Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Immunol Immunother ; 73(1): 16, 2024 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-38236251

RESUMO

Collagen expression and structure in the tumour microenvironment are associated with tumour development and therapy response. Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1) is a widely expressed inhibitory collagen receptor. LAIR-2 is a soluble homologue of LAIR-1 that competes for collagen binding. Multiple studies in mice implicate blockade of LAIR-1:collagen interaction in cancer as a promising therapeutic strategy. Here, we investigated the role of LAIR-1 in anti-tumour responses. We show that although LAIR-1 inhibits activation, proliferation, and cytokine production of mouse T cells in vitro, tumour outgrowth in LAIR-1-deficient mice did not differ from wild type mice in several in vivo tumour models. Furthermore, treatment with NC410, a LAIR-2-Fc fusion protein, did not result in increased tumour clearance in tested immunocompetent mice, which contrasts with previous data in humanized mouse models. This discrepancy may be explained by our finding that NC410 blocks human LAIR-1:collagen interaction more effectively than mouse LAIR-1:collagen interaction. Despite the lack of therapeutic impact of NC410 monotherapy, mice treated with a combination of NC410 and anti-programmed death-ligand 1 did show reduced tumour burden and increased survival. Using LAIR-1-deficient mice, we showed that this effect seemed to be dependent on the presence of LAIR-1. Taken together, our data demonstrate that the absence of LAIR-1 signalling alone is not sufficient to control tumour growth in multiple immunocompetent mouse models. However, combined targeting of LAIR-1 and PD-L1 results in increased tumour control. Thus, additional targeting of the LAIR-1:collagen pathway with NC410 is a promising approach to treating tumours where conventional immunotherapy is ineffective.


Assuntos
Antígeno B7-H1 , Neoplasias , Animais , Humanos , Camundongos , Colágeno , Modelos Animais de Doenças , Leucócitos , Ligantes , Neoplasias/tratamento farmacológico , Microambiente Tumoral
2.
Blood ; 139(20): 3087-3098, 2022 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-35245360

RESUMO

Glycoprotein VI (GPVI) mediates collagen-induced platelet activation after vascular damage and is an important contributor to the onset of thrombosis, heart attack, and stroke. Animal models of thrombosis have identified GPVI as a promising target for antithrombotic therapy. Although for many years the crystal structure of GPVI has been known, the essential details of its interaction with collagen have remained elusive. Here, we present crystal structures of the GPVI ectodomain bound to triple-helical collagen peptides, which reveal a collagen-binding site across the ß-sheet of the D1 domain. Mutagenesis and binding studies confirm the observed binding site and identify Trp76, Arg38, and Glu40 as essential residues for binding to fibrillar collagens and collagen-related peptides (CRPs). GPVI binds a site on collagen comprising two collagen chains with the core formed by the sequence motif OGPOGP. Potent GPVI-binding peptides from Toolkit-III all contain OGPOGP; weaker binding peptides frequently contain a partial motif varying at either terminus. Alanine-scanning of peptide III-30 also identified two AGPOGP motifs that contribute to GPVI binding, but steric hindrance between GPVI molecules restricts the maximum binding capacity. We further show that no cooperative interactions could occur between two GPVI monomers binding to a stretch of (GPO)5 and that binding of ≥2 GPVI molecules to a fibril-embedded helix requires non-overlapping OGPOGP motifs. Our structure confirms the previously suggested similarity in collagen binding between GPVI and leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1) but also indicates significant differences that may be exploited for the development of receptor-specific therapeutics.


Assuntos
Glicoproteínas da Membrana de Plaquetas , Trombose , Animais , Sítios de Ligação , Plaquetas/metabolismo , Colágeno/metabolismo , Peptídeos/química , Ativação Plaquetária , Glicoproteínas da Membrana de Plaquetas/metabolismo , Ligação Proteica , Trombose/metabolismo
3.
Int J Mol Sci ; 24(20)2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37894834

RESUMO

Mutations in the FKBP14 gene encoding the endoplasmic reticulum resident collagen-related proline isomerase FK506 binding protein 22 kDa (FKBP22) result in kyphoscoliotic Ehlers-Danlos Syndrome (EDS), which is characterized by a broad phenotypic outcome. A plausible explanation for this outcome is that FKBP22 participates in the biosynthesis of subsets of collagen types: FKBP22 selectively binds to collagens III, IV, VI, and X, but not to collagens I, II, V, and XI. However, these binding mechanisms have never been explored, and they may underpin EDS subtype heterogeneity. Here, we used collagen Toolkit peptide libraries to investigate binding specificity. We observed that FKBP22 binding was distributed along the collagen helix. Further, it (1) was higher on collagen III than collagen II peptides and it (2) was correlated with a positive peptide charge. These findings begin to elucidate the mechanism by which FKBP22 interacts with collagen.


Assuntos
Síndrome de Ehlers-Danlos , Proteínas de Ligação a Tacrolimo , Humanos , Proteínas de Ligação a Tacrolimo/metabolismo , Colágeno/genética , Peptidilprolil Isomerase/genética , Mutação , Síndrome de Ehlers-Danlos/genética
4.
Nat Chem Biol ; 16(4): 423-429, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31907373

RESUMO

The most abundant member of the collagen protein family, collagen I (also known as type I collagen; COL1), is composed of one unique (chain B) and two similar (chain A) polypeptides that self-assemble with one amino acid offset into a heterotrimeric triple helix. Given the offset, chain B can occupy either the leading (BAA), middle (ABA) or trailing (AAB) position of the triple helix, yielding three isomeric biomacromolecules with different protein recognition properties. Despite five decades of intensive research, there is no consensus on the position of chain B in COL1. Here, three triple-helical heterotrimers that each contain a putative von Willebrand factor (VWF) and discoidin domain receptor (DDR) recognition sequence from COL1 were designed with chain B permutated in all three positions. AAB demonstrated a strong preference for both VWF and DDR, and also induced higher levels of cellular DDR phosphorylation. Thus, we resolve this long-standing mystery and show that COL1 adopts an AAB register.


Assuntos
Colágeno Tipo I/química , Colágeno Tipo I/metabolismo , Colágeno/química , Sequência de Aminoácidos , Aminoácidos , Colágeno/metabolismo , Biologia Computacional/métodos , Humanos , Modelos Moleculares , Peptídeos/química , Conformação Proteica
5.
Arterioscler Thromb Vasc Biol ; 41(2): e97-e111, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33267658

RESUMO

OBJECTIVE: Fibrin is considered to strengthen thrombus formation via integrin αIIbß3, but recent findings indicate that fibrin can also act as ligand for platelet glycoprotein VI. Approach and Results: To investigate the thrombus-forming potential of fibrin and the roles of platelet receptors herein, we generated a range of immobilized fibrin surfaces, some of which were cross-linked with factor XIIIa and contained VWF-BP (von Willebrand factor-binding peptide). Multicolor microfluidics assays with whole-blood flowed at high shear rate (1000 s-1) indicated that the fibrin surfaces, regardless of the presence of factor XIIIa or VWF-BP, supported platelet adhesion and activation (P-selectin expression), but only microthrombi were formed consisting of bilayers of platelets. Fibrinogen surfaces produced similar microthrombi. Markedly, tiggering of coagulation with tissue factor or blocking of thrombin no more than moderately affected the fibrin-induced microthrombus formation. Absence of αIIbß3 in Glanzmann thrombasthenia annulled platelet adhesion. Blocking of glycoprotein VI with Fab 9O12 substantially, but incompletely reduced platelet secretion, Ca2+ signaling and aggregation, while inhibition of Syk further reduced these responses. In platelet suspension, glycoprotein VI blockage or Syk inhibition prevented fibrin-induced platelet aggregation. Microthrombi on fibrin surfaces triggered only minimal thrombin generation, in spite of thrombin binding to the fibrin fibers. CONCLUSIONS: Together, these results indicate that fibrin fibers, regardless of their way of formation, act as a consolidating surface in microthrombus formation via nonredundant roles of platelet glycoprotein VI and integrin αIIbß3 through signaling via Syk and low-level Ca2+ rises.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Fibrina/metabolismo , Adesividade Plaquetária , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Trombose/sangue , Plaquetas/ultraestrutura , Sinalização do Cálcio , Estudos de Casos e Controles , Feminino , Fibrina/ultraestrutura , Humanos , Masculino , Técnicas Analíticas Microfluídicas , Quinase Syk/sangue , Trombastenia/sangue , Trombose/patologia
6.
Platelets ; 33(3): 451-461, 2022 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-34348571

RESUMO

Kv1.3 is a voltage-gated K+-selective channel with roles in immunity, insulin-sensitivity, neuronal excitability and olfaction. Despite being one of the largest ionic conductances of the platelet surface membrane, its contribution to platelet function is poorly understood. Here we show that Kv1.3-deficient platelets display enhanced ADP-evoked platelet aggregation and secretion, and an increased surface expression of platelet integrin αIIb. In contrast, platelet adhesion and thrombus formation in vitro under arterial shear conditions on surfaces coated with collagen were reduced for samples from Kv1.3-/- compared to wild type mice. Use of collagen-mimetic peptides revealed a specific defect in the engagement with α2ß1. Kv1.3-/- platelets developed significantly fewer, and shorter, filopodia than wild type platelets during adhesion to collagen fibrils. Kv1.3-/- mice displayed no significant difference in thrombus formation within cremaster muscle arterioles using a laser-induced injury model, thus other pro-thrombotic pathways compensate in vivo for the adhesion defect observed in vitro. This may include the increased platelet counts of Kv1.3-/- mice, due in part to a prolonged lifespan. The ability of Kv1.3 to modulate integrin-dependent platelet adhesion has important implications for understanding its contribution to normal physiological platelet function in addition to its reported roles in auto-immune diseases and thromboinflammatory models of stroke.


Assuntos
Plaquetas/metabolismo , Colágeno/metabolismo , Integrina alfa2beta1/metabolismo , Adesividade Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Humanos
7.
Toxicol Appl Pharmacol ; 428: 115669, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34363821

RESUMO

Integrins are a family of 24 adhesion receptors which are both widely-expressed and important in many pathophysiological cellular processes, from embryonic development to cancer metastasis. Hence, integrin inhibitors are valuable research tools which may have promising therapeutic uses. Here, we focus on the four collagen-binding integrins α1ß1, α2ß1, α10ß1 and α11ß1. TC-I-15 is a small molecule inhibitor of α2ß1 that inhibits platelet adhesion to collagen and thrombus deposition, and obtustatin is an α1ß1-specific disintegrin that inhibits angiogenesis. Both inhibitors were applied in cellular adhesion studies, using synthetic collagen peptide coatings with selective affinity for the different collagen-binding integrins and testing the adhesion of C2C12 cells transfected with each. Obtustatin was found to be specific for α1ß1, as described, whereas TC-I-15 is shown to be non-specific, since it inhibits both α1ß1 and α11ß1 as well as α2ß1. TC-I-15 was 100-fold more potent against α2ß1 binding to a lower-affinity collagen peptide, suggestive of a competitive mechanism. These results caution against the use of integrin inhibitors in a therapeutic or research setting without testing for cross-reactivity.


Assuntos
Inibidores da Angiogênese/farmacologia , Colágeno/metabolismo , Integrina alfa2beta1/antagonistas & inibidores , Integrina alfa2beta1/metabolismo , Venenos de Víboras/metabolismo , Venenos de Víboras/farmacologia , Inibidores da Angiogênese/química , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular , Relação Dose-Resposta a Droga , Camundongos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia
8.
Anal Chem ; 91(13): 8025-8035, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31074960

RESUMO

Analysis of oxylipins by liquid chromatography mass spectrometry (LC/MS) is challenging because of the small mass range occupied by this diverse lipid class, the presence of numerous structural isomers, and their low abundance in biological samples. Although highly sensitive LC/MS/MS methods are commonly used, further separation is achievable by using drift tube ion mobility coupled with high-resolution mass spectrometry (DTIM-MS). Herein, we present a combined analytical and computational method for the identification of oxylipins and fatty acids. We use a reversed-phase LC/DTIM-MS workflow able to profile and quantify (based on chromatographic peak area) the oxylipin and fatty acid content of biological samples while simultaneously acquiring full scan and product ion spectra. The information regarding accurate mass, collision-cross-section values in nitrogen (DTCCSN2), and retention times of the species found are compared to an internal library of lipid standards as well as the LIPID MAPS Structure Database by using specifically developed processing tools. Features detected within the DTCCSN2 and m/ z ranges of the analyzed standards are flagged as oxylipin-like species, which can be further characterized using drift-time alignment of product and precursor ions distinctive of DTIM-MS. This not only helps identification by reducing the number of annotations from LIPID MAPS but also guides discovery studies of potentially novel species. Testing the methodology on Salmonella enterica serovar Typhimurium-infected murine bone-marrow-derived macrophages and thrombin activated human platelets yields results in agreement with literature. This workflow has also annotated features as potentially novel oxylipins, confirming its ability in providing further insights into lipid analysis of biological samples.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Eicosanoides/análise , Ácidos Graxos/análise , Oxilipinas/análise , Espectrometria de Massas em Tandem/métodos , Animais , Células Cultivadas , Humanos , Espectrometria de Mobilidade Iônica/métodos , Camundongos Endogâmicos C57BL
9.
Int J Mol Sci ; 20(11)2019 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-31181592

RESUMO

Platelet interaction with collagens, via von Willebrand factor, is a potent trigger of shear-dependent thrombus formation mediated by subsequent engagement of the signaling collagen receptor glycoprotein (GP)VI, enforced by integrin α2ß1. Protein tyrosine kinase Syk is central in the GPVI-induced signaling pathway, leading to elevated cytosolic Ca2+. We aimed to determine the Syk-mediated thrombogenic activity of several collagen peptides and (fibrillar) type I and III collagens. High-shear perfusion of blood over microspots of these substances resulted in thrombus formation, which was assessed by eight parameters and was indicative of platelet adhesion, activation, aggregation, and contraction, which were affected by the Syk inhibitor PRT-060318. In platelet suspensions, only collagen peptides containing the consensus GPVI-activating sequence (GPO)n and Horm-type collagen evoked Syk-dependent Ca2+ rises. In whole blood under flow, Syk inhibition suppressed platelet activation and aggregation parameters for the collagen peptides with or without a (GPO)n sequence and for all of the collagens. Prediction models based on a regression analysis indicated a mixed role of GPVI in thrombus formation on fibrillar collagens, which was abolished by Syk inhibition. Together, these findings indicate that GPVI-dependent signaling through Syk supports platelet activation in thrombus formation on collagen-like structures regardless of the presence of a (GPO)n sequence.


Assuntos
Plaquetas/metabolismo , Colágeno/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Quinase Syk/metabolismo , Trombose/metabolismo , Plaquetas/efeitos dos fármacos , Plaquetas/fisiologia , Células Cultivadas , Colágeno/química , Cicloexilaminas/farmacologia , Humanos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Agregação Plaquetária , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Quinase Syk/antagonistas & inibidores
10.
J Struct Biol ; 203(3): 255-262, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29758270

RESUMO

Gly missense mutations in type I collagen, which replace a conserved Gly in the repeating (Gly-Xaa-Yaa)n sequence with a larger residue, are known to cause Osteogenesis Imperfecta (OI). The clinical consequences of such mutations range from mild to lethal, with more serious clinical severity associated with larger Gly replacement residues. Here, we investigate the influence of the identity of the residue replacing Gly within and adjacent to the integrin binding 502GFPGER507 sequence on triple-helix structure, stability and integrin binding using a recombinant bacterial collagen system. Recombinant collagens were constructed with Gly substituted by Ala, Ser or Val at four positions within the integrin binding region. All constructs formed a stable triple-helix structure with a small decrease in melting temperature. Trypsin was used to probe local disruption of the triple helix, and Gly to Val replacements made the triple helix trypsin sensitive at three of the four sites. Any mutation at Gly505, eliminated integrin binding, while decreased integrin binding affinity was observed in the replacement of Gly residues at Gly502 following the order Val > Ser > Ala. Molecular dynamics simulations indicated that all Gly replacements led to transient disruption of triple-helix interchain hydrogen bonds in the region of the Gly replacement. These computational and experimental results lend insight into the complex molecular basis of the varying clinical severity of OI.


Assuntos
Colágeno Tipo I/química , Osteogênese Imperfeita/genética , Conformação Proteica , Sequência de Aminoácidos/genética , Substituição de Aminoácidos/genética , Dicroísmo Circular , Colágeno Tipo I/genética , Colágeno Tipo I/ultraestrutura , Glicina/genética , Humanos , Ligação de Hidrogênio , Mutação de Sentido Incorreto , Osteogênese Imperfeita/patologia , Ligação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína
11.
Blood ; 127(5): 529-37, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26552697

RESUMO

The osteoclast-associated receptor (OSCAR) is a collagen-binding immune receptor with important roles in dendritic cell maturation and activation of inflammatory monocytes as well as in osteoclastogenesis. The crystal structure of the OSCAR ectodomain is presented, both free and in complex with a consensus triple-helical peptide (THP). The structures revealed a collagen-binding site in each immunoglobulin-like domain (D1 and D2). The THP binds near a predicted collagen-binding groove in D1, but a more extensive interaction with D2 is facilitated by the unusually wide D1-D2 interdomain angle in OSCAR. Direct binding assays, combined with site-directed mutagenesis, confirm that the primary collagen-binding site in OSCAR resides in D2, in marked contrast to the related collagen receptors, glycoprotein VI (GPVI) and leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1). Monomeric OSCAR D1D2 binds to the consensus THP with a KD of 28 µM measured in solution, but shows a higher affinity (KD 1.5 µM) when binding to a solid-phase THP, most likely due to an avidity effect. These data suggest a 2-stage model for the interaction of OSCAR with a collagen fibril, with transient, low-affinity interactions initiated by the membrane-distal D1, followed by firm adhesion to the primary binding site in D2.


Assuntos
Colágeno/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Colágeno/química , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Glicoproteínas da Membrana de Plaquetas/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Imunológicos/metabolismo
12.
J Mater Sci Mater Med ; 29(4): 39, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29564650

RESUMO

The article "Evaluation of cell binding to collagen and gelatin: a study of the effect of 2D and 3D architecture and surface chemistry", written by Natalia Davidenko, Carlos F. Schuster, Daniel V. Bax, Richard W. Farndale, Samir Hamaia, Serena M. Best and Ruth E. Cameron, was originally published Online First without open access. After publication in volume 27, issue 10, page 148 it was noticed that the copyright was wrong in the PDF version of the article. The copyright of the article should read as "© The Author(s) 2016". The Open Access license terms were also missing.

13.
J Biol Chem ; 291(15): 7951-60, 2016 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-26893379

RESUMO

The hallmark of fibrotic disorders is a highly cross-linked and dense collagen matrix, a property driven by the oxidative action of lysyl oxidase. Other fibrosis-associated proteins also contribute to the final collagen matrix properties, one of which is fibromodulin. Its interactions with collagen affect collagen cross-linking, packing, and fibril diameter. We investigated the possibility that a specific relationship exists between fibromodulin and lysyl oxidase, potentially imparting a specific collagen matrix phenotype. We mapped the fibromodulin-collagen interaction sites using the collagen II and III Toolkit peptide libraries. Fibromodulin interacted with the peptides containing the known collagen cross-linking sites and the MMP-1 cleavage site in collagens I and II. Interestingly, the interaction sites are closely aligned within the quarter-staggered collagen fibril, suggesting a multivalent interaction between fibromodulin and several collagen helices. Furthermore, we detected an interaction between fibromodulin and lysyl oxidase (a major collagen cross-linking enzyme) and mapped the interaction site to 12 N-terminal amino acids on fibromodulin. This interaction also increases the activity of lysyl oxidase. Together, the data suggest a fibromodulin-modulated collagen cross-linking mechanism where fibromodulin binds to a specific part of the collagen domain and also forms a complex with lysyl oxidase, targeting the enzyme toward specific cross-linking sites.


Assuntos
Colágeno/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Proteoglicanas/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Colágeno/análise , Ativação Enzimática , Proteínas da Matriz Extracelular/genética , Fibromodulina , Deleção de Genes , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mapas de Interação de Proteínas , Proteína-Lisina 6-Oxidase/análise , Proteoglicanas/genética
14.
J Biol Chem ; 291(36): 19196-207, 2016 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-27432884

RESUMO

The replacement of one Gly in the essential repeating tripeptide sequence of the type I collagen triple helix results in the dominant hereditary bone disorder osteogenesis imperfecta. The mechanism leading to pathology likely involves misfolding and autophagy, although it has been hypothesized that some mutations interfere with known collagen interactions. Here, the effect of Gly replacements within and nearby the integrin binding GFPGER sequence was investigated using a recombinant bacterial collagen system. When a six-triplet human type I collagen sequence containing GFPGER was introduced into a bacterial collagen-like protein, this chimeric protein bound to integrin. Constructs with Gly to Ser substitutions within and nearby the inserted human sequence still formed a trypsin-resistant triple helix, suggesting a small local conformational perturbation. Gly to Ser mutations within the two Gly residues in the essential GFPGER sequence prevented integrin binding and cell attachment as predicted from molecular dynamics studies of the complex. Replacement of Gly residues C-terminal to GFPGER did not affect integrin binding. In contrast, Gly replacements N-terminal to the GFPGER sequence, up to four triplets away, decreased integrin binding and cell adhesion. This pattern suggests either an involvement of the triplets N-terminal to GFPGER in initial binding or a propagation of the perturbation of the triple helix C-terminal to a mutation site. The asymmetry in biological consequences relative to the mutation site may relate to the observed pattern of osteogenesis imperfecta mutations near the integrin binding site.


Assuntos
Colágeno Tipo I/química , Integrina alfa2beta1/química , Substituição de Aminoácidos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Humanos , Integrina alfa2beta1/genética , Integrina alfa2beta1/metabolismo , Mutação de Sentido Incorreto , Ligação Proteica , Estrutura Secundária de Proteína
15.
J Biol Chem ; 291(9): 4343-55, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26702058

RESUMO

A bacterial collagen-like protein Scl2 has been developed as a recombinant collagen model system to host human collagen ligand-binding sequences, with the goal of generating biomaterials with selective collagen bioactivities. Defined binding sites in human collagen for integrins, fibronectin, heparin, and MMP-1 have been introduced into the triple-helical domain of the bacterial collagen and led to the expected biological activities. The modular insertion of activities is extended here to the discoidin domain receptors (DDRs), which are collagen-activated receptor tyrosine kinases. Insertion of the DDR-binding sequence from human collagen III into bacterial collagen led to specific receptor binding. However, even at the highest testable concentrations, the construct was unable to stimulate DDR autophosphorylation. The recombinant collagen expressed in Escherichia coli does not contain hydroxyproline (Hyp), and complementary synthetic peptide studies showed that replacement of Hyp by Pro at the critical Gly-Val-Met-Gly-Phe-Hyp position decreased the DDR-binding affinity and consequently required a higher concentration for the induction of receptor activation. The ability of the recombinant bacterial collagen to bind the DDRs without inducing kinase activation suggested it could interfere with the interactions between animal collagen and the DDRs, and such an inhibitory role was confirmed in vitro and with a cell migration assay. This study illustrates that recombinant collagen can complement synthetic peptides in investigating structure-activity relationships, and this system has the potential for the introduction or inhibition of specific biological activities.


Assuntos
Proteínas de Bactérias/metabolismo , Colágeno Tipo III/metabolismo , Colágeno/metabolismo , Megacariócitos/metabolismo , Modelos Moleculares , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Mitogênicos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Movimento Celular , Células Cultivadas , Colágeno/química , Colágeno/genética , Colágeno Tipo III/química , Colágeno Tipo III/genética , Receptores com Domínio Discoidina , Sangue Fetal/citologia , Células HEK293 , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/genética , Proteínas Imobilizadas/metabolismo , Ligantes , Megacariócitos/citologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Engenharia de Proteínas , Domínios e Motivos de Interação entre Proteínas , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/genética , Receptores Mitogênicos/antagonistas & inibidores , Receptores Mitogênicos/química , Receptores Mitogênicos/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Streptococcus pyogenes
16.
Blood ; 124(14): 2173-83, 2014 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-25082876

RESUMO

Mitochondrial DNA (mtDNA) is a highly potent inflammatory trigger and is reportedly found outside the cells in blood in various pathologies. Platelets are abundant in blood where they promote hemostasis. Although lacking a nucleus, platelets contain functional mitochondria. On activation, platelets produce extracellular vesicles known as microparticles. We hypothesized that activated platelets could also release their mitochondria. We show that activated platelets release respiratory-competent mitochondria, both within membrane-encapsulated microparticles and as free organelles. Extracellular mitochondria are found in platelet concentrates used for transfusion and are present at higher levels in those that induced acute reactions (febrile nonhemolytic reactions, skin manifestations, and cardiovascular events) in transfused patients. We establish that the mitochondrion is an endogenous substrate of secreted phospholipase A2 IIA (sPLA2-IIA), a phospholipase otherwise specific for bacteria, likely reflecting the ancestral proteobacteria origin of mitochondria. The hydrolysis of the mitochondrial membrane by sPLA2-IIA yields inflammatory mediators (ie, lysophospholipids, fatty acids, and mtDNA) that promote leukocyte activation. Two-photon microscopy in live transfused animals revealed that extracellular mitochondria interact with neutrophils in vivo, triggering neutrophil adhesion to the endothelial wall. Our findings identify extracellular mitochondria, produced by platelets, at the midpoint of a potent mechanism leading to inflammatory responses.


Assuntos
Plaquetas/metabolismo , Fosfolipases A2 do Grupo II/metabolismo , Inflamação/metabolismo , Mitocôndrias/metabolismo , Animais , DNA Mitocondrial/metabolismo , Endotélio Vascular/metabolismo , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ativação Plaquetária , Rickettsia prowazekii/metabolismo
17.
Circ Res ; 114(5): 780-91, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24406984

RESUMO

RATIONALE: Platelets are the most important cells in the primary prevention of blood loss after injury. In addition, platelets are at the interface between circulating leukocytes and the (sub)endothelium regulating inflammatory responses. OBJECTIVE: Our aim was to study the dynamic process that leads to the formation of procoagulant and proinflammatory platelets under physiological flow. METHODS AND RESULTS: In the present study, we describe the formation of extremely long, negatively charged membrane strands that emerge from platelets adhered under flow. These flow-induced protrusions (FLIPRs) are formed in vitro on different physiological substrates and are also detected in vivo in a mouse carotid injury model. FLIPRs are formed downstream the adherent and activated platelets and reach lengths of 250 µm. FLIPR formation is shear-dependent and requires cyclophilin D, calpain, and Rac1 activation. It is accompanied by a disassembly of the F-actin and microtubule organization. Monocytes and neutrophils roll over FLIPRs in a P-selectin/P-selectin glycoprotein ligand-1-dependent manner, retrieving fragments of FLIPRs as microparticles on their surface. Consequently, monocytes and neutrophils become activated, as demonstrated by increased CD11b expression and L-selectin shedding. CONCLUSIONS: The formation of long platelet membrane extensions, such as the ones presented in our flow model, may pave the way to generate an increased membrane surface for interaction with monocytes and neutrophils. Our study provides a mechanistic model for platelet membrane transfer and the generation of monocyte/neutrophil-microparticle complexes. We propose that the formation of FLIPRs in vivo contributes to the well-established proinflammatory function of platelets and platelet-derived microparticles.


Assuntos
Plaquetas/citologia , Plaquetas/imunologia , Lesões das Artérias Carótidas/imunologia , Micropartículas Derivadas de Células/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Animais , Cálcio/metabolismo , Lesões das Artérias Carótidas/patologia , Citoesqueleto/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Voluntários Saudáveis , Humanos , Camundongos , Monócitos/citologia , Neutrófilos/citologia , Ativação Plaquetária/imunologia , Fluxo Sanguíneo Regional/imunologia , Estresse Mecânico
18.
J Mater Sci Mater Med ; 27(1): 14, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26676860

RESUMO

Short wavelength (λ = 254 nm) UV irradiation was evaluated over a range of intensities (0.06 to 0.96 J/cm(2)) as a means of cross-linking collagen- and gelatin-based scaffolds, to tailor their material characteristics whilst retaining biological functionality. Zero-link carbodiimide treatments are commonly applied to collagen-based materials, forming cross-links from carboxylate anions (for example the acidic E of GFOGER) that are an essential part of integrin binding sites on collagen. Cross-linking these amino acids therefore disrupts the bioactivity of collagen. In contrast, UV irradiation forms bonds from less important aromatic tyrosine and phenylalanine residues. We therefore hypothesised that UV cross-linking would not compromise collagen cell reactivity. Here, highly porous (~99 %) isotropic, collagen-based scaffolds were produced via ice-templating. A series of scaffolds (pore diameters ranging from 130-260 µm) with ascending stability in water was made from gelatin, two different sources of collagen I, or blends of these materials. Glucose, known to aid UV crosslinking of collagen, was added to some lower-stability formulations. These scaffolds were exposed to different doses of UV irradiation, and the scaffold morphology, dissolution stability in water, resistance to compression and cell reactivity was assessed. Stabilisation in aqueous media varied with both the nature of the collagen-based material employed and the UV intensity. Scaffolds made from the most stable materials showed the greatest stability after irradiation, although the levels of cross-linking in all cases were relatively low. Scaffolds made from pure collagen from the two different sources showed different optimum levels of irradiation, suggesting altered balance between stabilisation from cross-linking and destabilisation from denaturation. The introduction of glucose into the scaffold enhanced the efficacy of UV cross-linking. Finally, as hypothesized, cell attachment, spreading and proliferation on collagen materials were unaffected by UV cross-linking. UV irradiation may therefore be used to provide relatively low level cross-linking of collagen without loss of biological functionality.


Assuntos
Colágeno Tipo I/química , Alicerces Teciduais , Raios Ultravioleta , Animais , Sítios de Ligação , Bovinos , Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Microscopia Eletrônica de Varredura
19.
J Mater Sci Mater Med ; 27(10): 148, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27582068

RESUMO

Studies of cell attachment to collagen-based materials often ignore details of the binding mechanisms-be they integrin-mediated or non-specific. In this work, we have used collagen and gelatin-based substrates with different dimensional characteristics (monolayers, thin films and porous scaffolds) in order to establish the influence of composition, crosslinking (using carbodiimide) treatment and 2D or 3D architecture on integrin-mediated cell adhesion. By varying receptor expression, using cells with collagen-binding integrins (HT1080 and C2C12 L3 cell lines, expressing α2ß1, and Rugli expressing α1ß1) and a parent cell line C2C12 with gelatin-binding receptors (αvß3 and α5ß1), the nature of integrin binding sites was studied in order to explain the bioactivity of different protein formulations. We have shown that alteration of the chemical identity, conformation and availability of free binding motifs (GxOGER and RGD), resulting from addition of gelatin to collagen and crosslinking, have a profound effect on the ability of cells to adhere to these formulations. Carbodiimide crosslinking ablates integrin-dependent cell activity on both two-dimensional and three-dimensional architectures while the three-dimensional scaffold structure also leads to a high level of non-specific interactions remaining on three-dimensional samples even after a rigorous washing regime. This phenomenon, promoted by crosslinking, and attributed to cell entrapment, should be considered in any assessment of the biological activity of three-dimensional substrates. Spreading data confirm the importance of integrin-mediated cell engagement for further cell activity on collagen-based compositions. In this work, we provide a simple, but effective, means of deconvoluting the effects of chemistry and dimensional characteristics of a substrate, on the cell activity of protein-derived materials, which should assist in tailoring their biological properties for specific tissue engineering applications.


Assuntos
Colágeno/química , Gelatina/química , Tendão do Calcâneo/metabolismo , Motivos de Aminoácidos , Animais , Carbodi-Imidas/química , Bovinos , Adesão Celular , Linhagem Celular , Linhagem Celular Tumoral , Materiais Revestidos Biocompatíveis , Reagentes de Ligações Cruzadas/química , Matriz Extracelular/metabolismo , Humanos , Integrinas/química , Ligantes , Teste de Materiais , Camundongos , Ligação Proteica , Propriedades de Superfície , Engenharia Tecidual/métodos , Alicerces Teciduais/química
20.
J Biol Chem ; 289(35): 24091-101, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25008319

RESUMO

Remodeling of collagen by matrix metalloproteinases (MMPs) is crucial to tissue homeostasis and repair. MMP-13 is a collagenase with a substrate preference for collagen II over collagens I and III. It recognizes a specific, well-known site in the tropocollagen molecule where its binding locally perturbs the triple helix, allowing the catalytic domain of the active enzyme to cleave the collagen α chains sequentially, at Gly(775)-Leu(776) in collagen II. However, the specific residues upon which collagen recognition depends within and surrounding this locus have not been systematically mapped. Using our triple-helical peptide Collagen Toolkit libraries in solid-phase binding assays, we found that MMP-13 shows little affinity for Collagen Toolkit III, but binds selectively to two triple-helical peptides of Toolkit II. We have identified the residues required for the adhesion of both proMMP-13 and MMP-13 to one of these, Toolkit peptide II-44, which contains the canonical collagenase cleavage site. MMP-13 was unable to bind to a linear peptide of the same sequence as II-44. We also discovered a second binding site near the N terminus of collagen II (starting at helix residue 127) in Toolkit peptide II-8. The pattern of binding of the free hemopexin domain of MMP-13 was similar to that of the full-length enzyme, but the free catalytic subunit bound none of our peptides. The susceptibility of Toolkit peptides to proteolysis in solution was independent of the very specific recognition of immobilized peptides by MMP-13; the enzyme proved able to cleave a range of dissolved collagen peptides.


Assuntos
Colágeno Tipo II/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Colágeno Tipo II/química , Primers do DNA , Metaloproteinase 13 da Matriz/química , Dados de Sequência Molecular , Proteólise , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA