Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Pharmaceutics ; 16(5)2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38794344

RESUMEN

The successful substitution of complex physiological fluids, such as human saliva, remains a major challenge in drug development. Although there are a large number of saliva substitutes on the market, their efficacy is often inadequate due to short residence time in the mouth, unpleasant mouthfeel, or insufficient protection of the teeth. Therefore, systems need to be identified that mimic the functions of saliva, in particular the salivary mucin MUC5B and the unique physiological properties of saliva. To this end, plant extracts known to contain hydrocolloid polysaccharides and to have mucus-forming properties were studied to evaluate their suitability as saliva substitutes. The aqueous plant extracts of Calendula officinalis, Fucus sp. thalli, and lichenan from Lichen islandicus were examined for composition using a range of techniques, including GC-MS, NMR, SEC, assessment of pH, osmolality, buffering capacity, viscoelasticity, viscoelastic interactions with human saliva, hydrocolloid network formation, and in vitro cell adhesion. For this purpose, a physiologically adapted adhesive test was developed using human buccal epithelial cells. The results show that lichenan is the most promising candidate to mimic the properties of MUC5B. By adjusting the pH, osmolality, and buffering capacity with K2HPO4, it was shown that lichenan exhibited high cell adhesion, with a maximum detachment force that was comparable to that of unstimulated whole mouth saliva.

2.
Int J Mol Sci ; 24(19)2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37834140

RESUMEN

Tenascin-C (TNC) is a complex glycoprotein of the extracellular matrix (ECM) involved in a plethora of (patho-)physiological processes, such as oncogenesis and inflammation. Since chemokines play an essential role in both disease processes, we have investigated here the binding of TNC to some of the key chemokines, namely CCL2, CCL26, CXCL8, CXCL10, and CXCL12. Thereby, a differential chemokine-TNC binding pattern was observed, with CCL26 exhibiting the highest and CCL2 the lowest affinity for TNC. Heparan sulfate (HS), another member of the ECM, proved to be a similarly high-affinity ligand of TNC, with a Kd value of 730 nM. Chemokines use glycosa-minoglycans such as HS as co-receptors to induce immune cell migration. Therefore, we assumed an influence of TNC on immune cell chemotaxis due to co-localization within the ECM. CCL26- and CCL2-induced mobilization experiments of eosinophils and monocytes, respectively, were thus performed in the presence and the absence of TNC. Pre-incubation of the immune cells with TNC resulted in a 3.5-fold increase of CCL26-induced eosinophil chemotaxis, whereas a 1.3-fold de-crease in chemotaxis was observed when monocytes were pre-incubated with CCL2. As both chemokines have similar HS binding but different TNC binding affinities, we speculate that TNC acts as an attenuator in monocyte and as an amplifier in eosinophil mobilization by impeding CCL2 from binding to HS on the one hand, and by reinforcing CCL26 to bind to HS on the other hand.


Asunto(s)
Matriz Extracelular , Tenascina , Movimiento Celular , Matriz Extracelular/metabolismo , Heparitina Sulfato/metabolismo , Monocitos/metabolismo , Transducción de Señal , Tenascina/metabolismo , Humanos
3.
Int J Mol Sci ; 24(9)2023 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-37176074

RESUMEN

Bidirectional dialogue between cellular and non-cellular components of the tumor microenvironment (TME) drives cancer survival. In the extracellular space, combinations of matrix molecules and soluble mediators provide external cues that dictate the behavior of TME resident cells. Often studied in isolation, integrated cues from complex tissue microenvironments likely function more cohesively. Here, we study the interplay between the matrix molecule tenascin-C (TNC) and chemokine CCL2, both elevated in and associated with the progression of breast cancer and playing key roles in myeloid immune responses. We uncover a correlation between TNC/CCL2 tissue levels in HER2+ breast cancer and examine the physical and functional interactions of these molecules in a murine disease model with tunable TNC levels and in in vitro cellular and cell-free models. TNC supported sustained CCL2 synthesis, with chemokine binding to TNC via two distinct domains. TNC dominated the behavior of tumor-resident myeloid cells; CCL2 did not impact macrophage survival/activation whilst TNC facilitated an immune suppressive macrophage phenotype that was not dependent on or altered by CCL2 co-expression. Together, these data map new binding partners within the TME and demonstrate that whilst the matrix exerts transcriptional control over the chemokine, each plays a distinct role in subverting anti-tumoral immunity.


Asunto(s)
Neoplasias , Tenascina , Animales , Ratones , Quimiocinas/metabolismo , Matriz Extracelular/metabolismo , Macrófagos/metabolismo , Neoplasias/metabolismo , Transducción de Señal , Tenascina/metabolismo , Quimiocina CCL2/metabolismo
4.
Int J Mol Sci ; 23(12)2022 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-35742962

RESUMEN

Proinflammatory chemokine ligand 26 (CCL26, eotaxin-3) mediates transendothelial cell migration of eosinophils by binding and activating the G-protein-coupled (GPC) chemokine receptor 3 on the surface of eosinophilic cells. Here we have investigated the role of glycosaminoglycans (GAGs) as potential co-receptors in the process of CCL26-induced eosinophil chemotaxis. For this purpose, we have first identified the GAG-binding site of CCL26 by a site-directed mutagenesis approach in the form of an alanine screening. A panel of GAG-binding-deficient mutants has been designed, generated, and analyzed with respect to their binding affinities to heparan sulphate (HS) by isothermal fluorescence titration studies. This showed that basic amino acids in the α-helical part of CCL26 are strongly involved in GAG-binding. In chemotaxis experiments, we found that decreased GAG-binding affinity correlated with decreased chemotactic activity, which indicates an involvement of GAGs in eosinophil migration. This was further proven by the negative impact of heparinase III treatment and, independently, by the incubation of eosinophils with an anti heparan sulfate antibody. We finally investigated eosinophils' proteoglycan (PG) expression patterns by real-time PCR, which revealed the highest expression level for serglycin. Including an anti-serglycin antibody in CCL26-induced eosinophil migration experiments reduced the chemotaxis of these immune cells, thereby proving the dependence of eosinophil mobilization on the proteoglycan serglycin.


Asunto(s)
Quimiotaxis , Eosinófilos , Quimiocina CCL26 , Quimiocinas CC/metabolismo , Quimiotaxis de Leucocito , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/metabolismo , Proteoglicanos/metabolismo
5.
Carbohydr Res ; 511: 108480, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34837849

RESUMEN

A wide variety of diseases throughout the mammalian organism is characterized by abnormal deposition of various components of the extracellular matrix (ECM), including the heterogeneous family of glycosaminoglycans (GAGs), which contribute considerably to the ECM architecture as part of the so-called proteoglycans. The GAG's unique sulfation pattern, derived from highly dynamic and specific modification processes, has a massive impact on critical mediators such as cytokines and growth factors. Due to the strong connection between the specific sulfation pattern and GAG function, slight alterations of this pattern are often associated with enormous changes at the cell as well as at the organ level. This review aims to investigate the connection between modifications of GAG sulfation patterns and the wide range of pathological conditions, mainly focusing on a range of chronic diseases of the central nervous system (CNS) as well as the respiratory tract.


Asunto(s)
Matriz Extracelular , Glicosaminoglicanos , Animales , Encéfalo/metabolismo , Sulfatos de Condroitina/metabolismo , Matriz Extracelular/metabolismo , Glicosaminoglicanos/metabolismo , Humanos , Pulmón , Mamíferos/metabolismo , Proteoglicanos
6.
FASEB J ; 35(11): e21970, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34637549

RESUMEN

Single domain shark variable domain of new antigen receptor (VNAR) antibodies can offer a viable alternative to conventional Ig-based monoclonal antibodies in treating COVID-19 disease during the current pandemic. Here we report the identification of neutralizing single domain VNAR antibodies selected against the severe acute respiratory syndrome coronavirus 2 spike protein derived from the Wuhan variant using phage display. We identified 56 unique binding clones that exhibited high affinity and specificity to the spike protein. Of those, 10 showed an ability to block both the spike protein receptor binding domain from the Wuhan variant and the N501Y mutant from interacting with recombinant angiotensin-converting enzyme 2 (ACE2) receptor in vitro. In addition, three antibody clones retained in vitro blocking activity when the E484K spike protein mutant was used. The inhibitory property of the VNAR antibodies was further confirmed for all 10 antibody clones using ACE2 expressing cells with spike protein from the Wuhan variant. The viral neutralizing potential of the VNAR clones was also confirmed for the 10 antibodies tested using live Wuhan variant virus in in vitro cell infectivity assays. Single domain VNAR antibodies, due to their low complexity, small size, unique epitope recognition, and formatting flexibility, should be a useful adjunct to existing antibody approaches to treat COVID-19.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19 , SARS-CoV-2/inmunología , Anticuerpos de Dominio Único/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , COVID-19/inmunología , COVID-19/prevención & control , Chlorocebus aethiops , Humanos , Unión Proteica , Tiburones/inmunología , Células Vero
7.
Molecules ; 26(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34576979

RESUMEN

Glycosaminoglycans are a class of linear, highly negatively charged, O-linked polysaccharides that are involved in many (patho)physiological processes. In vitro experimental investigations of such processes typically involve porcine-derived heparan sulfate (HS). Structural information about human, particularly organ-specific heparan sulfate, and how it compares with HS from other organisms, is very limited. In this study, heparan sulfate was isolated from human lung tissues derived from five donors and was characterized for their overall size distribution and disaccharide composition. The expression profiles of proteoglycans and HS-modifying enzymes was quantified in order to identify the major core proteins for HS. In addition, the binding affinities of human HS to two chemokines-CXCL8 and CCL2-were investigated, which represent important inflammatory mediators in lung pathologies. Our data revealed that syndecans are the predominant proteoglycan class in human lungs and that the disaccharide composition varies among individuals according to sex, age, and health stage (one of the donor lungs was accidentally discovered to contain a solid tumor). The compositional difference of the five human lung HS preparations affected chemokine binding affinities to various degrees, indicating selective immune cell responses depending on the relative chemokine-glycan affinities. This represents important new insights that could be translated into novel therapeutic concepts for individually treating lung immunological disorders via HS targets.


Asunto(s)
Heparitina Sulfato , Animales , Glicosaminoglicanos , Humanos , Pulmón , Porcinos
8.
Eur J Pharm Biopharm ; 168: 152-165, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34474111

RESUMEN

Spray-dried products, such as synthetic peptides and hormones, have already been approved by the U.S. Food and Drug Agency and the European Medicines Agency, while spray-dried antibodies or interleukins, are not yet available on the market. Concerning the latter group, knowledge on whether and how spray-drying (SD) can be performed without adversely affecting their biological activity is lacking. Accordingly, this study aimed at establishing a SD process (Büchi B-90 spray dryer) using three Interleukin-8 based proteins (7-74 kDa) that were dispersed in phosphate buffered saline to maintain their stability. A Box-Behnken Design of Experiments was conducted to identify the appropriate process parameters taking into account the thermal stability of interleukin-8. In parallel, a FD process was developed. Both powders were stored for up to 12 weeks. Powder characterization included residual moisture evaluation and the mean particle size of the SD powder was investigated with Laser Diffraction Analysis. The hydrodynamic volume was measured via size exclusion chromatography and sodium dodecyl sulfate-polyacrylamide gel electrophoresis. The secondary structure of the model proteins in the solid state was assessed with Fourier-transformation infrared spectroscopy for detecting the protein folding patterns and reconstituted with Circular Dichroism Spectroscopy. Finally, the binding affinity was studied with Surface Plasmon Resonance and Isothermal Fluorescence Titration, the protein stability with Chaotropic Unfolding, and the activity studies were carried out with the chemotaxis assay. The results showed that SD and FD powders with a residual moisture of less than 5 wt% were obtained. The interleukins showed no unfolding upon processing, neither in solid state nor reconstituted. Oligomerization was observed for FD, but not for SD interleukins. However, the unfolding, binding affinity and activity of all interleukins examined did not decrease in neither SD nor FD powders, even after 12 weeks of storage. Thus, it can be concluded that SD of interleukin formulations at outlet temperatures close to ambient temperature is a promising process for transferring them into a stable powder.


Asunto(s)
Química Farmacéutica/métodos , Interleucina-8/química , Composición de Medicamentos/métodos , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Electroforesis en Gel de Poliacrilamida , Liofilización , Tamaño de la Partícula , Polvos , Estructura Secundaria de Proteína , Espectroscopía Infrarroja por Transformada de Fourier , Secado por Pulverización , Temperatura
9.
Hepatol Commun ; 5(10): 1737-1754, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34532999

RESUMEN

Hepatic cell death occurs in response to diverse stimuli such as chemical and physical damage. The exposure of intracellular contents such as DNA during necrosis induces a severe inflammatory response that has yet to be fully explored therapeutically. Here, we sought means to neutralize the ability of extracellular DNA to induce deleterious tissue inflammation when drug-induced liver injury had already ensued. DNA exposure and inflammation were investigated in vivo in drug-induced liver injury using intravital microscopy. The necrotic DNA debris was studied in murine livers in vivo and in DNA debris models in vitro by using a positively charged chemokine-derived peptide (MIG30; CXCL9[74-103]). Acetaminophen-induced liver necrosis was associated with massive DNA accumulation, production of CXC chemokines, and neutrophil activation inside the injured tissue. The MIG30 peptide bound the healthy liver vasculature and, to a much greater extent, to DNA-rich necrotic tissue. Moreover, MIG30 bound extracellular DNA directly in vivo in a charge-dependent manner and independently of glycosaminoglycans and chemokines. Post-treatment of mice with MIG30 reduced mortality, liver damage, and inflammation significantly. These effects were not observed with a control peptide that does not bind DNA. Mechanistically, MIG30 inhibited the interaction between DNA and histones, and promoted the dissociation of histones from necrotic debris. MIG30 also inhibited the pro-inflammatory effect of CpG DNA, as measured by a reduction in CXCL8 production, indicating that MIG30 disturbs the ability of DNA to induce hepatic inflammation. Conclusion: The use of DNA-binding peptides reduces necrotic liver injury and inflammation, even at late timepoints.


Asunto(s)
Antiinflamatorios/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Degradación Necrótica del ADN/efectos de los fármacos , Hígado/patología , Péptidos/farmacología , Acetaminofén/efectos adversos , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Quimiocina CXCL9/efectos de los fármacos , Quimiocinas CXC/efectos de los fármacos , Modelos Animales de Enfermedad , Matriz Extracelular/genética , Histonas/efectos de los fármacos , Humanos , Interleucina-8/efectos de los fármacos , Hígado/efectos de los fármacos , Ratones , Necrosis/inducido químicamente , Necrosis/patología , Activación Neutrófila/efectos de los fármacos , Electricidad Estática
10.
Front Immunol ; 12: 635166, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33790905

RESUMEN

The extracellular matrix (ECM) molecule Tenascin-C (TNC) is well-known to promote tumor progression by multiple mechanisms. However, reliable TNC detection in tissues of tumor banks remains limited. Therefore, we generated dromedary single-domain nanobodies Nb3 and Nb4 highly specific for human TNC (hTNC) and characterized the interaction with TNC by several approaches including ELISA, western blot, isothermal fluorescence titration and negative electron microscopic imaging. Our results revealed binding of both nanobodies to distinct sequences within fibronectin type III repeats of hTNC. By immunofluroescence and immunohistochemical imaging we observed that both nanobodies detected TNC expression in PFA and paraffin embedded human tissue from ulcerative colitis, solid tumors and liver metastasis. As TNC impairs cell adhesion to fibronectin we determined whether the nanobodies abolished this TNC function. Indeed, Nb3 and Nb4 restored adhesion of tumor and mesangial cells on a fibronectin/TNC substratum. We recently showed that TNC orchestrates the immune-suppressive tumor microenvironment involving chemoretention, causing tethering of CD11c+ myeloid/dendritic cells in the stroma. Here, we document that immobilization of DC2.4 dendritic cells by a CCL21 adsorbed TNC substratum was blocked by both nanobodies. Altogether, our novel TNC specific nanobodies could offer valuable tools for detection of TNC in the clinical practice and may be useful to inhibit the immune-suppressive and other functions of TNC in cancer and other diseases.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Camelus/inmunología , Anticuerpos de Dominio Único/inmunología , Tenascina/antagonistas & inhibidores , Animales , Anticuerpos Neutralizantes/farmacología , Especificidad de Anticuerpos , Sitios de Unión de Anticuerpos , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Colitis Ulcerosa/inmunología , Colon/inmunología , Ensayo de Inmunoadsorción Enzimática , Células HEK293 , Humanos , Inmunohistoquímica , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Unión Proteica , Anticuerpos de Dominio Único/farmacología , Tenascina/administración & dosificación , Tenascina/inmunología
11.
Semin Thromb Hemost ; 47(3): 316-332, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33794555

RESUMEN

Heparan sulfate proteoglycans (HSPGs) occur in almost every tissue of the human body and consist of a protein core, with covalently attached glycosaminoglycan polysaccharide chains. These glycosaminoglycans are characterized by their polyanionic nature, due to sulfate and carboxyl groups, which are distributed along the chain. These chains can be modified by different enzymes at varying positions, which leads to huge diversity of possible structures with the complexity further increased by varying chain lengths. According to their location, HSPGs are divided into different families, the membrane bound, the secreted extracellular matrix, and the secretory vesicle family. As members of the extracellular matrix, they take part in cell-cell communication processes on many levels and with different degrees of involvement. Of particular therapeutic interest is their role in cancer and inflammation as well as in infectious diseases. In this review, we give an overview of the current status of medical approaches to antagonize HSPG function in pathology.


Asunto(s)
Proteoglicanos de Heparán Sulfato/metabolismo , Humanos
12.
Cytokine ; 142: 155503, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33781652

RESUMEN

The role of glycosaminoglycans on the surface of immune cells has so far been less studied compared to their participation in inflammatory responses as members of the endothelium and the extracellular matrix. In this study we have therefore investigated if glycosaminoglycans on immune cells act in concert with GPC receptors (i.e. both being cis-located on leukocytes) in chemokine-induced leukocyte mobilisation. For this purpose, freshly-prepared human neutrophils and monocytes were treated with heparinase III or chondroitinase ABC to digest heparan sulfate -chains or chondroitin sulfate-chains, respectively, from the leukocyte surfaces. Subsequent analysis of CXCL8- and CCL2-induced chemotaxis revealed that leukocyte migration was strongly reduced after eliminating heparan sulfate from the surface of neutrophils and monocytes. In the case of monocytes, an additional dependence of CCL2-induced chemotaxis on chondroitin sulfate was observed. We compared these results with the effect on chemotaxis of a heparan sulfate masking antibody and obtained similarly reduced migration. Following our findings, we postulate that glycosaminoglycans located on target leukocytes act synergistically with GPC receptors on immune cell migration, which is further influenced by glycosaminoglycans located on the inflamed tissue (i.e. trans with respect to the immune cell/GPC receptor). Both glycosaminoglycan localization sites seem to be important during inflammatory processes and could potentially be tackled in chemokine-related diseases.


Asunto(s)
Movimiento Celular , Quimiocina CCL2/farmacología , Glicosaminoglicanos/metabolismo , Interleucina-8/farmacología , Monocitos/metabolismo , Neutrófilos/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Condroitinasas y Condroitín Liasas/metabolismo , Femenino , Glipicanos/genética , Glipicanos/metabolismo , Liasa de Heparina/metabolismo , Humanos , Monocitos/efectos de los fármacos , Neutrófilos/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Porcinos , Sindecanos/genética , Sindecanos/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos
13.
Biomolecules ; 11(1)2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33445810

RESUMEN

The recently identified severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, the cause of coronavirus disease (COVID-19) and the associated ongoing pandemic, frequently leads to severe respiratory distress syndrome and pneumonia with fatal consequences. Although several factors of this infection and its consequences are not completely clear, the presence and involvement of specific chemokines is undoubtedly crucial for the development and progression of COVID-19. Cytokine storm and the often-resulting cytokine release syndrome (CRS) are pathophysiological hallmarks in COVID-19 infections related to its most severe and fatal cases. In this hyperinflammatory event, chemokines and other cytokines are highly upregulated and are therefore not fulfilling their beneficial function in the host response anymore but causing harmful effects. Here, we present the recent views on the involvement of chemokines and selected cytokines in COVID-19 and the therapeutics currently in clinical development targeting or interfering with them, discussing their potentials in the treatment of COVID-19 infections.


Asunto(s)
COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Citocinas/inmunología , SARS-CoV-2 , Síndrome de Liberación de Citoquinas/tratamiento farmacológico , Humanos , Tratamiento Farmacológico de COVID-19
14.
Biomedicines ; 10(1)2021 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-35052728

RESUMEN

As with many other pathogens, SARS-CoV-2 cell infection is strongly dependent on the interaction of the virus-surface Spike protein with the glycosaminoglycans of target cells. The SARS-CoV-2 Spike glycoprotein was previously shown to interact with cell-surface-exposed heparan sulfate and heparin in vitro. With the aim of using Enoxaparin as a treatment for COVID-19 patients and as prophylaxis to prevent interpersonal viral transmission, we investigated GAG binding to the Spike full-length protein, as well as to its receptor binding domain (RBD) in solution by isothermal fluorescence titration. We found that Enoxaparin bound to both protein variants with similar affinities, compared to the natural GAG ligand heparan sulfate (with Kd-values in the range of 600-680 nM). Using size-defined Enoxaparin fragments, we discovered the optimum binding for dp6 or dp8 for the full-length Spike protein, whereas the RBD did not exhibit a significant chain-length-dependent affinity for heparin oligosaccharides. The soluble ACE2 receptor was found to interact with unfractionated GAGs in the low µM Kd range, but with size-defined heparins with clearly sub-µM Kd-values. Interestingly, the structural heparin analogue, pentosan polysulfate (PPS), exhibited high binding affinities to both Spike variants as well as to the ACE2 receptor. In viral infection experiments, Enoxaparin and PPS both showed a strong inhibition of infection in a concentration range of 50-500 µg/mL. Both compounds were found to retain their inhibitory effects at 500 µg/mL in a natural biomatrix-like human sputum. Our data suggest the early topical treatment of SARS-CoV-2 infections with inhaled Enoxaparin; some clinical studies in this direction are already ongoing, and they further imply an oral or nasal prophylactic inactivation of the virus by Enoxaparin or PPS for the prevention of inter-personal viral transmission.

15.
Int J Mol Sci ; 23(1)2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-35008874

RESUMEN

Although glycosaminoglycan (GAG)-protein interactions are important in many physiological and pathological processes, the structural requirements for binding are poorly defined. Starting with GAG-binding peptide CXCL9(74-103), peptides were designed to elucidate the contribution to the GAG-binding affinity of different: (1) GAG-binding motifs (i.e., BBXB and BBBXXB); (2) amino acids in GAG-binding motifs and linker sequences; and (3) numbers of GAG-binding motifs. The affinity of eight chemically synthesized peptides for various GAGs was determined by isothermal fluorescence titration (IFT). Moreover, the binding of peptides to cellular GAGs on Chinese hamster ovary (CHO) cells was assessed using flow cytometry with and without soluble GAGs. The repetition of GAG-binding motifs in the peptides contributed to a higher affinity for heparan sulfate (HS) in the IFT measurements. Furthermore, the presence of Gln residues in both GAG-binding motifs and linker sequences increased the affinity of trimer peptides for low-molecular-weight heparin (LMWH), partially desulfated (ds)LMWH and HS, but not for hyaluronic acid. In addition, the peptides bound to cellular GAGs with differential affinity, and the addition of soluble HS or heparin reduced the binding of CXCL9(74-103) to cellular GAGs. These results indicate that the affinity and specificity of peptides for GAGs can be tuned by adapting their amino acid sequence and their number of GAG-binding motifs.


Asunto(s)
Heparina de Bajo-Peso-Molecular/metabolismo , Heparitina Sulfato/metabolismo , Péptidos/química , Animales , Sitios de Unión , Células CHO , Cricetulus , Unión Proteica
16.
J Mol Biol ; 432(4): 930-951, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-31794729

RESUMEN

The mechanisms by which protein complexes convert from functional to pathogenic are the subject of intensive research. Here, we report how functionally unfavorable protein interactions can be induced by structural fuzziness, i.e., by persisting conformational disorder in protein complexes. We show that extreme disorder in the bound state transforms the intrinsically disordered protein SERF1a from an RNA-organizing factor into a pathogenic enhancer of alpha-synuclein (aSyn) amyloid toxicity. We demonstrate that SERF1a promotes the incorporation of RNA into nucleoli and liquid-like artificial RNA-organelles by retaining an unusually high degree of conformational disorder in the RNA-bound state. However, this type of structural fuzziness also determines an undifferentiated interaction with aSyn. RNA and aSyn both bind to one identical, positively charged site of SERF1a by an analogous electrostatic binding mode, with similar binding affinities, and without any observable disorder-to-order transition. The absence of primary or secondary structure discriminants results in SERF1a being unable to select between nucleic acid and amyloidogenic protein, leading the pro-amyloid aSyn:SERF1a interaction to prevail in the cytosol under conditions of cellular stress. We suggest that fuzzy disorder in SERF1a complexes accounts for an adverse gain-of-interaction which favors toxic binding to aSyn at the expense of nontoxic RNA binding, thereby leading to a functionally distorted and pathogenic process. Thus, structural fuzziness constitutes a direct link between extreme conformational flexibility, amyloid aggregation, and the malfunctioning of RNA-associated cellular processes, three signatures of neurodegenerative proteinopathies.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , ARN/química , alfa-Sinucleína/metabolismo , Animales , Citosol/metabolismo , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Ratones , Proteínas del Tejido Nervioso/química , Ácidos Nucleicos/química , Unión Proteica , ARN/metabolismo , Electricidad Estática , alfa-Sinucleína/química
17.
Arthritis Res Ther ; 21(1): 172, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31300004

RESUMEN

BACKGROUND: Syndecans are heparan sulfate proteoglycans that occur in membrane-bound or soluble forms. Syndecan-3, the least well-characterised of the syndecan family, is highly expressed on synovial endothelial cells in rheumatoid arthritis patients. Here, it binds pro-inflammatory chemokines with evidence for a role in chemokine presentation and leukocyte trafficking into the joint, promoting the inflammatory response. In this study, we explored the role of soluble syndecan-3 as a binder of chemokines and as an anti-inflammatory and therapeutic molecule. METHODS: A human monocytic cell line and CD14+ PBMCs were utilised in both Boyden chamber and trans-endothelial migration assays. Soluble syndecan-3 was tested in antigen-induced and collagen-induced in vivo arthritis models in mice. ELISA and isothermal fluorescence titration assays assessed the binding affinities. Syndecan-3 expression was identified by flow cytometry and PCR, and levels of shedding by ELISA. RESULTS: Using in vitro and in vivo models, soluble syndecan-3 inhibited leukocyte migration in vitro in response to CCL7 and its administration in murine models of rheumatoid arthritis reduced histological disease severity. Using isothermal fluorescence titration, the binding affinity of soluble syndecan-3 to inflammatory chemokines CCL2, CCL7 and CXCL8 was determined, revealing little difference, with Kds in the low nM range. TNFα increased cell surface expression and shedding of syndecan-3 from cultured human endothelial cells. Furthermore, soluble syndecan-3 occurred naturally in the sera of patients with rheumatoid arthritis and periodontitis, and its levels correlated with syndecan-1. CONCLUSIONS: This study shows that the addition of soluble syndecan-3 may represent an alternative therapeutic approach in inflammatory disease.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Movimiento Celular , Quimiocinas/metabolismo , Leucocitos/metabolismo , Sindecano-3/metabolismo , Animales , Artritis Reumatoide/patología , Células Cultivadas , Quimiocina CCL7/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Leucocitos/citología , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Unión Proteica , Índice de Severidad de la Enfermedad , Solubilidad , Sindecano-3/administración & dosificación , Sindecano-3/genética , Células THP-1 , Factor de Necrosis Tumoral alfa/farmacología
18.
J Mol Biol ; 431(14): 2581-2598, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31034892

RESUMEN

The recent discovery of biologically active fully disordered, so called random fuzzy protein-protein interactions leads to the question of how the high flexibility of these protein complexes correlates to aggregation and pathologic misfolding. We identify the structural mechanism by which a random fuzzy protein complex composed of the intrinsically disordered proteins alpha-Synuclein and SERF1a is able to potentiate cytotoxic aggregation. A structural model derived from an integrated NMR/SAXS analysis of the reconstituted aSyn:SERF1a complex enabled us to observe the partial deprotection of one precise aSyn amyloid nucleation element in the fully unstructured ensemble. This minimal exposure was sufficient to increase the amyloidogenic tendency of SERF1a-bound aSyn. Our findings provide a structural explanation of the previously observed pro-amyloid activity of SERF1a. They further demonstrate that random fuzziness can trigger a structurally organized disease-associated reaction such as amyloid polymerization.


Asunto(s)
Amiloide/química , Encéfalo/metabolismo , Proteínas Intrínsecamente Desordenadas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuroblastoma/metabolismo , alfa-Sinucleína/química , alfa-Sinucleína/metabolismo , Secuencia de Aminoácidos , Animales , Encéfalo/citología , Humanos , Proteínas Intrínsecamente Desordenadas/química , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Neuroblastoma/patología , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Homología de Secuencia
19.
Protein Eng Des Sel ; 32(8): 367-373, 2019 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-31974585

RESUMEN

The chemokine CXCL10 is released by a plethora of cells, including immune and metastatic cancer cells, following stimulation with interferon-gamma. It acts via its GPC receptor on T-cells attracting them to various target tissues. Glycosaminoglycans (GAGs) are regarded as co-receptors of chemokines, which enable the establishment of a chemotactic gradient for target cell migration. We have engineered human CXCL10 towards improved T-cell mobilisation by implementing a single site-directed mutation N20K into the protein, which leads to a higher GAG binding affinity compared to the wild type. Interestingly, this mutation not only increased T-cell migration in a transendothelial migration assay, the mutant intensified T-cell chemotaxis also in a Boyden chamber set-up thereby indicating a strong role of T-cell-localised GAGs on leukocyte migration. A CXCL10 mutant with increased GAG-binding affinity could therefore potentially serve as a T-cell mobiliser in pathological conditions where the immune surveillance of the target tissue is impaired, as is the case for most solid tumors.


Asunto(s)
Quimiocina CXCL10/metabolismo , Glicosaminoglicanos/metabolismo , Simulación de Dinámica Molecular , Linfocitos T/metabolismo , Secuencia de Aminoácidos , Unión Competitiva , Movimiento Celular , Células Cultivadas , Quimiocina CXCL10/química , Quimiocina CXCL10/genética , Quimiotaxis de Leucocito , Glicosaminoglicanos/química , Humanos , Masculino , Mutación Missense , Unión Proteica , Conformación Proteica , Ingeniería de Proteínas/métodos
20.
Biochim Biophys Acta Gen Subj ; 1863(3): 528-533, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30586626

RESUMEN

BACKGROUND: Binding of chemokines to glycosaminoglycans (GAGs) is a crucial step in leukocyte recruitment to inflamed tissues. METHODS: A disaccharide compositional analysis of the HS dp6 fraction in combination with MS analysis of the CCL2-depleted dp6 fraction was the basis for target GAG ligand structure suggestions. Four experimentally-derived heparan sulfate hexasaccharides, two potentially chemokine-specific and two unspecific, have been docked to CCL2. Subsequent 300 ns molecular dynamics simulations were used to improve the docked complexes. RESULTS: Hexasaccharides with four sulfations and no acetylations are suggested for selective and high affinity chemokine binding. Using the Antithromin-III/heparin complex as positive control for docking, we were able to recover the correct complex structure only if the previously liganded ATIII structure was used as input. Since the liganded structure is not known for a CCL2-GAG complex, we investigated if molecular dynamics simulations could improve initial docking results. We found that all four GAG oligosaccharides ended up in close contact with the known binding residues after about 100 ns simulation time. CONCLUSIONS: A discrimination of specific vs. unspecific CCL2 GAG ligands is not possible by this approach. Long-time molecular dynamics simulations are, however, well suited to capture the delicate enthalpy/entropy balance of GAG binding and improve results obtained from docking. GENERAL SIGNIFICANCE: With the comparison of two methods, MS-based ligand identification and molecular modelling, we have shown the current limitations of our molecular understanding of complex ligand binding which is could be due to the numerical inaccessibility of ligand-induced protein conformational changes.


Asunto(s)
Quimiocina CCL2/química , Quimiocina CCL2/metabolismo , Heparitina Sulfato/metabolismo , Simulación de Dinámica Molecular , Oligosacáridos/metabolismo , Secuencia de Carbohidratos , Fraccionamiento Químico/métodos , Cromatografía Líquida de Alta Presión/métodos , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Oligosacáridos/química , Oligosacáridos/aislamiento & purificación , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA