Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
Carbohydr Polym ; 304: 120492, 2023 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-36641179

RESUMO

Regeneration of injured articular cartilage is limited by low early-stage recruitment of stem cells and insufficient chondrogenic differentiation. Hydrogels are widely used to repair cartilage because they have excellent mechanical and biological properties. In this study, a dual drug-loaded thermosensitive hydroxypropyl chitin hydrogel (HPCH) system was prepared to release stromal-derived factor-1α-like polypeptides (SDFP) and kartogenin (KGN) for stem-cell recruitment and chondrogenic differentiation. The hydrogel had a network structure that promoted cell growth and nutrient exchange. Moreover, it was temperature sensitive and suitable for filling irregular defects. The system showed good biocompatibility in vitro and promoted stem-cell recruitment and chondrogenic differentiation. Furthermore, it reduced chondrocyte catabolism under inflammatory conditions. Animal experiments demonstrated that the dual-drug hydrogel systems can promote the regeneration of articular cartilage in rats. This study confirmed that an HPCH system loaded with KGN and SDFP could effectively repair articular cartilage defects and represents a viable treatment strategy.


Assuntos
Cartilagem Articular , Hidrogéis , Ratos , Animais , Hidrogéis/farmacologia , Hidrogéis/química , Quimiocina CXCL12/química , Regeneração , Diferenciação Celular , Condrogênese
2.
ACS Appl Mater Interfaces ; 15(2): 2578-2589, 2023 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-36598791

RESUMO

Transplantation of exogenous cardiomyocytes (CMs) is a hopeful method to treat myocardial infarction (MI). However, its clinical application still remains challenging due to low retention and survival rates of the transplanted cells. Herein, a stromal cell-derived factor 1 (SDF-1)-loaded injectable hydrogel based on a decellularized porcine extracellular matrix (dECM) is developed to encapsulate and deliver CMs locally to the infarct area of the heart. The soluble porcine cardiac dECM is composed of similar components such as the human cardiac ECM, which could be self-assembled into a nanofibrous hydrogel at physiological temperature to improve the retention of transplanted CMs. Furthermore, the chemokine SDF-1 could recruit endogenous cells to promote angiogenesis, mitigating the ischemic microenvironment and improving the survival of CMs. The results in vitro show that this composite hydrogel exhibits good biocompatibility, anti-apoptosis property, and chemotactic effects for mesenchymal stromal cells and endothelial cells through SDF-1-CXCR4 axis. Moreover, intramyocardial injection of this composite hydrogel to the infarcted area leads to the promotion of angiogenesis and inhibition of fibrosis, reducing the infarction size and improving the cardiac function. The combination of natural biomaterials, exogenous cells, and bioactive factors shows potential for MI treatment in the clinical application.


Assuntos
Quimiocina CXCL12 , Matriz Extracelular Descelularizada , Hidrogéis , Infarto do Miocárdio , Miócitos Cardíacos , Animais , Humanos , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacologia , Matriz Extracelular Descelularizada/química , Matriz Extracelular Descelularizada/farmacologia , Células Endoteliais , Matriz Extracelular , Hidrogéis/farmacologia , Infarto do Miocárdio/terapia , Miócitos Cardíacos/metabolismo , Regeneração , Suínos
3.
ACS Biomater Sci Eng ; 8(10): 4486-4496, 2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36178141

RESUMO

Stromal cell-derived factor-1 alpha (SDF-1α, CXCL12) mediates the migration of circulating cells to desired sites for tissue development, homeostasis, and regeneration and can be used to promote cardiac regeneration by recruiting stem cells. However, the use of SDF-1α in the injured heart necessitates not only higher binding affinity to its receptor, CXCR4+, but also better robustness against enzymatic degradation than other SDF-1 isoforms. Here, we conduct a screening of SDF-1α analog peptides that were designed by structure-based drug design (SBDD), a type of computer-aided drug design (CADD). We have developed in vitro and in vivo methods that enable us to estimate the effect of peptides on the migration of human mesenchymal stem cells (hMSCs) and cardiac regeneration in acute myocardial infarction (AMI)-induced animals, respectively. We demonstrate that one type of SDF-1α analog peptide, SDP-4, among the four analog peptides preselected by SBDD, is more potent than native SDF-1α for cardiac regeneration in myocardial infarction. It is interesting to note that the migratory effects of SDP-4 determined by a wound healing assay, a Transwell assay, and a 2D migration assay are comparable to those of SDF-1α. These results suggest that in vivo, as well as in vitro, screening of peptides developed by SBDD is a quintessential process to the development of a novel therapeutic compound for cardiac regeneration. Our finding also has an implication that the SDP-4 peptide is an excellent candidate for use in the regeneration of an AMI heart.


Assuntos
Quimiocina CXCL12 , Infarto do Miocárdio , Animais , Movimento Celular , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacologia , Quimiocina CXCL12/uso terapêutico , Desenho de Fármacos , Humanos , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Peptídeos/farmacologia , Peptídeos/uso terapêutico , Receptores CXCR4/metabolismo , Receptores CXCR4/uso terapêutico
4.
ACS Biomater Sci Eng ; 8(8): 3576-3588, 2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35899941

RESUMO

Critically sized skin flaps used to treat skin defects often suffer from necrosis due to insufficient blood supply. Hence there is an urgent need to improve the survival rate of skin flaps by promoting local angiogenesis. The delivery of growth factor loaded microcarriers have shown promise in enhancing defect repair, however, their rapid clearance from the defect site limits their regenerative potential. Thus, it is critical to develop microcarriers which can promote the sustained release of bioactive factors to effectively stimulate tissue repair. This study aimed to develop a stromal cell-derived factor 1 (SDF-1) loaded microcarrier coated with Matrigel (MC@SDF-1@Mat) to promote skin flap repair. SEM imaging showed that the surface of the microcarrier was coated by a porous Matrigel film. The drug release experiment showed that the Matrigel-coated microcarriers enhanced the sustained release of the model drug methylene blue when compared to uncoated group. MC@SDF-1@Mat significantly promoted the proliferation, migration, and angiogenesis of HUVECs via CCK-8, wound healing assay, and tube formation assay, respectively. Moreover, the murine random skin flap model was further established and treated. It was found that the flap necrosis area in the MC@SDF-1@Mat treated group was significantly reduced. H&E and Masson staining showed the histological structure and collagen organization exhibited a normal phenotype in the MC@SDF-1@Mat treated group. Additionally, CD31 immunohistochemical analysis showed that the MC@SDF-1@Mat treated group exhibited the greatest degree of neovascularization. In conclusion, our SDF-1 functionalized gelatin-based hydrogel microcarrier has potential clinical applications in promoting skin flap repair and drug delivery.


Assuntos
Quimiocina CXCL12 , Hidrogéis , Animais , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacologia , Preparações de Ação Retardada/farmacologia , Gelatina/química , Hidrogéis/farmacologia , Camundongos , Necrose
5.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35046049

RESUMO

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.


Assuntos
Carcinoma/etiologia , Carcinoma/metabolismo , Quimiocina CXCL12/metabolismo , Citotoxicidade Imunológica , Queratina-19/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Animais , Neoplasias da Mama , Carcinoma/patologia , Linhagem Celular Tumoral , Quimiocina CXCL12/química , Feminino , Humanos , Queratina-19/química , Masculino , Camundongos , Repetições de Microssatélites , Neoplasias Pancreáticas , Ligação Proteica , Multimerização Proteica , Neoplasias Pancreáticas
6.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-34769095

RESUMO

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.


Assuntos
Alginatos/química , Quimiocina CXCL12/química , Fator 2 de Crescimento de Fibroblastos/química , Heparina/química , Fator A de Crescimento do Endotélio Vascular/química , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana , Humanos , Microesferas , Engenharia Tecidual
7.
ACS Appl Mater Interfaces ; 13(31): 36880-36893, 2021 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-34324286

RESUMO

Existing local drug delivery systems for periodontitis suffer from poor antibacterial effect and unsatisfied periodontal regeneration. In this study, a smart gingipain-responsive hydrogel (PEGPD@SDF-1) was synthesized as an environmentally sensitive carrier for on-demand drug delivery. The PEGPD@SDF-1 hydrogel was synthesized from polyethylene glycol diacrylate (PEG-DA) based scaffolds, dithiothreitol (DTT), and a novel designed functional peptide module (FPM) via Michael-type addition reaction, and the hydrogel was further loaded with stromal cell derived factor-1 (SDF-1). The FPM exhibiting a structure of anchor peptide-short antimicrobial peptide (SAMP)-anchor peptide could be cleaved by gingipain specifically, and the SAMP was released out of the hydrogel for antibacterial effect in response to gingipain. The hydrogel properties were characterized by scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR), swelling ratio analysis, degradation evaluation, and release curve description of the SAMP and SDF-1. Results in vitro indicated the PEGPD@SDF-1 hydrogel exhibited preferable biocompatibility and could promote the proliferation, migration, and osteogenic differentiation of periodontal ligament stem cells (PDLSCs). Antibacterial testing demonstrated that the PEGPD@SDF-1 hydrogel released the SAMP stressfully in response to gingipain stimulation, thereby strongly inhibiting the growth of Porphyromonas gingivalis. Furthermore, the study in vivo indicated that the PEGPD@SDF-1 hydrogel inhibited P. gingivalis reproduction, created a low-inflammatory environment, facilitated the recruitment of CD90+/CD34- stromal cells, and induced osteogenesis. Taken together, these results suggest that the gingipain-responsive PEGPD@SDF-1 hydrogel could facilitate in situ periodontal tissue regeneration and is a promising candidate for the on-demand local drug delivery system for periodontitis.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Quimiocina CXCL12/uso terapêutico , Portadores de Fármacos/química , Cisteína Endopeptidases Gingipaínas/metabolismo , Hidrogéis/química , Periodontite/tratamento farmacológico , Animais , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/uso terapêutico , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/metabolismo , Peptídeos Catiônicos Antimicrobianos/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Movimento Celular , Quimiocina CXCL12/química , Portadores de Fármacos/síntese química , Liberação Controlada de Fármacos , Hidrogéis/síntese química , Masculino , Osteogênese/efeitos dos fármacos , Ligamento Periodontal/citologia , Periodontite/metabolismo , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Ácidos Polimetacrílicos/síntese química , Ácidos Polimetacrílicos/química , Porphyromonas gingivalis/efeitos dos fármacos , Ratos Wistar , Células-Tronco
8.
Biol Chem ; 402(11): 1453-1464, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34218538

RESUMO

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.


Assuntos
Quimiocina CXCL12/metabolismo , Glicosaminoglicanos/metabolismo , Hidrogéis/metabolismo , Quimiocina CXCL12/química , Glicosaminoglicanos/química , Humanos , Hidrogéis/síntese química , Hidrogéis/química , Células-Tronco Mesenquimais/metabolismo , Estrutura Molecular
9.
Int J Nanomedicine ; 15: 9337-9353, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33262591

RESUMO

PURPOSE: Autologous bone grafts are the gold standard for treating bone defects. However, limited bone supply and morbidity at the donor site restrict its extensive use. Therefore, developing bone graft materials as an alternative to autologous grafts has gained considerable attention. Injectable hydrogels endowed with osteogenic potential have the ability to fill irregular bone defects using minimally invasive procedures and have thus been attracting researchers' attention. However, from a clinical perspective, most fabrication methods employed for the current injectable osteogenic hydrogels are difficult and inconvenient. In the current study, we fabricated an injectable osteogenic hydrogel using a simple and convenient strategy. MATERIALS AND METHODS: Gelatin-methacryloyl (GelMA) pre-polymer was synthetized. Nano silicate (SN) and stromal cell-derived factor-1 alpha (SDF-1α) were introduced into the pre-polymer to achieve injectability, controlled release property, excellent osteogenic ability, and efficient stem cell homing. RESULTS: The GelMA-SN-SDF-1α demonstrated excellent injectability via a 17-G needle at room temperature. The loaded SDF-1α exhibited a long-term controlled release pattern and efficiently stimulated MSC migration and homing. The GelMA-SN-SDF-1α hydrogel amplified cell spreading, migration, osteogenic-related biomarker expression, and matrix mineralization. The GelMA-SN-SDF-1α hydrogel filled critical-sized calvaria defects in rats and demonstrated excellent bone regeneration ability, as assessed using micro-CT scanning and histomorphometric staining. CONCLUSION: The GelMA-SN-SDF-1α hydrogel provides a simple and convenient strategy for the fabrication of injectable osteogenic graft materials.


Assuntos
Osso e Ossos/citologia , Quimiocina CXCL12/química , Gelatina/química , Hidrogéis/química , Nanoestruturas/química , Silicatos/química , Engenharia Tecidual , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Regeneração Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiologia , Movimento Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Ratos
10.
Molecules ; 25(20)2020 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-33092204

RESUMO

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.


Assuntos
Aminoácidos/antagonistas & inibidores , Quimiocina CXCL12/antagonistas & inibidores , Avaliação Pré-Clínica de Medicamentos , Ligantes , Aminoácidos/química , Sítios de Ligação/efeitos dos fármacos , Quimiocina CXCL12/química , Química Computacional , Humanos , Ligação de Hidrogênio/efeitos dos fármacos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Ligação Proteica/efeitos dos fármacos , Relação Quantitativa Estrutura-Atividade , Receptores CXCR4/química , Interface Usuário-Computador
11.
Sci Signal ; 13(640)2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32665413

RESUMO

Because of their prominent roles in development, cancer, and HIV, the chemokine receptor CXCR4 and its ligand CXCL12 have been the subject of numerous structural and functional studies, but the determinants of ligand binding, selectivity, and signaling are still poorly understood. Here, building on our latest structural model, we used a systematic mutagenesis strategy to dissect the functional anatomy of the CXCR4-CXCL12 complex. Key charge swap mutagenesis experiments provided evidence for pairwise interactions between oppositely charged residues in the receptor and chemokine, confirming the accuracy of the predicted orientation of the chemokine relative to the receptor and providing insight into ligand selectivity. Progressive deletion of N-terminal residues revealed an unexpected contribution of the receptor N terminus to chemokine signaling. This finding challenges a longstanding "two-site" hypothesis about the essential features of the receptor-chemokine interaction in which the N terminus contributes only to binding affinity. Our results suggest that although the interaction of the chemokine N terminus with the receptor-binding pocket is the key driver of signaling, the signaling amplitude depends on the extent to which the receptor N terminus binds the chemokine. Together with systematic characterization of other epitopes, these data enable us to propose an experimentally consistent structural model for how CXCL12 binds CXCR4 and initiates signal transmission through the receptor transmembrane domain.


Assuntos
Quimiocina CXCL12/química , Modelos Moleculares , Complexos Multiproteicos/química , Receptores CXCR4/química , Animais , Células CHO , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Cricetulus , Células HEK293 , Humanos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Mutagênese Sítio-Dirigida , Estrutura Quaternária de Proteína , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
12.
J Leukoc Biol ; 107(6): 1123-1135, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32374043

RESUMO

Chemokines play critical roles in numerous physiologic and pathologic processes through their action on seven-transmembrane (TM) receptors. The N-terminal domain of chemokines, which is a key determinant of signaling via its binding within a pocket formed by receptors' TM helices, can be the target of proteolytic processing. An illustrative case of this regulatory mechanism is the natural processing of CXCL12 that generates chemokine variants lacking the first two N-terminal residues. Whereas such truncated variants behave as antagonists of CXCR4, the canonical G protein-coupled receptor of CXCL12, they are agonists of the atypical chemokine receptor 3 (ACKR3/CXCR7), suggesting the implication of different structural determinants in the complexes formed between CXCL12 and its two receptors. Recent analyses have suggested that the CXCL12 N-terminus first engages the TM helices of ACKR3 followed by the receptor N-terminus wrapping around the chemokine core. Here we investigated the first stage of ACKR3-CXCL12 interactions by comparing the activity of substituted or N-terminally truncated variants of CXCL12 toward CXCR4 and ACKR3. We showed that modification of the first two N-terminal residues of the chemokine (K1R or P2G) does not alter the ability of CXCL12 to activate ACKR3. Our results also identified the K1R variant as a G protein-biased agonist of CXCR4. Comparative molecular dynamics simulations of the complexes formed by ACKR3 either with CXCL12 or with the P2G variant identified interactions between the N-terminal 2-4 residues of CXCL12 and a pocket formed by receptor's TM helices 2, 6, and 7 as critical determinants for ACKR3 activation.


Assuntos
Quimiocina CXCL12/química , AMP Cíclico/química , Receptores CXCR4/química , Receptores CXCR/química , Sequência de Aminoácidos , Benzilaminas , Sítios de Ligação , Quimiocina CXCL11/química , Quimiocina CXCL11/genética , Quimiocina CXCL11/metabolismo , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Ciclamos , AMP Cíclico/metabolismo , Expressão Gênica , Células HEK293 , Compostos Heterocíclicos/química , Compostos Heterocíclicos/farmacologia , Humanos , Simulação de Dinâmica Molecular , Mutação , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Receptores CXCR/genética , Receptores CXCR/metabolismo , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , beta-Arrestinas/genética , beta-Arrestinas/metabolismo
13.
PLoS Biol ; 18(4): e3000656, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32271748

RESUMO

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.


Assuntos
Quimiocina CXCL12/química , Quimiocina CXCL12/metabolismo , Receptores CXCR4/química , Receptores CXCR4/metabolismo , Animais , Sítios de Ligação , Western Blotting , Quimiocina CXCL12/genética , Cisteína/química , Cisteína/genética , Dissulfetos/química , Citometria de Fluxo , Células HEK293 , Humanos , Insetos/citologia , Modelos Moleculares , Mutação , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Receptores CXCR4/genética , beta-Arrestinas/metabolismo
14.
Carbohydr Polym ; 237: 116107, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32241442

RESUMO

This work aims to design biocompatible aerogel sponges that can host and control the release of stromal cell-derived factor-1α (SDF-1α or CXCL12), a key protein for applications ranging from regenerative medicine to cancer therapy (notably for neural tissues). Miscibility of silk fibroin (SF) and hyaluronic acid (HA) was investigated by means of fluorescence and scanning electron microscopy to identify processing conditions. Series of freeze-dried sponges were prepared by associating and cross-linking within the same 3D structure, HA, SF, poly-l-lysine (PLL) and heparin (hep). Aerogel sponges presented high swelling degree and porosity (∼90 %), adequate mean pore diameter (ca. 60 µm) and connectivity for welcoming cells, and a soft texture close to that of the brain (6-13 kPa Young's Modulus). Addition of SF yielded sponges with slower biodegradation. SF-HA and SF-HA-hep sponges retained 75 % and 93 % of the SDF-1α respectively after 7 days and were found to be cytocompatible in vitro.


Assuntos
Materiais Biocompatíveis/química , Géis , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/síntese química , Quimiocina CXCL12/química , Fibroínas/química , Géis/síntese química , Géis/química , Heparina/química , Ácido Hialurônico/química , Camundongos , Células NIH 3T3 , Porosidade
15.
Biomed Mater ; 15(3): 035013, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-31694001

RESUMO

Optimized biocompatibility is crucial for the durability of cardiovascular implants. Previously, a combined coating with fibronectin (FN) and stromal cell-derived factor 1α (SDF1α) has been shown to accelerate the in vivo cellularization of synthetic vascular grafts and to reduce the calcification of biological pulmonary root grafts. In this study, we evaluate the effect of side-specific luminal SDF1α coating and adventitial FN coating on the in vivo cellularization and degeneration of decellularized rat aortic implants. Aortic arch vascular donor grafts were detergent-decellularized. The luminal graft surface was coated with SDF1α, while the adventitial surface was coated with FN. SDF1α-coated and uncoated grafts were infrarenally implanted (n = 20) in rats and followed up for up to eight weeks. Cellular intima population was accelerated by luminal SDF1α coating at two weeks (92.4 ± 2.95% versus 61.1 ± 6.51% in controls, p < 0.001). SDF1α coating inhibited neo-intimal hyperplasia, resulting in a significantly decreased intima-to-media ratio after eight weeks (0.62 ± 0.15 versus 1.35 ± 0.26 in controls, p < 0.05). Furthermore, at eight weeks, media calcification was significantly decreased in the SDF1α group as compared to the control group (area of calcification in proximal arch region 1092 ± 517 µm2 versus 11 814 ± 1883 µm2, p < 0.01). Luminal coating with SDF1α promotes early autologous intima recellularization in vivo and attenuates neo-intima hyperplasia as well as calcification of decellularized vascular grafts.


Assuntos
Prótese Vascular , Quimiocina CXCL12/química , Materiais Revestidos Biocompatíveis , Fibronectinas/química , Músculo Esquelético/inervação , Regeneração Nervosa , Animais , Bioprótese , Diferenciação Celular , Quimiotaxia , Reagentes de Ligações Cruzadas/química , Eletrofisiologia , Matriz Extracelular/metabolismo , Heparina , Laminina/química , Masculino , Músculo Esquelético/metabolismo , Neuritos/metabolismo , Células PC12 , Polímeros/química , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/patologia , Células Estromais , Enxerto Vascular , Caminhada
16.
Stem Cells Dev ; 29(4): 235-248, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31797735

RESUMO

Glial scars formed after brain injuries provide permissive cues for endogenous neural precursor/stem cells (eNP/SCs) to undergo astrogenesis rather than neurogenesis. Following brain injury, eNP/SCs from the subventricular zone leave their niche, migrate to the injured cortex, and differentiate into reactive astrocytes that contribute to glial scar formation. In vivo neuronal reprogramming, directly converting non-neuronal cells such as reactive astrocytes or NG2 glia into neurons, has greatly improved brain injury repair strategies. However, reprogramming carries a high risk of future clinical applications such as tumorigenicity, involving virus. In this study, we constructed a neural matrix to alter the adverse niche at the injured cortex, enabling eNP/SCs to differentiate into functional neurons. We found that the neural matrix functioned as a "glial trap" that largely concentrated and limited reactive astrocytes to the core of the lesion area, thus altering the adverse niche. The eNP/SCs migrated toward the injured cortex and differentiated into functional neurons. In addition, regenerated neurites extended across the boundary of the injured cortex. Mice treated with the neural matrix demonstrated significant behavioral recovery. For the first time, we induced eNP/SC-derived functional neurons in the cortex after brain injury without the use of viruses, microRNAs, or small molecules. Our novel strategy of applying this "glial trap" to obtain functional neurons in the injured cortex may provide a safer and more natural therapeutic alternative to reprogramming in future clinical applications.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Reprogramação Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Fator Neurotrófico Derivado do Encéfalo/química , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacologia , Condroitina ABC Liase/química , Condroitina ABC Liase/farmacologia , Modelos Animais de Doenças , Proteínas Imobilizadas/química , Proteínas Imobilizadas/farmacologia , Ventrículos Laterais/citologia , Ventrículos Laterais/fisiologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Fator de Crescimento Neural/química , Fator de Crescimento Neural/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Teste de Desempenho do Rota-Rod , Nicho de Células-Tronco/efeitos dos fármacos
17.
ACS Appl Mater Interfaces ; 11(16): 14608-14618, 2019 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-30938503

RESUMO

Continuous delivery of growth factors to the injury site is crucial to creating a favorable microenvironment for cartilage injury repair. In the present study, we fabricated a novel sustained-release scaffold, stromal-derived factor-1α (SDF-1α)/transforming growth factor-ß1 (TGF-ß1)-loaded silk fibroin-porous gelatin scaffold (GSTS). GSTS persistently releases SDF-1α and TGF-ß1, which enhance cartilage repair by facilitating cell homing and chondrogenic differentiation. Scanning electron microscopy showed that GSTS is a porous microstructure and the protein release assay demonstrated the sustainable release of SDF-1α and TGF-ß1 from GSTS. Bone marrow-derived mesenchymal stem cells (MSCs) maintain high in vitro cell activity and excellent cell distribution and phenotype after seeding into GSTS. Furthermore, MSCs acquired enhanced chondrogenic differentiation capability in the TGF-ß1-loaded scaffolds (GSTS or GST: loading TGF-ß1 only) and the conditioned medium from SDF-1α-loaded scaffolds (GSTS or GSS: loading SDF-1α only) effectively promoted MSCs migration. GSTS was transplanted into the osteochondral defects in the knee joint of rats, and it could promote cartilage regeneration and repair the cartilage defects at 12 weeks after transplantation. Our study shows that GSTS can facilitate in vitro MSCs homing, migration, chondrogenic differentiation and SDF-1α and TGF-ß1 have a synergistic effect on the promotion of in vivo cartilage forming. This SDF-1α and TGF-ß1 releasing GSTS have promising therapeutic potential in cartilage repair.


Assuntos
Cartilagem , Quimiocina CXCL12 , Condrogênese/efeitos dos fármacos , Fibroínas , Gelatina , Fator de Crescimento Transformador beta1 , Animais , Cartilagem/lesões , Cartilagem/metabolismo , Cartilagem/patologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacocinética , Quimiocina CXCL12/farmacologia , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Preparações de Ação Retardada/farmacologia , Fibroínas/química , Fibroínas/farmacocinética , Fibroínas/farmacologia , Gelatina/química , Gelatina/farmacocinética , Gelatina/farmacologia , Masculino , Porosidade , Ratos , Fator de Crescimento Transformador beta1/química , Fator de Crescimento Transformador beta1/farmacocinética , Fator de Crescimento Transformador beta1/farmacologia
18.
Oncol Rep ; 41(1): 312-324, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30365110

RESUMO

Notable advances in treatment have been made and increases in the cure rates of pediatric leukemia have been achieved. However, the majority of children with relapsed disease are not expected to survive, with chemotherapy resistance acting as the principal cause of treatment failure. Interaction between leukemic cells and the bone marrow microenvironment is the primary cause of relapse. It was identified that a multi­protein membrane complex, formed by potassium voltage­gated channel subfamily H member 2 (hERG1) channels, the ß1 integrin subunit and the stromal cell­derived factor 12 (CXCL12) receptor, C­X­C chemokine receptor type 4 (CXCR4), exerts a role in mesenchymal stromal cell (MSC)­mediated chemoresistance in pediatric leukemias. hERG1 blockade was able to overcome chemoresistance in vitro and in vivo. As an alternative strategy to overcome chemoresistance, the present study evaluated the effects of novel tools targeting the CXCR4/CXCL12 axis. The analysis of CXCL12 structural dynamics was used for the selection of a peptide (4­1­17) and a small molecule (8673), which interact with a transient hot spot, identified by a dynamic drug design approach. The present findings indicated that peptide 4­1­17 and small molecule 8673 inhibited leukemia cell proliferation and induced a pro­apoptotic effect, which was not reduced by the presence of MSCs. The combined treatment with 4­1­17 and 8673 had a stronger pro­apoptotic effect, particularly on cells cultured on MSCs in normoxic and hypoxic conditions, and was able to overcome MSC­induced resistance to cytarabine. Overall, the targeting of CXCL12 and the ensuing inhibition of the CXCR4/CXCL12 axis may be proposed as an alternative strategy to overcome chemoresistance in leukemia.


Assuntos
Quimiocina CXCL12/metabolismo , Citarabina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Leucemia/metabolismo , Peptídeos/farmacologia , Receptores CXCR4/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Medula Óssea/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL12/química , Humanos , Leucemia/tratamento farmacológico , Simulação de Dinâmica Molecular , Peptídeos/síntese química , Peptídeos/química , Ligação Proteica/efeitos dos fármacos , Receptores CXCR4/química , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/síntese química , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
19.
J Med Chem ; 61(17): 7671-7686, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30106292

RESUMO

We previously reported Chalcone-4 (1) that binds the chemokine CXCL12, not its cognate receptors CXCR4 or CXCR7, and neutralizes its biological activity. However, this neutraligand suffers from limitations such as poor chemical stability, solubility, and oral activity. Herein, we report on the discovery of pyrimidinone 57 (LIT-927), a novel neutraligand of CXCL12 which displays a higher solubility than 1 and is no longer a Michael acceptor. While both 1 and 57 reduce eosinophil recruitment in a murine model of allergic airway hypereosinophilia, 57 is the only one to display inhibitory activity following oral administration. Thereby, we here describe 57 as the first orally active CXCL12 neutraligand with anti-inflammatory properties. Combined with a high binding selectivity for CXCL12 over other chemokines, 57 represents a powerful pharmacological tool to investigate CXCL12 physiology in vivo and to explore the activity of chemokine neutralization in inflammatory and related diseases.


Assuntos
Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/farmacologia , Quimiocina CXCL12/metabolismo , Síndrome Hipereosinofílica/tratamento farmacológico , Pirimidinonas/química , Pirimidinonas/farmacologia , Administração Oral , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/farmacocinética , Quimiocina CXCL12/química , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Transferência Ressonante de Energia de Fluorescência , Humanos , Hipersensibilidade/tratamento farmacológico , Hipersensibilidade/etiologia , Masculino , Camundongos Endogâmicos BALB C , Modelos Moleculares , Pirimidinonas/administração & dosagem , Pirimidinonas/metabolismo , Pirimidinonas/farmacocinética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Relação Estrutura-Atividade
20.
Arterioscler Thromb Vasc Biol ; 38(7): e117-e134, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29853570

RESUMO

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.


Assuntos
Indutores da Angiogênese/farmacologia , Aorta Abdominal/cirurgia , Implante de Prótese Vascular/instrumentação , Prótese Vascular , Quimiocina CXCL12/farmacologia , Materiais Revestidos Biocompatíveis , Colágeno Tipo I/química , Neovascularização Fisiológica/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Poliésteres/química , Substância P/farmacologia , Indutores da Angiogênese/química , Animais , Aorta Abdominal/diagnóstico por imagem , Aorta Abdominal/patologia , Aorta Abdominal/fisiopatologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Quimiocina CXCL12/química , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Neointima , Fragmentos de Peptídeos/química , Desenho de Prótese , Ratos Sprague-Dawley , Substância P/química , Fatores de Tempo , Ultrassonografia Doppler em Cores , Grau de Desobstrução Vascular , Remodelação Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA