Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Int J Biol Macromol ; 269(Pt 1): 132040, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38702003

RESUMO

Decellularized vascular tissue has high potential as a tissue-engineered vascular graft because of its similarity to native vessels in terms of mechanical strength. However, exposed collagen on the tissue induces blood coagulation, and low hemocompatibility is a major obstacle to its vascular application. Here we report that freeze-drying and ethanol treatment effectively modify collagen fiber structure and drastically reduce blood coagulation on the graft surface without exogenous chemical modification. Decellularized carotid artery of ostrich was treated with freeze-drying and ethanol solution at concentrations ranging between 5 and 99.5 %. Collagen fiber distance in the graft was narrowed by freeze-drying, and the non-helical region increased by ethanol treatment. Although in vitro blood coagulation pattern was similar on the grafts, platelet adhesion on the grafts was largely suppressed by freeze-drying and ethanol treatments. Ex vivo blood circulation tests also indicated that the adsorption of platelets and Von Willebrand Factor was largely reduced to approximately 80 % by ethanol treatment. These results indicate that structural modification of collagen fibers in decellularized tissue reduces blood coagulation on the surface by inhibiting platelet adhesion.


Assuntos
Coagulação Sanguínea , Colágeno , Adesividade Plaquetária , Animais , Adesividade Plaquetária/efeitos dos fármacos , Coagulação Sanguínea/efeitos dos fármacos , Colágeno/química , Engenharia Tecidual/métodos , Teste de Materiais , Liofilização , Prótese Vascular , Alicerces Teciduais/química , Plaquetas/metabolismo , Plaquetas/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Artérias Carótidas/efeitos dos fármacos , Humanos , Etanol/química
2.
Acta Biomater ; 176: 116-127, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38232911

RESUMO

Decellularized vessels (DVs) have the potential to serve as available grafts for small-diameter vascular (<6 mm) reconstruction. However, the absence of functional endothelia makes them likely to trigger platelet aggregation and thrombosis. Luminal surface modification is an efficient approach to prevent thrombosis and promote endothelialization. Previously, we identified a hemocompatible peptide, HGGVRLY, that showed endothelial affinity and antiplatelet ability. By conjugating HGGVRLY with a phenylazide group, we generated a photoreactive peptide that can be modified onto multiple materials, including non-denatured extracellular matrices. To preserve the natural collagen of DVs as much as possible, we used a lower ultrahydrostatic pressure than that previously reported to prepare decellularized grafts. The photoreactive HGGVRLY peptide could be modified onto DV grafts via UV exposure for only 2 min. Modified DVs showed improved endothelial affinity and antiplatelet ability in vitro. When rat abdominal aortas were replaced with DVs, modified DVs with more natural collagen demonstrated the highest patent rate after 10 weeks. Moreover, the photoreactive peptide remained on the lumen surface of DVs over two months after implantation. Therefore, the photoreactive peptide could be efficiently and sustainably modified onto DVs with more natural collagens, resulting in improved hemocompatibility. STATEMENT OF SIGNIFICANCE: We employed a relatively lower ultrahydrostatic pressure to prepare decellularized vessels (DVs) with less denatured collagens to provide a more favorable environment for cell migration and proliferation. The hemocompatibility of DV luminal surface can be enhanced by peptide modification, but undenatured collagens are difficult to modify. We innovatively introduce a phenylazide group into the hemocompatible peptide HGGVRLY, which we previously identified to possess endothelial affinity and antiplatelet ability, to generate a photoreactive peptide. The photoreactive peptide can be efficiently and stably modified onto DVs with more natural collagens. DV grafts modified with photoreactive peptide exhibit enhanced in vivo patency. Furthermore, the sustainability of photoreactive peptide modification on DV grafts within bloodstream is evident after two months of transplantation.


Assuntos
Azidas , Prótese Vascular , Trombose , Ratos , Animais , Peptídeos/farmacologia , Trombose/prevenção & controle , Trombose/metabolismo , Colágeno/farmacologia
3.
Acta Biomater ; 176: 221-233, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38242190

RESUMO

Intramyocardial hydrogel injection is a promising therapy to prevent negative remodeling following myocardial infarction (MI). In this study, we report a mechanism for in-situ gel formation without external stimulation, resulting in an injectable and tissue-retainable hydrogel for MI treatment, and investigate its therapeutic outcomes. A liquid-like polymeric solution comprising poly(3-acrylamidophenylboronic acid-co-acrylamide) (BAAm), polyvinyl alcohol (PVA), and sorbitol (S) increases the viscous modulus by reducing the pre-added sorbitol concentration is developed. This solution achieves a sol-gel transition in-vitro in heart tissue by spontaneously diffusing the sorbitol. After intramyocardial injection, the BAAm/PVA/S with lower initial viscous modulus widely spreads in the myocardium and gelate compared to a viscoelastic alginate (ALG) hydrogel and is retained longer than the BAAm/S solution. Serial echocardiogram analyses prove that injecting the BAAm/PVA/S into the hearts of subacute MI rats significantly increases the fraction shortening and ejection shortening and attenuates the expansion of systolic LV diameter for up to 21 d after injection compared to the saline injection as a control, but the ALG injection does not. In addition, histological evaluation shows that only the BAAm/PVA/S decreases the infarct size and increases the wall thickness 21 d after injection. The BAAm/PVA/S intramyocardial injection is better at restraining systolic ventricular dilatation and cardiac failure in the rat MI model than in the control groups. Our findings highlight an effective injectable hydrogel therapy for MI by optimizing injectability-dependent distribution and retention of injected material. STATEMENT OF SIGNIFICANCE: In-situ gelling material is a promising strategy for intramyocardial hydrogel injection therapy for myocardial infarction (MI). Since the sol-gel transition of reported materials is driven by external stimulation such as temperature, pH, or ultraviolet, their application in vivo remains challenging. In this study, we first reported a synthetic in-situ gelling material (BAAm/PVA/S) whose gelation is stimulated by spontaneously reducing pre-added sorbitol after contacting the heart tissue. The BAAm/PVA/S solution spreads evenly, and is retained for at least 21 d in the heart tissue. Our study demonstrated that intramyocardial injection of the BAAm/PVA/S with more extensive distribution and longer retention had better effects on preventing LV dilation and improving cardiac function after MI than that of viscoelastic ALG and saline solution. We expect that these findings provide fundamental information for the optimum design of injectable biomaterials for treating MI.


Assuntos
Alprenolol/análogos & derivados , Hidrogel de Polietilenoglicol-Dimetacrilato , Infarto do Miocárdio , Ratos , Animais , Hidrogel de Polietilenoglicol-Dimetacrilato/uso terapêutico , Hidrogéis/farmacologia , Hidrogéis/uso terapêutico , Infarto do Miocárdio/patologia , Sorbitol/farmacologia , Sorbitol/uso terapêutico
4.
Mater Today Bio ; 23: 100847, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37953756

RESUMO

Although the clinical application of cell-free tissue-engineered vascular grafts (TEVGs) has been proposed, vascular tissue regeneration mechanisms have not been fully clarified. Here, we report that monocyte subpopulations reconstruct vascular-like tissues through integrin signaling. An Arg-Glu-Asp-Val peptide-modified acellular long-bypass graft was used as the TEVG, and tissue regeneration in the graft was evaluated using a cardiopulmonary pump system and porcine transplantation model. In 1 day, the luminal surface of the graft was covered with cells that expressed CD163, CD14, and CD16, which represented the monocyte subpopulation, and they exhibited proliferative and migratory abilities. RNA sequencing showed that captured cells had an immune-related phenotype similar to that of monocytes and strongly expressed cell adhesion-related genes. In vitro angiogenesis assay showed that tube formation of the captured cells occurred via integrin signal activation. After medium- and long-term graft transplantation, the captured cells infiltrated the tunica media layer and constructed vascular with a CD31/CD105-positive layer and an αSMA-positive structure after 3 months. This finding, including multiple early-time observations provides clear evidence that blood-circulating monocytes are directly involved in vascular remodeling.

5.
Colloids Surf B Biointerfaces ; 230: 113525, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37634287

RESUMO

Microvascular imaging is required to understand tumor angiogenesis development; however, an appropriate whole-body imaging method has not yet been established. Here, we successfully developed a supramolecular magnetic resonance (MR) contrast agent for long-term whole-tissue observation in a single individual. Fluorescein- and Gd-chelate-conjugated polyethylene glycols (PEGs) were synthesized, and their structures were optimized. Spectroscopic and pharmacokinetic analyses suggested that the fluorescein-conjugated linear and 8-arm PEGs with a molecular weight of approximately 10 kDa were suitable to form a supramolecular structure to visualize the microvessel structure and blood circulation. Microvascular formation was evaluated in a glioma cell transplantation model, and neovascularization around the glioma tissue at 5 days was observed, with the contrast agent leaking out into the cancer tissue. In contrast, after 12 days, microvessel structures were formed inside the glioma tissue, but the agents did not leak out. These imaging data for the first time proved that the microvessels formed inside cancer tissues at the early stage are very leaky, but that they form continuous microvessels after 12 days.


Assuntos
Meios de Contraste , Glioma , Humanos , Imageamento por Ressonância Magnética , Neovascularização Patológica/diagnóstico por imagem , Glioma/diagnóstico por imagem , Fluoresceína , Polietilenoglicóis , Espectroscopia de Ressonância Magnética
6.
Biomater Adv ; 147: 213324, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36796198

RESUMO

Abnormal proliferation of vascular smooth muscle cells (VSMCs) induces graft anastomotic stenosis, resulting in graft failure. Herein, we developed a drug-loaded tissue-adhesive hydrogel as artificial perivascular tissue to suppress VSMCs proliferation. Rapamycin (RPM), an anti-stenosis drug, is selected as the drug model. The hydrogel was composed of poly (3-acrylamidophenylboronic acid-co-acrylamide) (BAAm) and polyvinyl alcohol. Since phenylboronic acid reportedly binds to sialic acid of glycoproteins which is distributed on the tissues, the hydrogel is expected to be adherent to the vascular adventitia. Two hydrogels containing 25 or 50 mg/mL of BAAm (BAVA25 and BAVA50, respectively) were prepared. A decellularized vascular graft with a diameter of <2.5 mm was selected as a graft model. Lap-shear test indicates that both hydrogels adhered to the graft adventitia. In vitro release test indicated that 83 and 73 % of RPM in BAVA25 and BAVA50 hydrogels was released after 24 h, respectively. When VSMCs were cultured with RPM-loaded BAVA hydrogels, their proliferation was suppressed at an earlier stage in RPM-loaded BAVA25 hydrogels compared to RPM-loaded BAVA50 hydrogels. An in vivo preliminary test reveals that the graft coated with RPM-loaded BAVA25 hydrogel shows better graft patency for at least 180 d than the graft coated with RPM-loaded BAVA50 hydrogel or without hydrogel. Our results suggest that RPM-loaded BAVA25 hydrogel with tissue adhesive characteristics has potential to improve decellularized vascular graft patency.


Assuntos
Sirolimo , Enxerto Vascular , Sirolimo/farmacologia , Hidrogéis , Prótese Vascular
8.
J Biomed Mater Res A ; 110(3): 547-558, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34486215

RESUMO

Recently, a decellularized microvascular graft (inner diameter: 0.6 mm) modified with the integrin α4ß1 ligand, REDV, was developed to provide an alternative to autologous-vein grafting in reconstructive microsurgery, showing good early-stage patency under arterial flow in rats. This consecutive study evaluated its potential utility not only as an arterial substitute, but also as a venous substitute, using a rat-tail replantation model. Graft remodeling depending on hemodynamic status was also investigated. ACI rat tail arteries were decellularized via ultra-high-hydrostatic pressure treatment and modified with REDV to induce antithrombogenic interfaces and promote endothelialization after implantation. Grafts were implanted into the tail artery and vein to re-establish blood circulation in amputated Lewis rat tails (n = 12). The primary endpoint was the survival of replants. Secondary endpoints were graft patency, remodeling, and regeneration for 6 months. In all but three cases with technical errors or postoperative self-mutilation, tails survived without any evidence of ischemia or congestion. Six-month Kaplan-Meier patency was 100% for tail-artery implanted grafts and 62% for tail-vein implanted grafts. At 6 months, the neo-tunica media (thickness: 95.0 µm in tail-artery implanted grafts, 9.3 µm in tail-vein implanted grafts) was regenerated inside the neo-intima. In conclusion, the microvascular grafts functioned well both as arterial and venous paths of replanted-rat tails, with different remodeling under arterial and venous conditions.


Assuntos
Artérias , Túnica Média , Animais , Artérias/transplante , Ratos , Ratos Endogâmicos ACI , Ratos Endogâmicos Lew , Grau de Desobstrução Vascular
9.
J Mater Chem B ; 10(14): 2544-2550, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-34787632

RESUMO

Decellularized tissue is expected to be utilized as a regenerative scaffold. However, the migration of host cells into the central region of the decellularized tissues is minimal because the tissues are mainly formed with dense collagen and elastin fibers. This results in insufficient tissue regeneration. Herein, it is demonstrated that host cell migration can be accelerated by using decellularized tissue with a patterned pore structure. Patterned pores with inner diameters of 24.5 ± 0.4 µm were fabricated at 100, 250, and 500 µm intervals in the decellularized vascular grafts via laser ablation. The grafts were transplanted into rat subcutaneous tissue for 1, 2, and 4 weeks. All the microporous grafts underwent faster recellularization with macrophages and fibroblast cells than the non-porous control tissue. In the case of non-porous tissue, the cells infiltrated approximately 50% of the area four weeks after transplantation. However, almost the entire area was occupied by the cells after two weeks when the micropores were aligned at a distance of less than 250 µm. These results suggest that host cell infiltration depends on the micropore interval, and a distance shorter than 250 µm can accelerate cell migration into decellularized tissues.


Assuntos
Transplantes , Enxerto Vascular , Animais , Prótese Vascular , Colágeno , Ratos , Cicatrização
10.
Mater Sci Eng C Mater Biol Appl ; 129: 112381, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34579900

RESUMO

Ligand-immobilization to stents and vascular grafts is expected to promote endothelialization by capturing flowing endothelial progenitor cells (EPCs). However, the optimized ligand density and linker structure have not been fully elucidated. Here, we report that flowing EPCs were selectively captured by the REDV peptide conjugated with a short linker. The microchannel surface was modified with the REDV peptide via Gly-Gly-Gly (G3), (Gly-Gly-Gly)3 (G9), and diethylene glycol (diEG) linkers, and the moving velocity and captured ratio were evaluated. On the unmodified microchannels, the moving velocity of the cells exhibited a unimodal distribution similar to the liquid flow. The velocity of the endothelial cells and EPCs on the peptide-immobilized surface indicated a bimodal distribution, and approximately 20 to 30% of cells moved slower than the liquid flow, suggesting that the cells were captured and rolled on the surface. When the immobilized ligand density was lower than 1 molecule/nm2, selective cell capture was observed only in REDV with G3 and diEG linkers, but not in G9 linkers. An in silico study revealed that the G9 linker tends to form a bent structure, and the REDV peptide is oriented to the substrate side. These results indicated that REDV captured the flowing EPC in a sequence-specific manner, and that the short linker was more adequate.


Assuntos
Células Progenitoras Endoteliais , Prótese Vascular , Adesão Celular , Dispositivos Lab-On-A-Chip , Peptídeos
11.
Plast Reconstr Surg ; 148(1): 71e-76e, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-34181614

RESUMO

BACKGROUND: Giant congenital melanocytic nevi are large skin lesions associated with a risk of malignant transformation. The authors developed a novel treatment to reconstruct full-thickness skin defects by combining an inactivated nevus as the autologous dermis and a cultured epidermal autograft. The first-in-human trial of this treatment was performed. METHODS: Patients with melanocytic nevi that were not expected to be closed by primary closure were recruited. The full-thickness nevus of the target was removed and inactivated by high hydrostatic pressurization at 200 MPa for 10 minutes. The inactivated nevus was sutured to the original site, and a cultured epidermal autograft was grafted onto it 4 weeks later. Patients were followed for up to 52 weeks. RESULTS: Ten patients underwent reimplantation of the pressurized nevus, and one patient dropped out. The recurrence of nevus at 52 weeks was not detected by pathological diagnosis in any patients. The L* value at 52 weeks was significantly higher than that of the target nevus. One patient received skin grafting due to contracture of the reconstructed skin. The epithelized area of the reconstructed skin, as the percentage of the original target nevus, was 55.5 ± 19.4 percent at 12 weeks and 85.0 ± 32.4 percent at 52 weeks. CONCLUSIONS: The inactivated nevus caused inflammation and contracture for several months. However, no recurrence was observed, and combination therapy using an inactivated nevus with a cultured epidermal autograft may therefore be a novel treatment of giant congenital melanocytic nevi. CLINICAL QUESTION/LEVEL OF EVIDENCE: Therapeutic, IV.


Assuntos
Derme/transplante , Nevo Pigmentado/cirurgia , Neoplasias Cutâneas/cirurgia , Transplante de Pele/métodos , Adolescente , Adulto , Autoenxertos/transplante , Criança , Pré-Escolar , Epiderme , Feminino , Humanos , Pressão Hidrostática , Masculino , Recidiva Local de Neoplasia , Estudos Prospectivos , Técnicas de Cultura de Tecidos , Transplante Autólogo , Resultado do Tratamento , Adulto Jovem
12.
Biomater Sci ; 9(3): 1034-1043, 2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33336665

RESUMO

Blood compatibility generally requires two contradictory characteristics: reduced protein/platelet adhesion and excellent endothelium-related cell affinity. To understand the effect of cell adhesion peptides on blood compatibility, the peptides REDV, RGD, and hemocompatible peptide-1 (HCP-1) were immobilized on an expanded polytetrafluorethylene (ePTFE) surface and evaluated in vitro, in situ, and in vivo. Since the terminal amino groups of functional peptides often have an important effect, a cysteine residue was added to the C terminal and used for immobilization to keep the terminal amino groups free. Maleimide groups were added to carboxylic groups of highly hydrophilic and biologically inert (bioinert) polymer chains grafted onto ePTFE and coupled with cysteine residues. In vitro tests revealed that free N-terminal HCP-1 and RGD-immobilized surfaces improved the adhesion and spread of human umbilical vein endothelial cells (HUVECs), while, unexpectedly, a free N-terminal adjacent to REDV suppressed cell affinity. In situ evaluation with a porcine closed-circuit system for 2 h showed that no platelets adhered to the modified ePTFE sutures due to the bioinert graft chain containing phosphorylcholine groups. Simultaneously, leukocyte-related and endothelium-related cells were observed on RGD-immobilized ePTFE sutures because RGD was recognized by broad types of cells. These cells were not observed on the HCP-1- and REDV-immobilized ePTFE sutures, which may be due to insufficient exposure time. HCP-1-modified ePTFE graft implantation in a porcine femorofemoral (FF) bypass model for 10 days showed that the thrombus layer was clearly mitigated by HCP-1 immobilization. This study suggests that the HCP-1-immobilized ePTFE surface has potential for long-term application by mitigating thrombus and supporting endothelial cell adhesion.


Assuntos
Oligopeptídeos , Peptídeos , Animais , Adesão Celular , Endotélio , Humanos , Propriedades de Superfície , Suínos
13.
Peptides ; 136: 170470, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33279572

RESUMO

Recently, artificial blood vessels modified by integrin α4ß1 ligand, such as REDV, showed endothelialization improvement and antithrombotic properties have been reported. Early endothelialization was affected by the type of circulating cells captured by the peptide in the initial transplantation state, however, it is still not clarified. In this study, we identified in vitro circulating cells bound with the peptides arginine-glutamic acid-aspartic acid-valine (REDV) or histidine-glycine-glycine-valine-arginine-leucine-tyrosine (HGGVRLY). The effect of free C- or N-terminal of HGGVRLY on the type of peptide-binding cells was also studied. The rat circulating cells were isolated from blood and incubated with 5(6)-carboxyfluorescein (5/6-FAM, F) labeled F-REDV (C-terminal free), F-HGGVRLY (C-terminal free), or HGGVRLY-F (N-terminal free). Furthermore, peptide-binding cells were identified by co-staining with various antibodies labeled with PE, PerCP/Cy5.5, or APC. N-terminal free HGGVRLY-F was found to bind to more circulating cells than C-terminal free F-REDV and F-HGGVRLY. The ratio of integrin α4ß1 positive cell bound with F-REDV, F-HGGVRLY, or HGGVRLY-F reached over 90 %, demonstrating that HGGVRLY is also a ligand of integrin α4ß1. Among identified cell types, we found that F-REDV mainly bounds with EPC and BMSC, while F-HGGVRLY with BMSC. HGGVRLY-F bounds with EPC and BMSC, exhibiting a higher EPC binding ratio than F-REDV and F-HGGVRLY.


Assuntos
Anticorpos/química , Integrina alfa4beta1/genética , Oligopeptídeos/química , Peptídeos/genética , Animais , Anticorpos/genética , Micropartículas Derivadas de Células/efeitos dos fármacos , Fluoresceínas/química , Humanos , Integrina alfa4beta1/química , Ligantes , Oligopeptídeos/genética , Peptídeos/química , Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Ratos
14.
Chem Commun (Camb) ; 56(79): 11807-11810, 2020 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-33021251

RESUMO

We report the supramolecular self-assembly of one fluorescein and three Gd-chelate conjugated 8-arm polyethylene glycols (8-arm PEG-FGd3) for visualising the capillaries of the brain in magnetic resonance imaging (MRI).


Assuntos
Encéfalo/irrigação sanguínea , Capilares/diagnóstico por imagem , Meios de Contraste/química , Corantes Fluorescentes/química , Compostos Heterocíclicos/química , Polietilenoglicóis/química , Animais , Fluoresceínas/química , Gadolínio/química , Imageamento por Ressonância Magnética , Masculino , Nanopartículas/química , Ratos Sprague-Dawley
15.
Biomacromolecules ; 21(8): 3092-3101, 2020 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-32649189

RESUMO

Early-stage hemocompatibility is indispensable for manufacturing tissue-engineered vascular grafts used in regenerative medicine. In this study, we report the in vivo blood response and patency of small-diameter synthetic vascular grafts modified with the Arg-Glu-Asp-Val (REDV) peptide. Vascular grafts were prepared by casting REDV-conjugated poly(depsipeptide-co-caprolactone) on a stainless-steel mandril (diameter: 1.8 mm). After implanting the grafts into the abdominal aorta of rats for 24 h, all three control grafts without the peptide and three out of the four REDV (control sequence) peptide-modified grafts showed occlusion. The luminal surfaces of these grafts were covered with thick thrombi. In contrast, all the grafts containing the REDV peptide were patent, and their luminal surfaces were covered with a thin layer of fibrin. These results indicated that the REDV peptide on the luminal surface effectively reduced early-stage fibrin clot deposition and formed the pseudo-endothelium layer in a peptide sequence-specific manner, resulting in graft patency.


Assuntos
Trombose , Enxerto Vascular , Animais , Prótese Vascular , Fibrina , Peptídeos/farmacologia , Ratos , Trombose/tratamento farmacológico
16.
Colloids Surf B Biointerfaces ; 193: 111113, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32447201

RESUMO

We recently reported in vitro suppression of platelet adhesion on expanded polytetrafluoroethylene (ePTFE) by surface grafting of poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC). However, this may be inadequate for long-term hemocompatibility of blood-contacting biomaterials, and it has led us to develop a strategy of circulating mononuclear cell-capture. ePTFE was treated with argon (Ar) plasma, and grafted with 2-methacryloyloxyethyl phosphorylcholine (MPC) and methacrylic acid (MAA), by glycidyl methacrylate (GMA)-anchored graft polymerization. Next, it was immobilized with integrin α4ß1-positive circulating blood cell-specific peptides, i.e., the traditional arginine-glutamic acid-aspartic acid-valine (REDV), and our original hemocompatible peptide-1 (HCP-1). Both the surfaces retained the anti-platelet property just like the PMPC-grafted surface, and revealed considerable affinity to human umbilical vein endothelial cells (HUVEC), which is a well-known in vitro integrin α4ß1-positive model. Better HUVEC spreading and proliferation was also confirmed, in terms of the cell extension property. Since coagulation and endothelialization on the materials compete in the body, they cannot be properly evaluated separately, in vitro. They were assessed by using an in situ porcine closed-circuit system for 18 h in the present study. Our findings suggest that poly(MPC-co-MAA) is a great ePTFE surface modifier, exhibiting good hemocompatibility in association with REDV/HCP-1 immobilization, which suppresses anti-platelet adhesion and enhances circulating cell capture simultaneously.


Assuntos
Materiais Biocompatíveis/farmacologia , Células Progenitoras Endoteliais/efeitos dos fármacos , Peptídeos/farmacologia , Fosforilcolina/análogos & derivados , Ácidos Polimetacrílicos/farmacologia , Animais , Materiais Biocompatíveis/química , Células Cultivadas , Humanos , Teste de Materiais , Estrutura Molecular , Tamanho da Partícula , Peptídeos/química , Fosforilcolina/química , Fosforilcolina/farmacologia , Adesividade Plaquetária/efeitos dos fármacos , Ácidos Polimetacrílicos/química , Politetrafluoretileno , Propriedades de Superfície , Suínos
17.
J Biomed Mater Res A ; 108(10): 2005-2014, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32323458

RESUMO

Because acellular vascular xenografts induce an immunological reaction through macrophage infiltration, they are conventionally crosslinked with glutaraldehyde (GA). However, the GA crosslinking reaction inhibits not only the host immune reaction around the graft but also the graft's enzymatic degradability, which is one of the key characteristics of acellular grafts that allow them to be replaced by host tissue. In this study, we used an 8-arm polyethylene glycol (PEG) to successfully suppress macrophage infiltration, without eliminating graft degradation. Decellularized ostrich carotid arteries were modified with GA or N-hydroxysuccinimide-activated 8-arm PEG (8-arm PEG-NHS), which has a molecular weight of 17 kDa. To evaluate the enzymatic degradation in vitro, the graft was immersed in a collagenase solution for 12 hr. The 8-arm PEG-modified graft was degraded to the same extent as the unmodified graft, but the GA-modified graft was not degraded. The graft was transplanted into rat subcutaneous tissue for up to 8 weeks. Although CD68-positive cells accumulated in the unmodified graft, they did not infiltrate into either modified graft. However, the GA-modified grafts calcified, but the 8-arm PEG-modified graft did not calcify after transplantation. These data suggested that 8-arm PEG-NHS is a promising modification agent for biodegradable vascular xenografts, to suppress acute macrophage infiltration only.


Assuntos
Implantes Absorvíveis , Prótese Vascular , Glutaral/química , Macrófagos/citologia , Polietilenoglicóis/química , Implantes Absorvíveis/efeitos adversos , Animais , Prótese Vascular/efeitos adversos , Artérias Carótidas/química , Reagentes de Ligações Cruzadas/química , Macrófagos/imunologia , Masculino , Ratos Sprague-Dawley , Struthioniformes
18.
J Biomater Sci Polym Ed ; 31(11): 1475-1488, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32338157

RESUMO

Small-caliber artificial blood vessels with inner diameters of smaller than 4 mm have not been put into practical use because of early thrombus formation and graft occlusion. To realize small-caliber artificial blood vessels with anti-thrombus property and long-term patency, one of the promising approaches is endothelialization of the lumen by tissue engineering approaches. Integrin α4ß1 on the endothelial cell membrane is known to act as a receptor for Arg-Glu-Asp-Val (REDV) tetra-peptide, and this peptide can be used as a specific ligand to introduce endothelial cell attachment onto the surfaces of polymer scaffold. In this study, biodegradable polymer surface immobilizing REDV peptide were prepared, and the specific attachment of endothelial cells on it was investigated as a preliminary study for tissue-engineered small-caliber blood vessels in a future application. We synthesized copolymer of ε-caprolactone and depsipeptide having reactive carboxylic acid side-chain groups (PGDCL), and REDV peptide was attached to the copolymer to give PGDCL-REDV. The attachment of human umbilical vein endothelial cells (HUVECs) were investigated for the blend polymer film prepared by mixing PGDCL and PGDCL-REDV. The obtained blend polymer films exhibited sequence- and cell-specific HUVECs attachment through REDV peptide recognition. This technique should be useful not only to obtain artificial blood vessels which induce endothelialization and but also to provide biodegradable scaffolds with specific ligands immobilized surfaces for tissue regeneration.


Assuntos
Peptídeos , Polímeros , Adesão Celular , Células Endoteliais da Veia Umbilical Humana , Humanos , Engenharia Tecidual
19.
Biomed Res Int ; 2020: 3074742, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32219130

RESUMO

Cutaneous squamous cell carcinoma (cSCC) is one of the most common skin cancers. In the treatment of cSCC, it is necessary to remove it completely, and reconstructive surgery, such as a skin graft or a local or free flap, will be required, depending on the size, with donor-site morbidity posing a burden to the patient. The high hydrostatic pressure (HHP) technique has been developed as a physical method of decellularizing various tissues. We previously reported that HHP at 200 MPa for 10 min could inactivate all cells in the giant congenital melanocytic nevus, and we have already started a clinical trial using this technique. In the present study, we explored the critical pressurization condition for annihilating cSCC cells in vitro and confirmed that this condition could also annihilate cSCC in vivo. We prepared 5 pressurization conditions in this study (150, 160, 170, 180, and 190 MPa for 10 min) and confirmed that cSCC cells were killed by pressurization at ≥160 MPa for 10 min. In the in vivo study, the cSCC cells inactivated by HHP at 200 MPa for 10 min were unable to proliferate after injection into the intradermal space of mice, and transplanted cSCC tissues that had been inactivated by HHP showed a decreased weight at 5 weeks after implantation. These results suggested that HHP at 200 MPa for 10 min was able to annihilate SCC, so HHP technology may be a novel treatment of skin cancer.


Assuntos
Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/terapia , Pressão Hidrostática , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Animais , Linhagem Celular , Proliferação de Células , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Pele/patologia
20.
Biomed Res Int ; 2020: 9478789, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32076621

RESUMO

High hydrostatic pressure (HHP) is a physical method for inactivating cells or tissues without using chemicals such as detergents. We previously reported that HHP at 200 MPa for 10 min was able to inactivate all cells in skin and giant congenital melanocytic nevus (GCMN) without damaging the extracellular matrix. We also reported that HHP at 150 MPa for 10 min was not sufficient to inactivate them completely, while HHP at 200 MPa for 10 min was able to inactivate them completely. We intend to apply HHP to treat malignant skin tumor as the next step; however, the conditions necessary to kill each kind of cell have not been explored. In this work, we have performed a detailed experimental study on the critical pressure and pressurization time using five kinds of human skin cells and skin tumor cells, including keratinocytes (HEKas), dermal fibroblasts (HDFas), adipose tissue-derived stem cells (ASCs), epidermal melanocytes (HEMa-LPs), and malignant melanoma cells (MMs), using pressures between 150 and 200 MPa. We pressurized cells at 150, 160, 170, 180, or 190 MPa for 1 s, 2 min, and 10 min and evaluated the cellular activity using live/dead staining and proliferation assays. The proliferation assay revealed that HEKas were inactivated at a pressure higher than 150 MPa and a time period longer than 2 min, HDFas and MMs were inactivated at a pressure higher than 160 MPa and for 10 min, and ASCs and HEMa-LPs were inactivated at a pressure higher than 150 MPa and for 10 min. However, some HEMa-LPs were observed alive after HHP at 170 MPa for 10 min, so we concluded that HHP at a pressure higher than 180 MPa for 10 min was able to inactivate five kinds of cells completely.


Assuntos
Pressão Hidrostática , Neoplasias Cutâneas/patologia , Pele/patologia , Anexina A5 , Apoptose , Proliferação de Células , Epiderme/patologia , Matriz Extracelular/patologia , Fibroblastos/patologia , Humanos , Queratinócitos/patologia , Melanócitos , Nevo Pigmentado
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA