Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Pharm ; 16(2): 689-700, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30601012

RESUMEN

The excessive increase of intracellular reactive oxygen species (ROS) makes tumor cells usually in the state of oxidative stress. Although tumor cells can adapt to this state to a certain extent by upregulating antioxidant systems, the further ROS insults disrupt the transient intracellular redox balance, eventually leading to apoptosis and necrosis. Therefore, increasing the intracellular ROS level can effectively amplify the oxidative stress and induce apoptosis, which can be employed as a strategy for tumor treatment. Herein, a unique pH-responsive ROS inducing micellar system was reported in this study to specifically amplify the ROS signal in tumor cells. This micellar system was constructed by a new amphiphilic polymer, PIAThydCA, composed of poly(itaconic acid) (PIA) as the hydrophilic backbone, d-α-tocopherol polyethylene glycol 1000 succinate (TPGS) as the hydrophobic side chain, and cinnamaldehyde (CA) as the ROS-generating agent, which were linked to PIA by the pH-sensitive hydrazone bond. PIAThydCA micelles could be degraded in the intracellular acidic environment through the hydrolysis of hydrazone bond and release CA. CA and TPGS could amplify oxidative stress cooperatively to kill MCF-7 human breast cells preferentially through the mitochondrial apoptosis pathway. Therefore, we anticipate that the PIAThydCA micelles could exert great potential in anticancer therapy.


Asunto(s)
Estrés Oxidativo/efectos de los fármacos , Polímeros/química , Acroleína/análogos & derivados , Acroleína/química , Antineoplásicos/química , Antineoplásicos/farmacología , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Micelas , Oxidación-Reducción/efectos de los fármacos , Polietilenglicoles/química , Polímeros/farmacología , Especies Reactivas de Oxígeno/metabolismo
2.
Colloids Surf B Biointerfaces ; 244: 114181, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39216443

RESUMEN

Implant-associated infections impose great burden on patient health and public healthcare. Antimicrobial peptides and metal ions are generally incorporated onto implant surface to deter bacteria colonization. However, it is still challenging to efficiently prevent postoperative infections at non-cytotoxic dosages. Herein, a scaffold based on porous titanium coated with a mussel-inspired dual-diameter TiO2 nanotubes is developed for loading dual drugs of LL37 peptide and Zn2+ with different sizes and characteristics. Benefiting from in-situ formed polydopamine layer and dual-diameter nanotubular structure, the scaffold provides an efficient platform for controllable drugs elution: accelerated release under acidic condition and sustained release for up to 28 days under neutral/alkalescent circumstances. Such combination of dual drugs simultaneously enhanced antibacterial efficacy and osteogenesis. In antibacterial test, LL37 peptide serving as bacteria membrane puncture agent, and Zn2+ acting as ROS generator, cooperatively destroyed bacterial membrane integrity and subsequently damaged bacterial DNA, endowing dual-drug loaded scaffold with remarkable bactericidal efficiency of > 92 % in vitro and > 99 % in vivo. Noteworthily, dual-drug loaded scaffold promoted bone-implant osteointegration under infectious microenvironment, overmatching single-drug load ones. It provides a promising strategy on surface modification of implant for infected bone defect repairing.


Asunto(s)
Antibacterianos , Bivalvos , Titanio , Zinc , Titanio/química , Titanio/farmacología , Zinc/química , Zinc/farmacología , Porosidad , Animales , Bivalvos/química , Antibacterianos/farmacología , Antibacterianos/química , Catelicidinas , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/farmacología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Pruebas de Sensibilidad Microbiana , Propiedades de Superficie , Osteogénesis/efectos de los fármacos , Liberación de Fármacos , Staphylococcus aureus/efectos de los fármacos , Tamaño de la Partícula , Indoles , Polímeros
3.
Int Dent J ; 74(4): 836-846, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38369441

RESUMEN

OBJECTIVE: Pulp regeneration with bioactive dentin-pulp complex has been a research hotspot in recent years. Stem cell therapy provided an interest strategy to regenerate the dental-pulp complex. Hence, this study aimed to evaluate the effects of photosensitive gelatin methacrylate (GelMA) hydrogel encapsulating dental pulp stem cells (DPSCs) and silver nanoparticles (AgNPs) for dental pulp regeneration in vitro. METHODS: First, the AgNPs@GelMA hydrogels were prepared by lithium phenyl-2,4,6-trimethyl-benzoyl phosphinate (LAP) initiation via blue-light emitting diode light. The physical and chemical properties of AgNPs@GelMA hydrogels were comprehensively analysed via scanning electron microscopy (SEM), and mechanical characterisation, such as swelling ability, degradation properties, and AgNP release profile. Then, AgNPs@GelMA hydrogels encapsulated DPSCs were used to establish an AgNPs@GelMA biomimetic complex, further analysing its biocompatibility, antibacterial properties, and angiogenic capacity in vitro. RESULTS: The results indicated that GelMA hydrogels demontrated optimal characteristics with a monomer:LAP ratio of 16:1. The physico-chemical properties of AgNPs@GelMA hydrogels did not change significantly after loading with AgNPs. There was no significant difference in AgNP release rate amongst different concentrations of AgNPs@GelMA hydrogels. Fifty to 200 µg/mL AgNPs@GelMA hydrogels could disperse E faecalis biofilm and reduce its metabolic activity . Furthermore, cell proliferation was arrested in 100 and 200 µg/mL AgNPs@GelMA hydrogels. The inhibition of 50 µg/mL AgNPs@GelMA hydrogels on E faecalis biofilm was above 50%, and the cell viability of the hydrogels was higher than 90%. The angiogenesis assay indicated that AgNPs@GelMA hydrogels encapsulating DPSCs could induce the formation of capillary-like structures and express angiogenic markers CD31, vascular endothelial growth factor , and von willebrand factor (vWF) in vitro. CONCLUSIONS: Results of this study indicate that 50 µg/mL AgNPs@GelMA hydrogels encapsulating DPSCs had significant antibacterial properties and angiogenic capacity, which could provide a significant experimental basis for the regeneration of the dentin-pulp complex.


Asunto(s)
Pulpa Dental , Gelatina , Hidrogeles , Nanopartículas del Metal , Regeneración , Plata , Células Madre , Pulpa Dental/citología , Pulpa Dental/efectos de los fármacos , Humanos , Hidrogeles/química , Nanopartículas del Metal/uso terapéutico , Células Madre/efectos de los fármacos , Regeneración/efectos de los fármacos , Gelatina/química , Metacrilatos/química , Antibacterianos/farmacología , Microscopía Electrónica de Rastreo , Proliferación Celular/efectos de los fármacos
4.
ACS Appl Mater Interfaces ; 15(33): 39847-39863, 2023 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-37578471

RESUMEN

The wet environment of water or tissue in bleeding wounds poses significant challenges to the adhesion performance of existing hemostatic adhesives. An intelligent composite adhesive prepared by doping starch-based silicate micro-nanograded porous particles (MBC@CMS) with dopamine-hyperbranched polymers (HPD, 7800 Mw) synthesized by the Michael addition reaction could be triggered by water to form a glue (MBC@CMS-HPD). The results indicated that MBC@CMS-HPD could still have adhesion properties under running water washing and water immersion and could effectively seal the water outlet. The results of the glue-forming mechanism showed that MBC@CMS-HPD had better wettability than water, which could eliminate water molecules at the wet adhesive surface. When contacted with water, the agglomeration of the HPD hydrophobic chain increases the exposure of the catechol group, and the relative atomic mass of the N element on the surface increases from 2.8 to 4.8%. The adhesion of MBC@CMS-HPD was enhanced and stable. MBC@CMS-HPD showed significant hemostasis effects in five injury bleeding models of Sprague-Dawley (SD) rats and New Zealand rabbits. Especially in the fatal femoral artery bleeding model of New Zealand rabbits, MBC@CMS-HPD reduced the amount of bleeding by 75% and shortened the bleeding time by 78% compared with the a-cyanoacrylate adhesives. The results of the coagulation mechanism showed that compared with HPD, MBC@CMS-HPD could activate both endogenous and exogenous coagulation pathways. Among them, after contact with blood, HPD formed a gel to close the blood outlet, and MBC@CMS entered the wound to activate the internal and external coagulation pathways. In addition, HPD and MBC@CMS had good histocompatibility and degradability, which has the potential to be applied to different wounds.


Asunto(s)
Hemostáticos , Adhesivos Tisulares , Ratas , Animales , Conejos , Hemostáticos/farmacología , Hemostáticos/química , Adhesivos/farmacología , Dopamina/farmacología , Dopamina/química , Porosidad , Agua/química , Ratas Sprague-Dawley , Hemostasis , Hemorragia/terapia , Adhesivos Tisulares/química
5.
Toxicology ; 466: 153066, 2022 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-34919984

RESUMEN

In order to overcome the weakness of conventional approaches for cell culture, and provide cells with more in vivo-like microenvironment for studying hepatotoxicity of drugs, "multiple-in-one" strategy was adopted to fabricate a 3D scaffold of silk fibroin/hydroxyapatite/poly lacticco-glycolic acid (SF/HA/PLGA), where HepG2 cells were cultivated and the toxicity of drugs to the cells was investigated. The prepared 3D scaffold proves to bear proper porosity, excellent mechanical property, steady pH environment and good biocompatibility for cell culture. Furthermore, the validity of the developed 3D-SF/HA/PLGA-scaffold based platform was verified by probing the toxicity of a known drug-induced liver injury (DILI) concern acetaminophen (APAP) to HepG2 cells. Eventually, an application of the platform to dioscin (a medicinal plant extract) reveals the hepatotoxicity of dioscin, which involves the inhibition of the expression of CYP3A4 mRNA in the cells. The developed 3D-SF/HA/PLGA-scaffold platform may become a universal avenue for safety evaluation of drugs.


Asunto(s)
Acetaminofén/toxicidad , Técnicas de Cultivo de Célula/métodos , Resinas Compuestas/química , Evaluación Preclínica de Medicamentos/métodos , Andamios del Tejido/química , Diferenciación Celular , Proliferación Celular , Enfermedad Hepática Inducida por Sustancias y Drogas , Sistema Enzimático del Citocromo P-450/metabolismo , Diosgenina/análogos & derivados , Diosgenina/toxicidad , Glucosa/metabolismo , Células Hep G2 , Humanos , Preparaciones Farmacéuticas
6.
J Colloid Interface Sci ; 607(Pt 2): 1239-1252, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34583031

RESUMEN

Tissue adhesives have received much attention for their effectiveness in sealing wounds or incisions in clinical surgery, especially in minimally invasive surgery. To meet the safe and smart wound management requirements, ideal tissue adhesives are expected to have high biocompatibility, and be able to accelerate wound closing and healing, and monitor wound healing process. However, few adhesives fit all of the above descriptions. It has been demonstrated that inorganic nanoparticles can directly glue biological tissue based on nano-bridging effect. In this study, self-luminescence porous silicon (LPSi) particles were prepared with degradable and biocompatible properties. In addition, the self-luminescence property of LPSi particles was discovered by In Vivo Imaging System (IVIS) for the first time, which can avoid the limitations of photoluminescence imaging. Due to the oxidation and degradation reaction, LPSi particles not only can be degraded completely in several days, but also showed satisfactory biocompatibility. And their degradation product could promote tube formation of HUVECs. Moreover, owing to the high specific surface area and the outer oxide layer of LPSi particles, LPSi tissue adhesive exhibited strong adhesive strength to pig livers. Furthermore, this adhesive closed wound rapidly, promoted angiogenesis and epidermal regeneration, and facilitated wound healing in a mouse skin incision model. Importantly, the wound healing ratio can be monitored by measuring the self-luminescence intensity of LPSi particles in the wound site. This study reveals that LPSi particles could be employed as a safe and smart wound management tissue adhesive for wound closure, as well as accelerating and monitoring wound healing.


Asunto(s)
Adhesivos Tisulares , Animales , Luminiscencia , Ratones , Porosidad , Silicio , Porcinos , Cicatrización de Heridas
7.
Biomater Adv ; 133: 112651, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35034817

RESUMEN

Porous titanium implants were popularly fabricated to promote bone formation. A desirable porous scaffold was recommended to be with porosity of >60% or/and pore size of >300 µm for better osteointegration. However, whether the pore size and porosity could be randomly selected within the recommended values? And what is the correlation between pore size and porosity for accelerating osteointegration? In this study, porous titanium with cubic cell structure was produced by selective laser melting. The designed porosities of scaffolds with 700-µm pore size were 40%, 70% and 90%; and the pore sizes of scaffolds with 70% porosity were 400, 700 and 900 µm. The in vitro osteogenic potential and in vivo bone formation were investigated. Results showed that porosity and pore size could be tuned by altering strut size, which was further directly responsible for mechanical properties. Besides, pore size and porosity synergistically contributed to osteogenic activity in vitro and new bone formation in vivo. In regard to pore sizes herein, the optimized one for better osteogenic response and bone forming ability was ~600-700 µm (p70). Too smaller or too larger pore size might more or less hinder cellular behaviors and bone regeneration, even if both pore size (300-900 µm) and porosity (70%) were within the recommended value range. At a constant pore size (~600-700 µm), p70 and p90 with higher porosity was more conductive to biological effects, compared with p40. As a result, pore-size variation revealed more significant influence on osteogenesis, compared with variation of porosity within recommended values. However, the applicable porosity within recommended values should be designed with the consideration of specific load-bearing conditions. This study helps to provide guidance for designing porous scaffolds with appropriate mechanical strengths and effective bone-forming ability, so as to develop better custom-made bone substitutes.


Asunto(s)
Sustitutos de Huesos , Osteogénesis , Sustitutos de Huesos/química , Porosidad , Impresión Tridimensional , Titanio/química
9.
Int J Biol Macromol ; 193(Pt B): 2021-2028, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34767883

RESUMEN

Mg alloys are increasingly being investigated as a versatile and economical alternative for developing bone repair implants because of their high mechanical strength, wide availability, adjustable structure and properties. In this study, magnesium alloy WE43 is coated on both sides with gelatin nanosphere/chitosan (GNs/CTS), a coating enhanced by incorporating simvastatin (SIM). SIM-loaded GNs/CTS coated magnesium alloy can promote the osteogenic differentiation of bone mesenchymal stem cells (BMSCs). BMSCs and human umbilical vein endothelial cells (HUVECs) are co-cultured through transwell systems. The release of SIM from the coating is found to increase the secretion of chemokine and angiogenic factors from BMSCs, which promote the migration and tube formation of HUVECs, respectively. Bone morphogenetic protein secreted by HUVECs is seen to increase by the release of SIM from the coating, promoting the osteogenic differentiation of BMSCs. The secretion of chemokines from HUVECs promote the migration of BMSCs. The coated magnesium alloy substrate loaded with SIM is found to regulate the osteogenic differentiation of BMSCs. The study of the paracrine interaction between BMSCs and HUVECs proves that the applied coating promotes both osteogenic differentiation and vascularization, thus demonstrating a new approach for the design of bone repair materials based on magnesium alloys.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Quitosano/farmacología , Gelatina/farmacología , Magnesio/química , Nanosferas/química , Osteogénesis/efectos de los fármacos , Simvastatina/farmacología , Aleaciones/farmacología , Inductores de la Angiogénesis/farmacología , Huesos/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo/métodos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos
10.
Int J Biol Macromol ; 179: 507-518, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33711370

RESUMEN

The powdered hemostatic particles have broad application prospects in large open wounds, internal organ injuries and penetrating injuries of the body. In this study, nanoscale mescoporous and macroporous silica (MMSN), nanoscale mescoporous and macroporous bioactive glass (MBG), micron-scale cross-linked corn starch porous microspheres (CMS), MMSN@CMS and MBG@CMS starch-based nano-microporous particles were synthesized and their hemostatic effect and hemostatic mechanism were studied. The results showed that comparted with the single particle of CMS, the combination particles MBG@CMS and MMSN@CMS significantly increased the water absorption rate, activated both internal and external coagulation pathways, significantly shortened CBT, as well as the improved hemostatic effects in vitro. The immediately released Ca2+ from MBG@CMS in the blood to participate in the coagulation pathway, and MMSN@CMS activated platelets by concentrating blood coagulation factors, might be the main hemostatic mechanisms for the starch-based nano-microporous particles. Furthermore, the hemostatic efficacy of particles, both in the model of tail-amputation and liver injury in SD rats, showed the starch-based nano-microporous particles, especial MBG@CMS, could significantly reduce the weight of blood loss and shorten the bleeding time. Our research work stated that the starch-based nano-microporous particles MBG@CMS might be a hemostasis biomaterial with the potential applications for the emergency bleeding.


Asunto(s)
Materiales Biocompatibles/química , Coagulación Sanguínea , Hemostáticos/química , Nanopartículas/química , Almidón/química , Animales , Línea Celular , Ratones , Ratas , Ratas Sprague-Dawley
11.
Cell Prolif ; 54(2): e12969, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33332682

RESUMEN

OBJECTIVES: Various factors could interfere the biological performance of DPSCs during post-thawed process. Yet, little has been known about optimization of the recovery medium for DPSCs. Thus, our study aimed to explore the effects of adding recombinant bFGF on DPSCs after 3-month cryopreservation as well as the underlying mechanisms. MATERIALS AND METHODS: DPSCs were extracted from impacted third molars and purified by MACS. The properties of CD146+ DPSCs (P3) were identified by CCK-8 and flow cytometry. After cryopreservation for 3 months, recovered DPSCs (P4) were immediately supplied with a series of bFGF and analysed cellular proliferation by CCK-8. Then, the optimal dosage of bFGF was determined to further identify apoptosis and TRPC1 channel through Western blot. The succeeding passage (P5) from bFGF pre-treated DPSCs was cultivated in bFGF-free culture medium, cellular proliferation and stemness were verified, and pluripotency was analysed by neurogenic, osteogenic and adipogenic differentiation. RESULTS: It is found that adding 20 ng/mL bFGF in culture medium could significantly promote the proliferation of freshly thawed DPSCs (P4) through suppressing apoptosis, activating ERK pathway and up-regulating TRPC1. Such proliferative superiority could be inherited to the succeeding passage (P5) from bFGF pre-stimulated DPSCs, meanwhile, stemness and pluripotency have not been compromised. CONCLUSIONS: This study illustrated a safe and feasible cell culture technique to rapidly amplify post-thawed DPSCs with robust regenerative potency, which brightening the future of stem cells banking and tissue engineering.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Pulpa Dental/metabolismo , Factor 2 de Crecimiento de Fibroblastos/farmacología , Adipogénesis/efectos de los fármacos , Apoptosis/efectos de los fármacos , Antígeno CD146/metabolismo , Diferenciación Celular/efectos de los fármacos , Criopreservación , Medios de Cultivo/química , Pulpa Dental/citología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Homeótica Nanog/metabolismo , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células Madre/citología , Células Madre/metabolismo , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPC/metabolismo , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos
12.
Bioact Mater ; 6(3): 638-654, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33005828

RESUMEN

Due to the limitations in autogenous nerve grafting or Schwann cell transplantation, large gap peripheral nerve injuries require a bridging strategy supported by nerve conduit. Cell based therapies provide a novel treatment for peripheral nerve injuries. In this study, we first experimented an optimal scaffold material synthesis protocol, from where we selected the 10% GFD formula (10% GelMA hydrogel, recombinant human basic fibroblast growth factor and dental pulp stem cells (DPSCs)) to fill a cellulose/soy protein isolate composite membrane (CSM) tube to construct a third generation of nerve regeneration conduit, CSM-GFD. Then this CSM-GFD conduit was applied to repair a 15-mm long defect of sciatic nerve in a rat model. After 12 week post implant surgery, at histologic level, we found CSM-GFD conduit could regenerate nerve tissue like neuron and Schwann like nerve cells and myelinated nerve fibers. At physical level, CSM-GFD achieved functional recovery assessed by a sciatic functional index study. In both levels, CSM-GFD performed like what gold standard, the nerve autograft, could do. Further, we unveiled that almost all newly formed nerve tissue at defect site was originated from the direct differentiation of exogeneous DPSCs in CSM-GFD. In conclusion, we claimed that this third-generation nerve regeneration conduit, CSM-GFD, could be a promising tissue engineering approach to replace the conventional nerve autograft to treat the large gap defect in peripheral nerve injuries.

13.
Artículo en Inglés | MEDLINE | ID: mdl-32903416

RESUMEN

Norspermidine (Nspd) is a kind of polyamine molecule, which is common in eukaryotes and prokaryotes. It has been reported as a potential anti-biofilms agent of bacteria, but its anti-fungal effect remains unclear. Candida albicans (C. albicans) is a common opportunistic pathogen in oral cavity of human beings. C. albicans biofilm is often seen in dental caries. In this work, we aimed to study the effect of Nspd on mature Candida albicans biofilms and to investigate how Nspd would influence human dental pulp stem cells (DPSCs). Our biofilm assays indicated that 111.7 and 55.9 mM Nspd dispersed 48 h mature fungal biofilms and showed significant fungicidal effect. 27.9 and 14.0 mM Nspd showed moderate fungicidal effect. Live/dead staining echoed the fungicidal effect. 111.7-14.0 mM Nspd showed a dose- inhibitory effect on mature fungal biofilm, where 14.0 mM Nspd reduced the metabolic activity by half compared with blank control. Moreover, we demonstrated that 111.7-27.9 mM Nspd restrained the production of hyphae form of C. albicans via SEM. Low dose Nspd (27.9 and 14.0 mM) could significantly reduce virulence related gene expression in C. albicans biofilms. MTT assay displayed a dose effect relation between 2.5-0.08 mM Nspd and DPSCs viability, where 0.63 mM Nspd reduced the viable level of DPSCs to 75% compared with blank control. Live/dead staining of DPSCs did not show distinctive difference between 0.63 mM Nspd and blank control. Vascular differentiation assay showed capillary-like structure of inducted DPSCs culture with and without 0.63 mM Nspd suggesting that it did not significantly affect angiogenic differentiation of DPSCs. Nspd can penetrate remaining dentin at low level, which is confirmed by an in vitro caries model. In conclusion, our study indicated high dosage Nspd (111.7 and 55.9 mM) could effectively disrupt and kill mature fungal biofilms. Low dosage (27.9 and 14.0 mM) showed mild anti-fungal effect on mature C. albicans biofilms. Human DPSCs were tolerate to 0.08-0.63 mM Nspd, where viability was over 75%. 0.63 mM Nspd did not affect the proliferation and angiogenetic differentiation of DPSCs.

14.
Front Cell Dev Biol ; 8: 625151, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33344464

RESUMEN

Transplantation of undifferentiated dental pulp stem cells (DPSCs) may suffer from tumorigenesis. Neuronal differentiated DPSCs (d-DPSCs) have emerged as an ideal source to treat central nervous system (CNS) disorders. Moreover, different components of culture medium functioned on the characteristics of d-DPSCs in vitro. In this study, d-DPSCs were cultured in three types of medium: Neurobasal®®-A medium supplemented with 2% B27 (the 2% B27 NM group), Neurobasal® -A medium supplemented with 2% B27 and 5% FBS (the 2% B27 + 5% FBS NM group), and α-MEM containing 10% FBS (the 10% FBS α-MEM group). We found that d-DPSCs in the 2% B27 + 5% FBS NM group had lower proliferation and reduced expression of transient receptor potential canonical 1 (TRPC1) and CD146, whereas up-regulated Nestin and microtubule-associated protein-2 (MAP-2). Notably, d-DPSCs in the 10% FBS α-MEM group possessed high proliferative capacity, decreased expression of neuron-like markers and partially restored stemness. It was demonstrated that d-DPSCs cultured in the 2% B27 + 5% FBS NM could maintain their neuron-like characteristics. Besides, d-DPSCs cultivated in the 10% FBS α-MEM could partially recover their stem cells properties, indicating that neural differentiation of DPSCs was reversible and could open novel avenues for exploring the pluripotency of DPSCs.

15.
ACS Omega ; 5(26): 16064-16075, 2020 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-32656428

RESUMEN

Acute spinal cord injury (SCI) induces severe neuroinflammation, which increases intermediary filaments and neurodegeneration. Previous studies have shown that a basic fibroblast growth factor (bFGF) and dental pulp stem cells (DPSCs) contribute to a protective effect on injured neuronal cells, but the mechanism of SCI repair is still unclear. In this study, in situ heparin (HeP) hydrogel injection containing bFGF and DPSCs (HeP-bFGF-DPSCs), as well as in vitro studies of bFGF and DPSCs, proved an effective control over inflammation. The in vivo application of HeP-bFGF-DPSCs regulated inflammatory reactions and accelerated the nerve regeneration through microtubule stabilization and tissue vasculature. Our mechanistic investigation also showed that bFGF-DPSCs treatment inhibited microglia/macrophage proliferation and activation. Furthermore, HeP-bFGF-DPSCs prevented microglia/macrophage activation and reduced proinflammatory cytokine release. In this paper, we discovered that bFGF and DPSCs worked together to attenuate tissue inflammation of the injured spinal cord, resulting in a superior nerve repair. Our results indicated that a thermosensitive hydrogel delivering bFGF and DPSCs could serve as a promising treatment option for spinal cord injuries.

16.
Int J Nanomedicine ; 14: 5713-5728, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31413571

RESUMEN

Purpose: The levels of reactive oxygen species (ROS) in tumor cells are much higher than that in normal cells, and rise rapidly under the influence of exogenous or endogenous inducing factors, eventually leading to the apoptosis of tumor cells. Therefore, this study prepared a dual pH/reducing-responsive poly (N-isopropylacrylamide-co-Cinnamaldehyde-co-D-α-tocopheryl polyethylene glycol 1000 succinate, PssNCT) nanogels, which employed two exogenous ROS inducers, cinnamaldehyde (CA) and D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS), to selectively induce apoptosis by regulating ROS levels in tumor cells. Methods: The PssNCT nanogels were prepared by the free radical precipitation polymerization under the crosslink between pH-sensitive hydrazone and reducing-sensitive disulfide bonds, followed by the physicochemical and morphological characteristics investigations. Plasma stability, dual pH/reducing responsive degradation and in vitro release were also assessed. In cell experiments, cytotoxicity in different cells were first detected. The intracellular ROS levels and mitochondrial functions of tumor cells were then evaluated. Moreover, the apoptosis and western-blot assays were employed to verify the association between ROS levels elevation and apoptosis in tumor cells. Results: The nanogels exhibited a round-like hollow structure with the diameter smaller than 200nm. The nanogels were stable in plasma, while showed rapid degradation in acidic and reducing environments, thus achieving significant release of CA and TPGS in these media. Furthermore, the sufficient amplification of ROS signals was induced by the synergistically function of CA and TPGS on mitochondria, which resulted in the opening of the mitochondrial apoptotic pathway and enhanced cytotoxicity on MCF-7 cells. However, nanogels barely affected L929 cells owing to their lower intracellular ROS basal levels. Conclusion: The specific ROS regulation method achieved by these nanogels could be explored to selectively kill tumor cells according to the difference of ROS signals in different kinds of cells.


Asunto(s)
Apoptosis , Espacio Intracelular/química , Polietilenglicoles/farmacología , Polietileneimina/farmacología , Especies Reactivas de Oxígeno/metabolismo , Acroleína/análogos & derivados , Acroleína/farmacología , Animales , Apoptosis/efectos de los fármacos , Doxorrubicina/farmacología , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanogeles , Vitamina E/síntesis química , Vitamina E/química
17.
Stem Cells Int ; 2018: 2398521, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29765407

RESUMEN

Spinal cord injury (SCI) is one of serious traumatic diseases of the central nervous system and has no effective treatment because of its complicated pathophysiology. Tissue engineering strategy which contains scaffolds, cells, and growth factors can provide a promising treatment for SCI. Hydrogel that has 3D network structure and biomimetic microenvironment can support cellular growth and embed biological macromolecules for sustaining release. Dental pulp stem cells (DPSCs), derived from cranial neural crest, possess mesenchymal stem cell (MSC) characteristics and have an ability to provide neuroprotective and neurotrophic properties for SCI treatment. Basic fibroblast growth factor (bFGF) is able to promote cell survival and proliferation and also has beneficial effect on neural regeneration and functional recovery after SCI. Herein, a thermosensitive heparin-poloxamer (HP) hydrogel containing DPSCs and bFGF was prepared, and the effects of HP-bFGF-DPSCs on neuron restoration after SCI were evaluated by functional recovery tests, western blotting, magnetic resonance imaging (MRI), histology evaluation, and immunohistochemistry. The results suggested that transplanted HP hydrogel containing DPSCs and bFGF had a significant impact on spinal cord repair and regeneration and may provide a promising strategy for neuron repair, functional recovery, and tissue regeneration after SCI.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA