Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Plant J ; 111(2): 608-616, 2022 07.
Article in English | MEDLINE | ID: mdl-35510429

ABSTRACT

Though Medicago truncatula Tnt1 mutants are widely used by researchers in the legume community, they are mainly used for reverse genetics because of the availability of the BLAST-searchable large-scale flanking sequence tags database. However, these mutants should have also been used extensively for forward genetic screens, an effort that has been hindered due to the lack of a compatible genetic crossing partner for the M. truncatula genotype R108, from which Tnt1 mutants were generated. In this study, we selected three Medicago HapMap lines (HM017, HM018 and HM022) and performed reciprocal genetic crosses with R108. After phenotypic analyses in F1 and F2 progenies, HM017 was identified as a compatible crossing partner with R108. By comparing the assembled genomic sequences of HM017 and R108, we developed and confirmed 318 Indel markers evenly distributed across the eight chromosomes of the M. truncatula genome. To validate the effectiveness of these markers, by employing the map-based cloning approach, we cloned the causative gene in the dwarf mutant crs isolated from the Tnt1 mutant population, identifying it as gibberellin 3-ß-dioxygenase 1, using some of the confirmed Indel markers. The primer sequences and the size difference of each marker were made available for users in the web-based database. The identification of the crossing partner for R108 and the generation of Indel markers will enhance the forward genetics and the overall usage of the Tnt1 mutants.


Subject(s)
Medicago truncatula , Databases, Nucleic Acid , Genes, Plant , Genetic Testing , Medicago truncatula/genetics , Mutagenesis, Insertional
2.
Colloids Surf B Biointerfaces ; 238: 113889, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38574404

ABSTRACT

A novel core-shell with a tetradecyl dimethyl benzyl ammonium chloride-modified montmorillonite (TDMBA/MMT) interlayer silk fibroin (SF)/poly(lactic acid) (PLLA) nanofibrous membrane was fabricated using a simple conventional electrospinning method. Scanning electron microscopy and pore size analyses revealed that this core-shell with TDMBA/MMT interlayer maintained its nanofibrous morphology and larger pore structure more successfully than SF/PLLA nanofibrous membranes after treatment with 75% ethanol vapor. Transmission electron microscopy and energy-dispersive X-ray spectroscopy analyses testified that the SF/PLLA-TDMBA/MMT nanofibers exhibited a core-shell with an interlayer structure, with SF/PLLA in the core-shell layer and TDMBA/MMT in the interlayer. The formation of a core-shell with interlayer nanofibers was primarily attributed to the uniform dispersion of TDMBA/MMT nanosheets in a solution owing to its exfoliation using hexafluoroisopropanol and then preparing a stable spinning solution similar to an emulsion. Compared to SF/PLLA nanofibrous membranes, the core-shell structure with TDMBA/MMT interlayers of SF/PLLA nanofibrous membranes exhibited enhanced hydrophilicity, thermal stability, mechanical properties as well as improved and long-lasting antimicrobial performance against Escherichia coli and Staphylococcus aureus without cytotoxicity.


Subject(s)
Bentonite , Escherichia coli , Nanofibers , Staphylococcus aureus , Bentonite/chemistry , Bentonite/pharmacology , Nanofibers/chemistry , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacology , Fibroins/chemistry , Fibroins/pharmacology , Polyesters/chemistry , Polyesters/pharmacology , Membranes, Artificial , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Mice , Animals
3.
Int J Mol Sci ; 13(2): 2036-2047, 2012.
Article in English | MEDLINE | ID: mdl-22408436

ABSTRACT

In order to improve the water-resistant ability of silk fibroin (SF) and SF/P(LLA-CL) blended nanofibrous scaffolds for tissue engineering applications, 75% (v/v) ethanol vapor was used to post-treat electrospun nanofibers. SEM indicated that the treated SF and SF/P(LLA-CL) nanofibrous scaffolds maintained a nanofibrous structure and possessed good water-resistant ability. Characterization of (13)C CP-MAS NMR clarified that 75% (v/v) ethanol vapor could induce SF conformation from random coil or α-helix to ß-sheet. Although the water contact showed that treated SF/P(LLA-CL) blended nanofibrous scaffolds were hydrophobic, the water uptake demonstrated that their hydrophilicity was greatly superior to those of pure P(LLA-CL) nanofibrous scaffolds. Furthermore, the treated SF/P(LLA-CL) nanofibrous scaffolds, both in dry state and wet state, could retain good mechanical properties. Therefore, 75% (v/v) ethanol vapor treatment might be an ideal method to treat SF and SF/P(LLA-CL) nanofibrous scaffolds for biomedical applications.


Subject(s)
Ethanol/pharmacology , Fibroins/drug effects , Polyesters/chemistry , Tissue Scaffolds/chemistry , Cell Culture Techniques/instrumentation , Ethanol/chemistry , Fibroins/chemistry , Hydrophobic and Hydrophilic Interactions/drug effects , Nanofibers/chemistry , Silk/chemistry , Silk/drug effects , Surface Properties/drug effects , Tensile Strength/drug effects , Tissue Engineering , Volatilization , Wettability/drug effects
4.
Front Bioeng Biotechnol ; 10: 1114034, 2022.
Article in English | MEDLINE | ID: mdl-36698642

ABSTRACT

The cells and tissue in the human body are orderly and directionally arranged, and constructing an ideal biomimetic extracellular matrix is still a major problem to be solved in tissue engineering. In the field of the bioresorbable vascular grafts, the long-term functional prognosis requires that cells first migrate and grow along the physiological arrangement direction of the vessel itself. Moreover, the graft is required to promote the formation of neointima and the development of the vessel walls while ensuring that the whole repair process does not form a thrombus. In this study, poly (l-lactide-co-ε-caprolactone) (PLCL) shell layers and polyethylene oxide (PEO) core layers with different microstructures and loaded with sodium tanshinone IIA sulfonate (STS) were prepared by coaxial electrospinning. The mechanical properties proved that the fiber membranes had good mechanical support, higher than that of the human aorta, as well as great suture retention strengths. The hydrophilicity of the oriented-fiber membranes was greatly improved compared with that of the random-fiber membranes. Furthermore, we investigated the biocompatibility and hemocompatibility of different functional fiber membranes, and the results showed that the oriented-fiber membranes containing sodium tanshinone IIA sulfonate had an excellent antiplatelet adhesion effect compared to other fiber membranes. Cytological analysis confirmed that the functional fiber membranes were non-cytotoxic and had significant cell proliferation capacities. The oriented-fiber membranes induced cell growth along the orientation direction. Degradation tests showed that the pH variation range had little change, the material mass was gradually reduced, and the fiber morphology was slowly destroyed. Thus, results indicated the degradation rate of the oriented-fiber graft likely is suitable for the process of new tissue regeneration, while the random-fiber graft with a low degradation rate may cause the material to reside in the tissue for too long, which would impede new tissue reconstitution. In summary, the oriented-functional-fiber membranes possessing core-shell structures with sodium tanshinone IIA sulfonate/polyethylene oxide loading could be used as tissue engineering materials for applications such as vascular grafts with good prospects, and their clinical application potential will be further explored in future research.

5.
Neurochem Res ; 36(10): 1896-902, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21611833

ABSTRACT

Substance P (SP) is a neuropeptide that plays an important role in inflammation, respiration, pain, aggression, anxiety, and learning and memory mainly through its high affinity neurokinin 1 receptor (NK1R). The marginal division (MrD) is a pan-shaped subdivision in the caudomedial margin of the neostriatum in the mammalian brain and is known to be involved in learning and memory. We studied the expression of SP, NK1R and NK1R mRNA in the rat striatum by immunohistochemistry, immunofluorescence and in situ hybridization, and found that the levels of SP, NK1R protein and NK1R mRNA were high in the cell bodies, fibers and terminals of neurons in the neostriatum, especially in the MrD. Knocking down NK1R activity in the MrD by using an antisense oligonucleotide against NK1R mRNA inhibited learning and memory in a Y-maze behavioral test. Our results show that NK1R mediates the role of SP in the MrD in learning and memory.


Subject(s)
Learning/physiology , Memory/physiology , Neostriatum/anatomy & histology , Neostriatum/physiology , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Animals , Behavior, Animal/physiology , Male , Rats , Receptors, Neurokinin-1/genetics
6.
Int J Mol Sci ; 12(4): 2187-99, 2011.
Article in English | MEDLINE | ID: mdl-21731435

ABSTRACT

The native extracellular matrix (ECM) is composed of a cross-linked porous network of multifibril collagens and glycosaminoglycans. Nanofibrous scaffolds of silk fibroin (SF) and hydroxybutyl chitosan (HBC) blends were fabricated using 1,1,1,3,3,3-hexafluoro-2-propanol (HFIP) and trifluoroacetic acid (TFA) as solvents to biomimic the native ECM via electrospinning. Scanning electronic microscope (SEM) showed that relatively uniform nanofibers could be obtained when 12% SF was blended with 6% HBC at the weight ratio of 50:50. Meanwhile, the average nanofibrous diameter increased when the content of HBC in SF/HBC blends was raised from 20% to 100%. Fourier transform infrared spectra (FTIR) and (13)C nuclear magnetic resonance (NMR) showed SF and HBC molecules existed in hydrogen bonding interactions but HBC did not induce conformation of SF transforming from random coil form to ß-sheet structure. X-ray diffraction (XRD) confirmed the different structure of SF/HBC blended nanofibers from both SF and HBC. Thermogravimetry-Differential thermogravimetry (TG-DTG) results demonstrated that the thermal stability of SF/HBC blend nanofibrous scaffolds was improved. The results indicated that the rearrangement of HBC and SF molecular chain formed a new structure due to stronger hydrogen bonding between SF and HBC. These electrospun SF/HBC blended nanofibers may provide an ideal tissue engineering scaffold and wound dressing.


Subject(s)
Chitosan/chemistry , Fibroins/chemistry , Nanofibers/chemistry , Hydrogen Bonding , Protein Structure, Secondary , Silk/chemistry , Temperature , Thermogravimetry , Tissue Engineering , X-Ray Diffraction
7.
Int J Biol Macromol ; 191: 600-607, 2021 Nov 30.
Article in English | MEDLINE | ID: mdl-34582906

ABSTRACT

In order to obtain a synergistic antimicrobial effect of cuprous oxide nanoparticles (Cu2O NPs) and chitosan (CS) nanofibers, the nano Cu2O/CS nanofibrous scaffolds were synthesized in situ via two subsequent steps of chelation and reduction. The Cu2+ were stably chelated on CS nanofibrous scaffolds through the coordination of amino group (-NH2) and hydroxyl group (-OH) on CS with Cu2+, and then the chelated Cu2+ were reduced to nano Cu2O by Vitamin C under alkaline conditions. And by the measurements of XRD, XPS and FTIR-ATR, the results showed that Cu2O NPs were successfully deposited on the CS nanofibrous scaffolds. SEM clarified that the particle size of Cu2O gradually decreased and the shape changed from cubic to irregular with the increase of CuSO4 concentration. With the CuSO4 concentration of 0.02 and 0.04 mol·L-1, the Cu2O/CS nanofibrous scaffolds presented outstanding hydrophilicity and antibacterial activity against Gram-negative Escherichia coli (E. coli) and Gram-positive Staphylococcus aureus (S. aureus) comparing to the CS nanofibrous scaffolds, meanwhile, they possessed good biocompatibility. This kind of nanofibrous scaffolds deposited with nano Cu2O would have broad application prospects in the field of antibacterial biomaterials.


Subject(s)
Anti-Infective Agents/chemistry , Chitosan/analogs & derivatives , Copper/chemistry , Nanofibers/chemistry , Tissue Scaffolds/chemistry , Animals , Anti-Infective Agents/pharmacology , Hydrophobic and Hydrophilic Interactions , Mice , NIH 3T3 Cells , Staphylococcus aureus/drug effects
8.
ACS Appl Mater Interfaces ; 13(43): 50785-50801, 2021 Nov 03.
Article in English | MEDLINE | ID: mdl-34664947

ABSTRACT

Neovascularization is crucial for peripheral nerve regeneration and long-term functional restoration. Previous studies have emphasized strategies that enhance axonal repair over vascularization. Here, we describe the development and application of an in situ prevascularization strategy that uses 3D porous nerve guidance conduits (NGCs) to achieve angiogenesis-mediated neural regeneration. The optimal porosity of the NGC is a critical feature for achieving neovascularization and nerve growth patency. Hollow silk fibroin/poly(l-lactic acid-co-ε-caprolactone) NGCs with 3D sponge-like walls were fabricated using electrospinning and freeze-drying. In vitro results showed that 3D porous NGC favored cell biocompatibility had neuroregeneration potential and, most importantly, had angiogenic activity. Results from our mechanistic studies suggest that activation of HIF-1α signaling might be associated with this process. We also tested in situ prevascularized 3D porous NGCs in vivo by transplanting them into a 10 mm rat sciatic nerve defect model with the aim of regenerating the severed nerve. The prevascularized 3D porous NGCs greatly enhanced intraneural angiogenesis, resulting in demonstrable neurogenesis. Eight weeks after transplantation, the performance of the prevascularized 3D NGCs was similar to that of traditional autografts in terms of improved anatomical structure, morphology, and neural function. In conclusion, combining a reasonably fabricated 3D-pore conduit structure with in situ prevascularization promoted functional nerve regeneration, suggesting an alternative strategy for achieving functional recovery after peripheral nerve trauma.


Subject(s)
Fibroins/pharmacology , Nanofibers/chemistry , Neovascularization, Physiologic/drug effects , Nerve Regeneration/drug effects , Polyesters/pharmacology , Tissue Engineering , Animals , Cells, Cultured , Fibroins/chemistry , Freeze Drying , Humans , Materials Testing , Particle Size , Polyesters/chemistry , Porosity , Rats , Surface Properties
9.
Int J Mol Sci ; 11(9): 3529-39, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20957110

ABSTRACT

Chitosan, a naturally occurring polysaccharide with abundant resources, has been extensively exploited for various biomedical applications, typically as wound dressings owing to its unique biocompatibility, good biodegradability and excellent antibacterial properties. In this work, composite nanofibrous membranes of chitosan (CS) and silk fibroin (SF) were successfully fabricated by electrospinning. The morphology of electrospun blend nanofibers was observed by scanning electron microscopy (SEM) and the fiber diameters decreased with the increasing percentage of chitosan. Further, the mechanical test illustrated that the addition of silk fibroin enhanced the mechanical properties of CS/SF nanofibers. The antibacterial activities against Escherichia coli (Gram negative) and Staphylococcus aureus (Gram positive) were evaluated by the turbidity measurement method; and results suggest that the antibacterial effect of composite nanofibers varied on the type of bacteria. Furthermore, the biocompatibility of murine fibroblast on as-prepared nanofibrous membranes was investigated by hematoxylin and eosin (H&E) staining and MTT assays in vitro, and the membranes were found to promote the cell attachment and proliferation. These results suggest that as-prepared chitosan/silk fibroin (CS/SF) composite nanofibrous membranes could be a promising candidate for wound healing applications.


Subject(s)
Anti-Bacterial Agents/chemistry , Bandages , Biocompatible Materials/chemistry , Chitosan/chemistry , Fibroins/chemistry , Nanofibers/chemistry , Animals , Anti-Bacterial Agents/pharmacology , Biocompatible Materials/pharmacology , Cell Line , Fibroblasts/drug effects , Fibroins/pharmacology , Mice , Staphylococcus aureus/drug effects
10.
J Biomater Sci Polym Ed ; 31(5): 622-641, 2020 04.
Article in English | MEDLINE | ID: mdl-31852372

ABSTRACT

Graphite oxide (GO) and chitosan (CS) nanofibers have aroused intense interest as wound dressing due to their physicochemical, antimicrobial properties and nanotopography. In this study, GO nanosheets were coated on shell (chitosan, CS)-core (L-polylactic acid, PLLA) structured nanofibrous scaffolds to create a synergistic microenvironment for wound healing. Through scanning electron microscopy (SEM) and atomic force microscopy (AFM) tests, results showed that the surface of GO-coated CS/PLLA nanofibers presented corrugated wrinkles and rougher than that of CS/PLLA nanofibers, and the GO nanosheets did not destroy the structure of nanofibers. X-ray photoelectron spectroscopy (XPS) and thermogravimetric analysis (TGA) demonstrated that GO nanosheets were successfully coated on CS/PLLA nanofibrous scaffolds. Furthermore, the coatings of GO nanosheets significantly improved the hydrophilicity of CS/PLLA nanofibrous scaffolds. GO-coated CS/PLLA nanofibrous scaffolds revealed more excellent antimicrobial activity to Gram-negative Escherichia coli (E. coli) and Gram-positive Staphylococcus aureus (S. aureus) than that of CS/PLLA nanofibrous scaffolds, meanwhile, they promoted the proliferation of pig iliac endothelial cells (PIECs). Rats wounds covered by GO-coated CS/PLLA nanofibrous scaffolds were healed better than other groups on pathological section. This type of nanofibrous scaffolds with GO nanosheets would possess an excellent potential in wound healing process.


Subject(s)
Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Chitosan/chemistry , Graphite/chemistry , Nanofibers/chemistry , Polyesters/chemistry , Wound Healing/drug effects , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Proliferation/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Swine
11.
Acta Biomater ; 117: 180-191, 2020 11.
Article in English | MEDLINE | ID: mdl-33007489

ABSTRACT

Development of a functional nerve conduit to replace autografts remains a significant challenge particularly considering the compositional complexity and structural hierarchy of native peripheral nerves. In the present study, a multiscale strategy was adopted to fabricate 3D biomimetic nerve conduit from Antheraea pernyi silk fibroin (ApF)/(Poly(L-lactic acid-co-caprolactone)) (PLCL)/graphene oxide (GO) (ApF/PLCL/GO) nanofibers via nanofiber dispersion, template-molding, freeze-drying and crosslinking. The resultant conduits exhibit parallel multichannels (ϕ = 125 µm) surrounded by biomimetic fibrous fragments with tailored degradation rate and improved mechanical properties in comparison with the scaffold without GO. In vitro studies showed that such 3D biomimetic nerve scaffolds had the ability to offer an effective guiding interface for neuronal cell growth. Furthermore, these conduits showed a similarity to autografts in vivo repairing sciatic nerve defects based on a series of analysis (walking track, triceps weight, morphogenesis, vascularization, axonal regrowth and myelination). The conduits almost completely degraded within 12 weeks. These findings demonstrate that the 3D hierarchical nerve guidance conduit (NGC) with fascicle-like structure have great potential for peripheral nerve repair.


Subject(s)
Guided Tissue Regeneration , Nanofibers , Animals , Biomimetics , Nerve Regeneration , Rats , Rats, Sprague-Dawley , Sciatic Nerve , Tissue Scaffolds
12.
Acta Biomater ; 83: 291-301, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30541701

ABSTRACT

Topographical cues presented by aligned nanofibers have been demonstrated to stimulate peripheral nerve regeneration across long gaps, but the underlying mechanisms remain incompletely elucidated. Because macrophages play a crucial role in peripheral nerve regeneration and can be phenotypically modulated by topographical cues, we hypothesized that aligned nanofibers might induce the development of macrophage phenotypes that facilitate the regeneration of peripheral nerves. Here, macrophages were seeded on aligned and random poly(l-lactic acid-co-ε-caprolactone) nanofibers and their morphology and phenotypes were compared. Aligned nanofibers drastically stimulated macrophage elongation along the nanofibers, and, more importantly, induced the development of a pro-healing macrophage phenotype (M2 type), whereas random nanofibers induced a proinflammatory phenotype (M1 type). Notably, the macrophages polarized by aligned nanofibers potently promoted the proliferation and migration of Schwann cells in vitro. Thus, we constructed nerve-guidance conduits by using aligned and random nanofibers and evaluated their effects on macrophage polarization and nerve regeneration in a rat sciatic nerve defect model. Our in vivo results showed that the ratio of pro-healing macrophages was again higher in the aligned-nanofiber group, and further that Schwann cell infiltration and axon numbers were 2.0- and 2.84-fold higher in the aligned group than in the random group, respectively. This study demonstrates that nanofiber arrangement differentially regulates macrophage activation and that nerve-guidance conduits constructed from aligned nanofibers markedly facilitate peripheral nerve regeneration at least partly by promoting the pro-healing phenotype in macrophages. STATEMENT OF SIGNIFICANCE: The effect of aligned nanofibers on peripheral nerve regeneration has been well established. However, the underlying mechanism remains unclear. Since macrophages play an important role in peripheral nerve regeneration, and can be phenotypically modulated by topographical cues, we hypothesized that aligned nanofibers may exert their beneficial effects via modulating macrophage phenotypes. This study demonstrates for the first time that nanofiber arrangement differentially modulates macrophage shape and polarization, and this subsequently influences the outcome of peripheral nerve regeneration. These findings reveals a novel relationship between biomaterial structure and macrophage activation, contributes to clarifying the mechanism of surface topography in tissue regeneration, and highlight the potential application prospect of aligned nanofiber scaffolds in nerve regeneration and wound healing.


Subject(s)
Macrophages , Nanofibers/chemistry , Nerve Regeneration , Peripheral Nerve Injuries , Peripheral Nerves/physiology , Schwann Cells/pathology , Animals , Cell Proliferation , Macrophages/metabolism , Macrophages/pathology , Male , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/therapy , Polyesters/chemistry , Rats , Rats, Sprague-Dawley , Schwann Cells/metabolism
14.
Beijing Da Xue Xue Bao Yi Xue Ban ; 40(1): 43-6, 2008 Feb 18.
Article in Zh | MEDLINE | ID: mdl-18278136

ABSTRACT

OBJECTIVE: To investigate the effects of Epimedium on proliferation, function and apoptosis of mouse osteoblasts in vitro. METHODS: Primary osteoblasts were obtained by sequential digestion of mouse calvaria with collagenase and hyaluronidase. The identification of derived cells was done by histochemical staining of alkaline phosphatase (ALPase) and immunohistochemical staining of type I collagen, bone sialoprotein and osteopontin. MTT assay was employed to examine the proliferation of osteoblasts after treatment with Epimedium. The alkaline phosphatase activity level of mouse osteoblasts was also determined through an enzyme dynamical method. Apoptosis of osteoblasts was induced by dexamethasone and flow cytometry was utilized to examine the effects of Epimedium on the dexamethasone-induced apoptosis of osteoblasts. RESULTS: Five populations of bone cells were obtained by sequential digestion. Osteoblasts were purely obtained by discarding the first two populations and identified by the positive staining of ALPase, type I collagen, bone sialoprotein, and osteopontin. The alkaline phosphatase activity level of osteoblasts was significantly increased by the addition of Epimedium at 0.1 - 10 g/L, with the most significant increase at 1 g/L. On the other hand, the proliferation of osteoblasts was not affected after different doses of Epimedium added into the culture medium. Determined by flow cytometry, apoptosis of osteoblasts were induced by treatment with dexamethasone for 72 h. However, simultaneous administration of 1 g/L Epimedium had no effects on dexamethasone-induced apoptosis in osteoblasts. CONCLUSION: Epimedium did not affect the cell proliferation and cell survival of mouse osteoblasts, but could significantly increase alkaline phosphatase activity of the cells. The increase of alkaline phosphatase activity by Epimedium in osteoblasts may be one of the important mechanisms by which Epimedium can effectively prevent osteoporosis.


Subject(s)
Apoptosis/drug effects , Epimedium , Osteoblasts/drug effects , Plant Preparations/pharmacology , Alkaline Phosphatase/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Mice , Mice, Inbred Strains , Osteoblasts/cytology , Osteoblasts/metabolism
15.
J Biomed Mater Res A ; 106(7): 2070-2077, 2018 07.
Article in English | MEDLINE | ID: mdl-29575774

ABSTRACT

Silk fibroin (SF)-based nerve conduits have been widely used to bridge peripheral nerve defects. Our previous study showed that nerve regeneration in a SF-blended poly (l-lactide-co-ɛ-caprolactone) [P(LLA-CL)] nerve conduit is better than that in a P(LLA-CL) conduit. However, the involved mechanisms remain unclarified. Because angiogenesis within a nerve conduit plays an important role in nerve regeneration, vascularization of SF/P(LLA-CL) and P(LLA-CL) conduits was compared both in vitro and in vivo. In the present study, we observed that SF/P(LLA-CL) nanofibers significantly promoted fibroblast proliferation, and vascular endothelial growth factor secreted by fibroblasts seeded in SF/P(LLA-CL) nanofibers was more than seven-fold higher than that in P(LLA-CL) nanofibers. Conditioned medium of fibroblasts in the SF/P(LLA-CL) group stimulated more human umbilical vein endothelial cells (HUVEC) to form capillary-like networks and promoted faster HUVEC migration. The two kinds of nerve conduits were used to bridge 10-mm-length nerve defects in rats. At 3 weeks of reparation, the blood vessel area in the SF/P(LLA-CL) group was significantly larger than that in the P(LLA-CL) group. More regenerated axons and Schwann cells were also observed in the SF/P(LLA-CL) group, which was consistent with the results of blood vessels. Collectively, our data revealed that the SF/P(LLA-CL) nerve conduit enhances peripheral nerve regeneration by improving angiogenesis within the conduit. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2070-2077, 2018.


Subject(s)
Fibroins/pharmacology , Neovascularization, Physiologic/drug effects , Nerve Regeneration/drug effects , Peripheral Nerves/physiology , Tissue Scaffolds/chemistry , Animals , Antigens, CD34/metabolism , Axons/drug effects , Axons/metabolism , Culture Media, Conditioned/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Peripheral Nerves/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Rats, Sprague-Dawley , Schwann Cells/cytology , Schwann Cells/drug effects , Wound Healing/drug effects
16.
Br J Oral Maxillofac Surg ; 45(6): 467-70, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17254677

ABSTRACT

PURPOSE: To evaluate the role of nutrition in the development of postoperative complications in patients with oral and maxillofacial malignancy. PATIENTS AND METHODS: Ninety-six patients treated surgically for oral and maxillofacial malignancy, 27 of whom developed postoperative complications; the remaining 69 recovered uneventfully. Nutritional state and clinical variables in the two groups were compared. RESULTS: The incidence of poor nutrition was greater in the complication group (56%) than in the uncomplicated group (20%) (p<0.001); the values for body weight, triceps skinfold thickness, arm circumference, arm muscle circumference, and creatinine-height index decreased more in the complicated than in the uncomplicated group (p<0.001); nitrogen and calorie intake during the first postoperative week was less in the complicated than in the uncomplicated group (p<0.001). CONCLUSIONS: Poor nutrition plays an important part in the development of postoperative complications, and perioperative nutritional support of patients with oral and maxillofacial cancer must be properly managed.


Subject(s)
Jaw Neoplasms/surgery , Malnutrition/complications , Mouth Neoplasms/surgery , Postoperative Complications/etiology , Carcinoma, Squamous Cell/surgery , Female , Humans , Male , Middle Aged , Nutrition Assessment , Nutritional Status , Salivary Gland Neoplasms/surgery
17.
J Biomed Mater Res A ; 105(7): 1900-1910, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28256802

ABSTRACT

Biomimicing topological structure of natural nerve tissue to direct axon growth and controlling sustained release of moderate neurotrophic factors are extremely propitious to the functional recovery of damaged nervous systems. In this study, the heparin/collagen encapsulating nerve growth factor (NGF) multilayers were coated onto the aligned poly-L-lactide (PLLA) nanofibrous scaffolds via a layer-by-layer (LbL) self-assembly technique to combine biomolecular signals, and physical guidance cues for peripheral nerve regeneration. Scanning electronic microscopy (SEM) revealed that the surface of aligned PLLA nanofibrous scaffolds coated with heparin/collagen multilayers became rougher and appeared some net-like filaments and protuberances in comparison with PLLA nanofibrous scaffolds. The heparin/collagen multilayers did not destroy the alignment of nanofibers. X-ray photoelectron spectroscopy and water contact angles displayed that heparin and collagen were successfully coated onto the aligned PLLA nanofibrous scaffolds and improved its hydrophilicity. Three-dimensional (3 D) confocal microscopy images further demonstrated that collagen, heparin, and NGF were not only coated onto the surface of aligned PLLA nanofibrous scaffolds but also permeated into the inner of scaffolds. Moreover, NGF presented a sustained release for 2 weeks from aligned nanofibrous scaffolds coated with 5.5 bilayers or above and remained good bioactivity. The heparin/collagen encapsulating NGF multilayers coated aligned nanofibrous scaffolds, in particular 5.5 bilayers or above, was more beneficial to Schwann cells (SCs) proliferation and PC12 cells differentiation as well as the SC cytoskeleton and neurite growth along the direction of nanofibrous alignment compared to the aligned PLLA nanofibrous scaffolds. This novel scaffolds combining sustained release of bioactive NGF and aligned nanofibrous topography presented an excellent potential in peripheral nerve regeneration. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1900-1910, 2017.


Subject(s)
Collagen/chemistry , Heparin/chemistry , Immobilized Proteins , Nanofibers/chemistry , Nerve Growth Factor , Nerve Tissue/metabolism , Polyesters/chemistry , Tissue Scaffolds/chemistry , Animals , Immobilized Proteins/chemistry , Immobilized Proteins/pharmacokinetics , Immobilized Proteins/pharmacology , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacokinetics , Nerve Growth Factor/pharmacology , Nerve Tissue/cytology , PC12 Cells , Rats , Tissue Engineering/methods
18.
Acta Biomater ; 37: 131-42, 2016 06.
Article in English | MEDLINE | ID: mdl-27063493

ABSTRACT

UNLABELLED: The graphene oxide (GO) has attracted tremendous attention in biomedical fields. In order to combine the unique physicochemical properties of GO nanosheets with topological structure of aligned nanofibrous scaffolds for nerve regeneration, the GO nanosheets were coated onto aligned and aminolyzed poly-l-lactide (PLLA) nanofibrous scaffolds. Scanning electronic microscopy (SEM) and atomic force microscopy (AFM) revealed that the surface of aligned PLLA nanofibers after being coated with GO became rougher than those of the aligned PLLA and aminolyzed PLLA nanofibrous scaffolds. The GO nanosheets did not destroy the alignment of nanofibers. The characterizations of X-ray photoelectron spectroscopy (XPS) and water contact angle displayed that the aligned PLLA nanofibrous scaffolds were introduced with hydrophilic groups such as NH2, COOH, and OH after aminolysis and GO nanosheets coating, showing better hydrophilicity. The GO-coated and aligned PLLA nanofibrous scaffolds significantly promoted Schwann cells (SCs) proliferation with directed cytoskeleton along the nanofibers compared with the aligned PLLA and aminolyzed PLLA nanofibrous scaffolds. These scaffolds also greatly improved the proliferation of rat pheochromocytoma 12 (PC12) cells, and significantly promoted their differentiation and neurite growth along the nanofibrous alignment in the presence of nerve growth factor (NGF). This type of scaffolds with nanofibrous surface topography and GO nanosheets is expected to show better performance in nerve regeneration. STATEMENT OF SIGNIFICANCE: Recovery of damaged nerve functions remains a principal clinical challenge in spite of surgical intervention and entubulation. The use of aligned fibrous scaffolds provides suitable microenvironment for nerve cell attachment, proliferation and migration, enhancing the regeneration outcome of nerve tissue. Surface modification is generally required for the synthetic polymeric fibers by laminin, fibronectin and YIGSR peptides to stimulate specific cell functions and neurite outgrowth. Yet these proteins or peptides present the poor processibility, limited availability, and high cost, influencing their application in clinic. In this work, we combined GO nanosheets and topological structure of aligned nanofibrous scaffolds to direct cell migration, proliferation, and differentiation, and to induce neurite outgrowth for nerve regeneration. The GO coating improved several biomedical properties of the aligned PLLA nanofibrous scaffolds including surface roughness, hydrophilicity and promotion of cells/material interactions, which significantly promoted SCs growth and regulated cell orientation, and induced PC12 cells differentiation and neurite growth. The design of this type of structure is of both scientific and technical importance, and possesses broad interest in the fields of biomaterials, tissue engineering and regenerative medicine.


Subject(s)
Cell Proliferation/drug effects , Coated Materials, Biocompatible , Graphite , Nanofibers/chemistry , Neurons/metabolism , Polyesters , Tissue Scaffolds/chemistry , Animals , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Graphite/chemistry , Graphite/pharmacology , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacology , PC12 Cells , Polyesters/chemistry , Polyesters/pharmacology , Rats
19.
Di Yi Jun Yi Da Xue Xue Bao ; 22(2): 102-6, 2002 Feb.
Article in Zh | MEDLINE | ID: mdl-12390798

ABSTRACT

OBJECTIVE: To investigate the distribution of the substance P (SP) and its receptor in the marginal division (MrD) of rat striatum and to understand the relationship between SP and the learning and memory function of rats. METHODS: Using immunohistochemistry and in situ hybridization techniques, the distribution of SP and its receptor in the MrD was studied, and the relationship between the SP and learning and memory of the MrD was observed by means of SP receptor gene knockout in combination with Y-maze test. RESULTS: Numerous SP immunopositive fibers and large quantities of SP receptor protein and NK1 mRNA were identified in the MrD of rat striatum. After knockout of the SP receptor gene in the MrD, the ability of learning and memory of the rats was obviously decreased. CONCLUSION: SP and its receptor in the MrD may play important roles in the learning and memory function of rat, possibly through the regulation of the neurotransmitters as 5-HT by SP via NK1 receptor.


Subject(s)
Corpus Striatum/metabolism , Learning/physiology , Memory/physiology , Receptors, Neurokinin-1/metabolism , Substance P/metabolism , Analysis of Variance , Animals , Male , Rats , Rats, Sprague-Dawley , Receptors, Neurokinin-1/physiology , Substance P/physiology
20.
J Mater Chem B ; 2(45): 7945-7954, 2014 Dec 07.
Article in English | MEDLINE | ID: mdl-32262084

ABSTRACT

Currently, electroactive biomaterials have often been fabricated as tissue engineering scaffolds to provide electrical stimulation for neural tissue engineering. The goal of this work was to study the synergistic effect of electrical stimulation and nerve growth factor (NGF) on neuron growth. The composite meshes of polyaniline (PANi) and well-blended poly(l-lactic acid-co-ε-caprolactone)/silk fibroin (PS) incorporated with nerve growth factor (NGF) were prepared by coaxial electrospinning. The results showed that the increased concentration of PANi had a large effect on the fiber diameter, which was significantly reduced from 683 ± 138 nm to 411 ± 98 nm and then increased to 498 ± 100 nm. The contact angles and Young's modulus decreased to 28.3°± 5.4° and 7.2 ± 1.2 MPa, respectively, and the conductance increased to 30.5 ± 3.1 mS cm-1. The results of the viability and morphology of mouse Schwann cells on the nanofibrous meshes showed that PS-PANi-1 loaded with NGF exhibited the highest cell number after 5 days culture and the aligned nanofibers could guide cell orientation. The synergistic effects of electrical stimulation and NGF were also investigated via the growth and differentiation of rat pheochromocytoma 12 (PC12) cells. The scaffolds loaded with NGF under electrical stimulation could effectively support PC12 neurite outgrowth and increase the percentage of neurite-bearing cells as well as the median neurite length. More importantly, the NGF release from the conductive core-shell structure nanofiber could be increased by electrical stimulation. These promising results demonstrated that there was a potential use of this functional scaffold for nerve tissue regeneration.

SELECTION OF CITATIONS
SEARCH DETAIL