Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 903
Filter
1.
Int J Biol Macromol ; 266(Pt 1): 131221, 2024 May.
Article in English | MEDLINE | ID: mdl-38554926

ABSTRACT

Oral ulceration is the most common oral mucosal disease. Oral mucosal ulcers are extremely painful, may interfere with eating and speaking, and potentially complicate systemic symptoms in severe cases. The humid and highly dynamic environment of the oral cavity makes local drug administration for treating oral mucosal ulcers challenging. To overcome these challenges, we designed and prepared a novel dissolving microneedle (MN) patch containing multiple drugs in a core-shell to promote oral ulcer healing. The MNs contained a methacrylate gelatin shell layer of basic fibroblast growth factor (bFGF), a hyaluronic acid (HA) core loaded with dexamethasone (DXMS), and zeolite imidazoline framework-8 (ZIF-8) encapsulated in the HA-based backplane. Progressive degradation of gelatin methacryloyl (GelMA) from the tip of the MN patch in the oral mucosa resulted in sustained bFGF release at the lesion site, significantly promoting cell migration, proliferation, and angiogenesis. Moreover, the rapid release of HA and, subsequently, DXMS inhibited inflammation, and the remaining MN backing after the tip dissolved behaved as a dressing, releasing ZIF-8 for its antimicrobial effects. This novel, multifunctional, transmucosal core-shell MN patch exhibited excellent anti-inflammatory, antimicrobial, and pro-healing effects in vivo and in vitro, suggesting that it can promote oral ulcer healing.


Subject(s)
Gelatin , Hyaluronic Acid , Methacrylates , Mouth Mucosa , Needles , Oral Ulcer , Wound Healing , Hyaluronic Acid/chemistry , Gelatin/chemistry , Animals , Oral Ulcer/drug therapy , Oral Ulcer/pathology , Mouth Mucosa/drug effects , Mouth Mucosa/metabolism , Methacrylates/chemistry , Wound Healing/drug effects , Rats , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Fibroblast Growth Factor 2/administration & dosage , Male , Mice , Humans
2.
Theranostics ; 12(17): 7237-7249, 2022.
Article in English | MEDLINE | ID: mdl-36438504

ABSTRACT

Background: The only effective treatment for myocardial infarction (MI) is the timely restoration of coronary blood flow in the infarcted area, but further reperfusion exacerbates myocardial injury and leads to distal coronary no-reflow, which affects patient prognosis. Angiogenesis could be an important therapeutic strategy for re-establishing the blood supply to save the ischemic myocardium after MI. Basic fibroblast growth factor (bFGF) has been shown to promote angiogenesis. However, direct intravenous administration of bFGF is not a viable option given its poor half-life in vivo. Methods: Herein, we developed a peptide Lys-Lys-Pro-Leu-Gly-Leu-Ala-Gly-Phe-Phe (K2) to encapsulate bFGF to form bFGF@K2 micelle and proposed an enzyme-instructed self-assembly (EISA) strategy to deliver and slowly release bFGF in the ischemic myocardium. Results: The bFGF@K2 micelle exerted a stronger cardioprotective effect than free bFGF in a rat model of myocardial ischemia-reperfusion (MI/R). In vitro results revealed that the bFGF@K2 micelle could be cleaved by matrix metallopeptidase 9 (MMP-9) to yield bFGF@Nanofiber through amphipathic changes. In vivo experiments indicated that intravenous administration of bFGF@K2 micelle could lead to their restructuring into bFGF@Nanofiber and long term retention of bFGF in the ischemic myocardium of rat due to high expression of MMP-9 and assembly-induced retention (AIR) effect, respectively. Twenty-eight days after MI/R model establishment, bFGF@K2 micelle treatment significantly reduced fibrosis and improved cardiac function of the rats. Conclusion: We predict that our strategy could be applied in clinic for MI treatment in the future.


Subject(s)
Fibroblast Growth Factor 2 , Matrix Metalloproteinase 9 , Myocardial Infarction , Myocardial Reperfusion Injury , Nanofibers , Animals , Rats , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/therapeutic use , Matrix Metalloproteinase 9/metabolism , Micelles , Myocardial Infarction/complications , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/therapy , Myocardium/metabolism , Myocardium/pathology , Nanofibers/administration & dosage , Nanofibers/therapeutic use , Neovascularization, Pathologic , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/etiology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/therapy
3.
Colloids Surf B Biointerfaces ; 209(Pt 2): 112135, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34655915

ABSTRACT

Deep burns are a common form of trauma worldwide, and they are hard to be cured in a short time and enhance psychological pressure of the patients. How to effectively promote the healing of wounds after burns is a continuing challenge currently faced by burn physicians. Various strategies of promoting wound healing of deep burns have been developed, including gene therapy and growth factor therapy. In this study, we developed a combined therapy using PLGA nanoparticles as carriers to deliver bFGF and VEGFA genes to promote healing of burn wounds. We first inserted the bFGF and VEGFA genes into pEGFP-N1 vectors and loaded the mixed generated plasmids into PLGA nanoparticles. Next, we injected the nanoparticle/plasmid complexes into the rats intracutaneously and found that the complexes were successfully transfected in vivo one week later. Finally, we injected the nanoparticle/plasmid complexes containing bFGF and VEGFA around burn wounds. We found that the percentage of wound healing of rats treated with nanoparticles/bFGF+ VEGFA plasmid complexes was higher than that of rats in the scald control group, and the early percentage of wound complete epithelialization was also higher. Therefore, combining gene therapy with nanoparticles may be an effective biological strategy for wound repair.


Subject(s)
Burns , Fibroblast Growth Factor 2/administration & dosage , Nanoparticles , Vascular Endothelial Growth Factor A/administration & dosage , Wound Healing , Animals , Burns/therapy , Fibroblast Growth Factor 2/genetics , Rats , Re-Epithelialization , Vascular Endothelial Growth Factor A/genetics
4.
Invest Ophthalmol Vis Sci ; 62(9): 34, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34293078

ABSTRACT

Purpose: Mutations in the fibroblast growth factor (FGF) receptor can result in strabismus, but little is known about how FGFs affect extraocular muscle structure and function. These were assessed after short-term and long-term exposure to exogenously applied FGF2 to determine the effect of enhanced signaling. Methods: One superior rectus muscle of adult rabbits received either a series of three injections of 500 ng, 1 µg, or 5 µg FGF2 and examined after 1 week, or received sustained treatment with FGF2 and examined after 1, 2, or 3 months. Muscles were assessed for alterations in force generation, myofiber size, and satellite cell number after each treatment. Results: One week after the 5 µg FGF2 injections, treated muscles showed significantly increased force generation compared with naïve controls, which correlated with increased myofiber cross-sectional areas and Pax7-positive satellite cells. In contrast, 3 months of sustained FGF2 treatment resulted in decreased force generation, which correlated with decreased myofiber size and decreased satellite cells compared with naïve control and the untreated contralateral side. Conclusions: FGF2 had distinctly different effects when short-term and long-term treatments were compared. The decreased size and ability to generate force correlated with decreased myofiber areas seen in individuals with Apert syndrome, where there is sustained activation of FGF signaling. Knowing more about signaling pathways critical for extraocular muscle function, development, and disease will pave the way for improved treatment options for strabismus patients with FGF abnormalities in craniofacial disease, which also may be applicable to other strabismus patients.


Subject(s)
Fibroblast Growth Factor 2/administration & dosage , Muscle Contraction/drug effects , Oculomotor Muscles/cytology , Animals , Injections, Intramuscular , Models, Animal , Muscle Contraction/physiology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Oculomotor Muscles/physiology , Rabbits
5.
Gene ; 788: 145664, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33887371

ABSTRACT

The angiogenic gene therapy is an attractive approach for the treatment of ischemic muscle diseases, including peripheral arterial disease and ischemic heart diseases. Although a variety of gene transfer methods have been developed, the efficiency of gene transfer is still limited. We have been developing the needleless high-energy bioinjector device, Pyro-drive Jet Injector (PJI), based on pyrotechnics using a combination of ignition powder and gunpowder, however, the utility of PJI in gene transfer into muscle tissues remains unclear. pcDNA3.1 plasmid containing Flag was injected to the thigh muscles of C57BL/6J mice using PJI or needle, as a control. Histological analysis demonstrated that the protein expression of Flag was observed in a wider range in PJI group than in needle group. To assess the validity of PJI for gene therapy, pcDNA3.1-human fibroblast growth factor 2 (FGF2), which has angiogenic activity and tissue protective properties, was injected into the ischemic thigh muscles with PJI or needle. ELISA assay revealed that the protein expression of FGF2 was increased in the thigh muscle tissues by PJI-mediated gene delivery. Significantly, histological analyses revealed that muscle fiber cross-sectional area and the number of endothelial marker CD31 (+) cells was increased in ischemic hind-limb tissues of the PJI-FGF2 group but not in those of needle-FGF2 group. To expand the applicability of the PJI-mediated gene transfer, pcDNA3.1-venus plasmid was injected into murine hearts with PJI or needle. PJI method was successful in gene transfer into murine hearts, especially into cardiomyocytes, with high efficiency when compared to needle method. Collectively, the non-needle, non-liposomal and non-viral gene transfer by PJI could be a novel therapeutic approach for muscle diseases.


Subject(s)
Fibroblast Growth Factor 2/administration & dosage , Gene Transfer Techniques/instrumentation , Muscle, Skeletal/metabolism , Myocardium/metabolism , Animals , Cell Line , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Genetic Therapy/instrumentation , Genetic Vectors/administration & dosage , Genetic Vectors/pharmacology , Hindlimb , Humans , Male , Mice , Mice, Inbred C57BL , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/therapy , Plasmids/genetics
6.
Neurosci Lett ; 755: 135936, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33910061

ABSTRACT

Müller glia can act as endogenous stem cells and regenerate the missing neurons in the injured or degenerating retina in lower vertebrates. However, mammalian Müller glia, although can sometimes express stem cell markers and specific neuronal proteins in response to injury or degeneration, do not differentiate into functional neurons. We asked whether bFGF and insulin would stimulate the Müller glia to migrate, proliferate and differentiate into photoreceptors in rd1 mouse. We administered single or repeated (two or three) intravitreal injections of basic fibroblast growth factor (bFGF;200 µg) and insulin (2 µg) in 2-week-old rd1 mice. Müller glia were checked for proliferation, migration and differentiation using immunostaining. A single injection resulted within 5 days in a decrease in the numbers of Müller glia in the inner nuclear layer (INL) and a corresponding increase in the outer nuclear layer (ONL). The total number of Müller glia in the INL and ONL was unaltered, suggesting that they did not proliferate, but migrated from INL to ONL. However, maintaining the Müller cells in the ONL for two weeks or longer required repeated injections of bFGF and insulin. Interestingly, all Müller cells in the ONL expressed chx10, a stem cell marker. We did not find any immunolabeling for rhodopsin, m-opsin or s-opsin in the Müller glia in the ONL.


Subject(s)
Cell Movement/drug effects , Ependymoglial Cells/drug effects , Fibroblast Growth Factor 2/administration & dosage , Insulin/administration & dosage , Neuroglia/drug effects , Photoreceptor Cells/drug effects , Animals , Cell Movement/physiology , Ependymoglial Cells/chemistry , Ependymoglial Cells/metabolism , Intravitreal Injections , Mice , Mice, Inbred CBA , Mice, Transgenic , Nerve Regeneration/drug effects , Nerve Regeneration/physiology , Neuroglia/chemistry , Neuroglia/metabolism , Photoreceptor Cells/chemistry , Photoreceptor Cells/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology
7.
PLoS One ; 16(2): e0243727, 2021.
Article in English | MEDLINE | ID: mdl-33534866

ABSTRACT

In vitro embryo production systems are limited by their inability to consistently produce embryos with the competency to develop to the blastocyst stage, survive cryopreservation, and establish a pregnancy. Previous work identified a combination of three cytokines [fibroblast growth factor 2 (FGF2), leukemia inhibitory factor (LIF), and insulin-like growth factor 1 (IGF1)], called FLI, that we hypothesize improve preimplantation development of bovine embryos in vitro. To test this hypothesis, FLI was supplemented into oocyte maturation or embryo culture medium. Embryos were produced in vitro using abattoir-derived oocytes and fertilized with sperm from a single bull known to have high fertility. After an 18-20 h fertilization period, putative zygotes were cultured in synthetic oviductal fluid (SOF) for 8 days. The addition of FLI to the oocyte maturation medium increased (P < 0.05) the dissociation of transzonal projections at 12, 18, and 24 h of maturation, as well as, the proportion of oocytes that reached the metaphase II stage of meiosis. Additionally, lipid content was decreased (P < 0.05) in the blastocyst stage embryo. The addition of FLI during the culture period increased development to the blastocyst stage, cytoskeleton integrity, and survival following slow freezing, as well as, decreased post thaw cell apoptosis (P < 0.05). In conclusion, the supplementation of these cytokines in vitro has the potential to alleviate some of the challenges associated with the cryo-survival of in vitro produced bovine embryos through improving embryo development and embryo quality.


Subject(s)
Cattle/embryology , Cryopreservation/veterinary , Embryo, Mammalian/embryology , Fibroblast Growth Factor 2 , Insulin-Like Growth Factor I , Leukemia Inhibitory Factor , Animals , Blastocyst/cytology , Blastocyst/drug effects , Blastocyst/ultrastructure , Cryopreservation/methods , Embryo, Mammalian/drug effects , Embryo, Mammalian/ultrastructure , Female , Fertilization in Vitro/methods , Fertilization in Vitro/veterinary , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/pharmacology , In Vitro Oocyte Maturation Techniques/methods , In Vitro Oocyte Maturation Techniques/veterinary , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacology , Leukemia Inhibitory Factor/administration & dosage , Leukemia Inhibitory Factor/pharmacology , Pregnancy
8.
Laryngoscope ; 131(9): 2059-2064, 2021 09.
Article in English | MEDLINE | ID: mdl-33107605

ABSTRACT

OBJECTIVES/HYPOTHESIS: Vocal fold atrophy, scar, and sulcus reduce the vibratory function of the vocal fold mucosa, which causes severe refractory dysphonia. We have reported encouraging preliminary results using an intracordal injection of basic fibroblast growth factor (bFGF) and showed improvement in phonatory parameters and voice. The present study summarizes our experience with 100 cases of stiffened vocal folds that were treated with bFGF injections. STUDY DESIGN: Retrospective chart review with Interstitial Review Board (IRB) approval. METHODS: Local injection of bFGF was performed in 100 cases of vocal fold pathology, which included 43 cases of vocal fold atrophy, 41 cases with scar, and 16 cases with sulcus. Ten micrograms of bFGF were injected into the vocal folds under topical anesthesia 4 times in each patient. Therapeutic outcomes were examined with maximum phonation time (MPT), voice handicap index-10 (VHI-10), and GRBAS scale. RESULTS: MPT, VHI-10, and GRBAS scores significantly improved in all pathology groups. An improvement on the VHI-10 greater than five points was observed in 82% of atrophy cases, 78% of scar cases, and 67% of sulcus cases. Improvement on the VHI-10 was significantly better in the atrophy group than the scar or sulcus groups. The mild/moderate cases of scar and sulcus showed better improvement than severe cases. CONCLUSIONS: The current large case series indicates positive effects of intracordal injection of bFGF for improvement of voice with no severe adverse events. The effects appeared best for cases of atrophy, while the treatment of severe scar and sulcus requires further improvement. LEVEL OF EVIDENCE: 4 Laryngoscope, 131:2059-2064, 2021.


Subject(s)
Dysphonia/drug therapy , Fibroblast Growth Factor 2/administration & dosage , Hoarseness/drug therapy , Regeneration/drug effects , Vocal Cords/drug effects , Aged , Aged, 80 and over , Atrophy/diagnosis , Atrophy/pathology , Case-Control Studies , Cicatrix/diagnosis , Cicatrix/pathology , Dysphonia/etiology , Female , Fibroblast Growth Factor 2/adverse effects , Fibroblast Growth Factor 2/therapeutic use , Hoarseness/etiology , Humans , Injections, Intralesional/methods , Laryngeal Diseases/pathology , Male , Middle Aged , Phonation/drug effects , Retrospective Studies , Treatment Outcome , Vocal Cords/pathology , Voice/drug effects
9.
J Biomed Mater Res A ; 109(3): 374-384, 2021 03.
Article in English | MEDLINE | ID: mdl-32515102

ABSTRACT

Tuneable, bioactive hydrogels present an attractive option as cell-instructive substrates for tissue regeneration. Properties mimicking the extracellular matrix at the site of injury are sought after, in particular the ability to regulate growth factors that are key to the regeneration process. This study demonstrates the successful formation of hydrogels with heparin functionalities and fibroblast growth factor-2 (FGF-2). Poly(2-hydroxyethyl methacrylate)-heparin hydrogels were capable of retaining FGF-2 by specific binding to heparin and subsequently showed sustained presentation of the growth factor to mesenchymal stromal cells (MSC). Heparin acted as stable anchoring molecules for FGF-2 on the substrate and the synergistic effect of the ensuing heparin-FGF-2 complex was evident in supporting long term cell growth. The presence of heparin during 3D scaffold formation was also found to introduce surface roughness and microporosity to the resulting hydrogels. While FGF-2 has been known to encourage MSC growth and maintain their multilineage potential, other heparin-binding ligands such as bone morphogenetic proteins are potent differentiation stimuli for MSC. Therefore preserving MSC multipotency or a push toward a differentiation pathway may be pursued by the choice of ligand applied to and bound by the heparin functionalities on the current substrate.


Subject(s)
Delayed-Action Preparations/chemistry , Fibroblast Growth Factor 2/administration & dosage , Heparin/chemistry , Hydrogels/chemistry , Cell Line , Cell Proliferation/drug effects , Fibroblast Growth Factor 2/pharmacology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects
10.
J Mater Chem B ; 8(43): 9921-9932, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33034328

ABSTRACT

Incorporating poly(lactic-co-glycolic) acid (PLGA) microparticles into human mesenchymal stem cells (hMSC) aggregates has shown promising application prospects. However, the acidic degradation products and burst release of PLGA microparticles still need to be ameliorated. In this study, the PLGA/chitosan-heparin (P/C-h) composite microparticles were successfully fabricated by integrating the double emulsion and microfluidic technology through the precise manipulation of the emulsion composition and flow rate of the two-phase in a flow-focusing chip. The P/C-h microparticles were highly monodispersed with a diameter of 23.45 ± 0.25 µm and shell-core structure of the PLGA encapsulated C-h complex, which were suitable for the fabrication of hMSC aggregates. When the mass ratio of PLGA to the C-h complex was optimized to 2 : 1, the pH of the leach liquor of P/C-h microparticles remained neutral. Compared with those of PLGA microparticles, the cytotoxicity and the initial burst release (loaded FGF-2 and VEGF) were both significantly reduced in P/C-h microparticles. Furthermore, the survival, stemness, as well as secretion and migration abilities of cells in hMSC aggregates incorporating P/C-h microparticles were also enhanced. In summary, the P/C-h composite microparticles prepared by the droplet microfluidic technique support the optimal biological and functional profile of the hMSC aggregates, which may facilitate the clinical applications of MSC-based therapy.


Subject(s)
Chitosan/chemistry , Drug Carriers/chemistry , Heparin/chemistry , Mesenchymal Stem Cells/cytology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Cell Aggregation , Cell Line , Drug Liberation , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/chemistry , Humans , Lab-On-A-Chip Devices , Mesenchymal Stem Cells/chemistry , Particle Size , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/chemistry
11.
J Control Release ; 327: 61-69, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32768629

ABSTRACT

Growth factors are required for cell proliferation and differentiation under physiological conditions but especially in the context of regenerative medicine. The time-prolonged administration of those factors has been explored using different sustained drug delivery systems. These platforms include natural materials such as bacterial inclusion bodies (IBs) that contain chaperones and other bacterial components that might favour protein release. Being successful from a functional point of view, IBs pose regulatory concerns to clinical applications because of the mentioned presence of bacterial cell components, including endotoxins. We have here explored the release and activity of the human fibroblast growth factor-2 (hFGF-2) from a novel synthetic material, namely artificial IBs. Being chemically homogenous and compliant with regulatory restrictions, we wondered if these materials would effectively release functional proteins in absence of accompanying bacterial agents. The data provided here fully supports that artificial hFGF-2 IBs act as true and efficient secretory granules and they slowly disintegrate in cell culture to promote wound healing in an in vitro wound healing model. Free from undesired bacterial components, artificial inclusion bodies show promises as delivery agents in regenerative medicine.


Subject(s)
Fibroblast Growth Factor 2 , Inclusion Bodies , Endotoxins , Fibroblast Growth Factor 2/administration & dosage , Humans , Molecular Chaperones
12.
Mater Sci Eng C Mater Biol Appl ; 115: 111123, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32600722

ABSTRACT

Anti-inflammation and angiogenesis play an essential role in wound healing. In this study, we developed a composite hydrogel dressing with stepwise delivery of diclofenac sodium (DS) and basic fibroblast growth factor (bFGF) in the inflammation stage and new tissue formation stage respectively for wound repair. Sodium alginate (SA) crosslinked by calcium ion acted as the continuous phase, and thermosensitive bFGF-loaded poly(N-isopropylacrylamide) nanogels (pNIPAM NGs, LCST1 ~33 °C) and DS-loaded p(N-isopropylacrylamide-co-acrylic acid) nanogels [p(NIPAM-co-AA) NGs, LCST2 ~40 °C] acted as the dispersed phase. The synthesized SA/bFGF@pNIPAM/DS@p(NIPAM-co-AA) hydrogel presented a desirable storage modulus of ~4500 Pa, a high water equilibrium swelling ratio of ~90, an appropriate water vapor transmission rate of ~2300 g/m2/day, and nontoxicity to human skin fibroblasts. The in vitro thermosensitive cargo delivery of this hydrogel showed that 92% of DS was sustainably delivered at 37 °C within the early three days mimicking the inflammation stage, while 80% of bFGF was controlled released at 25 °C within the later eight days mimicking new tissue formation stage. The in vivo wound healing of rats showed that this composite hydrogel presented a better healing effect with a wound contraction of 96% at 14 d, less inflammation and higher angiogenesis, than all control groups. These findings indicate SA/bFGF@pNIPAM/DS@p(NIPAM-co-AA) composite hydrogel is a potential dressing for wound repair.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Anti-Inflammatory Agents/administration & dosage , Diclofenac/administration & dosage , Fibroblast Growth Factor 2/administration & dosage , Wound Healing/drug effects , Acrylic Resins/chemistry , Alginates/chemistry , Angiogenesis Inducing Agents/chemistry , Angiogenesis Inducing Agents/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Bandages , Diclofenac/chemistry , Diclofenac/pharmacology , Disease Models, Animal , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/pharmacology , Humans , Male , Nanogels , Rats
13.
J Cell Mol Med ; 24(14): 8166-8178, 2020 07.
Article in English | MEDLINE | ID: mdl-32515141

ABSTRACT

The application of growth factors (GFs) for treating chronic spinal cord injury (SCI) has been shown to promote axonal regeneration and functional recovery. However, direct administration of GFs is limited by their rapid degradation and dilution at the injured sites. Moreover, SCI recovery is a multifactorial process that requires multiple GFs to participate in tissue regeneration. Based on these facts, controlled delivery of multiple growth factors (GFs) to lesion areas is becoming an attractive strategy for repairing SCI. Presently, we developed a GFs-based delivery system (called GFs-HP) that consisted of basic fibroblast growth factor (bFGF), nerve growth factor (NGF) and heparin-poloxamer (HP) hydrogel through self-assembly mode. This GFs-HP was a kind of thermosensitive hydrogel that was suitable for orthotopic administration in vivo. Meanwhile, a 3D porous structure of this hydrogel is commonly used to load large amounts of GFs. After single injection of GFs-HP into the lesioned spinal cord, the sustained release of NGF and bFGF from HP could significantly improve neuronal survival, axon regeneration, reactive astrogliosis suppression and locomotor recovery, when compared with the treatment of free GFs or HP. Moreover, we also revealed that these neuroprotective and neuroregenerative effects of GFs-HP were likely through activating the phosphatidylinositol 3 kinase and protein kinase B (PI3K/Akt) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signalling pathways. Overall, our work will provide an effective therapeutic strategy for SCI repair.


Subject(s)
Drug Delivery Systems , Fibroblast Growth Factor 2/administration & dosage , Heparin/chemistry , Hydrogels/chemistry , Nerve Growth Factor/administration & dosage , Poloxamer/chemistry , Spinal Cord Injuries/drug therapy , Animals , Axons/drug effects , Axons/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Immunohistochemistry , Nerve Regeneration , Phosphatidylinositol 3-Kinases/metabolism , Porosity , Proto-Oncogene Proteins c-akt/metabolism , Rats , Recovery of Function , Signal Transduction , Spinal Cord Injuries/etiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Temperature , Treatment Outcome
14.
Am J Otolaryngol ; 41(4): 102532, 2020.
Article in English | MEDLINE | ID: mdl-32409162

ABSTRACT

OBJECTIVE: The aging larynx undergoes structural changes that have functional consequences for voice production known as presbyphonia. Treatment of presbyphonia includes voice therapy and surgery. This review seeks to examine voice outcomes after surgery for presbyphonia. DATA SOURCES: Three electronic databases (PreMed, ScienceDirect, Embase) were reviewed for articles published between 1 January 1900 and 1 June 2019. REVIEW METHODS: Original English-language studies examining surgical treatment of presbyphonia in elderly patients (≥65 years) were included according to PRISMA. Two researchers independently analyzed articles. Outcome measures were extracted from and qualitatively compared across studies. RESULTS: Of the 118 articles identified, five satisfied eligibility criteria. In all studies, diagnosis of presbyphonia was based on videostroboscopy. 85 patients (61M, 21F) were evaluated. 37.6% underwent implantation thyroplasty (IT), 48.2% underwent injection augmentation (IA), 7.1% underwent both, and 7.1% underwent basic-fibroblastic growth factor (b-FGF) injection. Average patient age was 71.3 years. Average follow-up time was 5.4 months. Three months post-intervention, IT patients self-reported greater improvement in quality of life (QoL) metrics compared to IA patients. Aerodynamic measures, like mean phonation time, were significantly improved in IT and IA, but not b-FGF-injected patients. All patients experienced improvements in the auditory perception of voice three months post-intervention. CONCLUSION: Surgical modalities currently utilized for presbyphonia include IT and IA, with bFGF-injection being trialed abroad. IT patients reported enhanced QoL relative to IA and bFGF-injected patients. Overall there is a paucity of high-power, prospective studies that explore the efficacy of these modalities. Moreover, wide variability exists in reported outcomes among published studies.


Subject(s)
Laryngoplasty/methods , Voice Disorders/surgery , Age Factors , Aged , Aged, 80 and over , Aging , Auditory Perception , Female , Fibroblast Growth Factor 2/administration & dosage , Follow-Up Studies , Humans , Injections , Male , Phonation , Quality of Life , Treatment Outcome , Voice , Voice Disorders/physiopathology
15.
J Biomed Mater Res A ; 108(10): 2023-2031, 2020 10.
Article in English | MEDLINE | ID: mdl-32319183

ABSTRACT

Growth factor (GF) delivery is a common strategy for spinal cord injury repair, however, GF degradation can impede long-term therapies. GF sequestration via heparin is known to protect bioactivity after delivery. We tested two heparin modifications, methacrylated heparin and thiolated heparin, and electrospun these with methacrylated hyaluronic acid (MeHA) to form HepMAHA and HepSHHA nanofibers. For loaded conditions, MeHA, HepMAHA, and HepSHHA fibers were incubated with soluble basic fibroblast growth factor (bFGF) or nerve growth factor (NGF) and rinsed with PBS. Control groups were hydrated in PBS. L929 fibroblast proliferation was analyzed after 24 hr of culture in either growth media or bFGF-supplemented media. Dissociated chick dorsal root ganglia neurites were measured after 3 days of cell culture in serum free media (SFM) or NGF-supplemented SFM (SFM + NGF). In growth media, fibroblast proliferation was significantly increased in loaded HepMAHA (α < .05) compared to other groups. In SFM, loaded HepMAHA had the longest average neurite length compared to all other groups. In SFM + NGF, HepMAHA and HepSHHA had increased neurite lengths compared to MeHA, regardless of loading (α < .01), suggesting active sequestration of soluble NGF. HepMAHA is a promising biomaterial for sequestering released GFs in a spinal cord injury environment and will be combined with GF filled microspheres for future studies.


Subject(s)
Fibroblast Growth Factor 2/administration & dosage , Heparin/chemistry , Hyaluronic Acid/chemistry , Nanofibers/chemistry , Spinal Cord Injuries/therapy , Animals , Cell Line , Cells, Cultured , Chick Embryo , Drug Carriers/chemistry , Fibroblast Growth Factor 2/pharmacology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Mice , Spinal Cord Regeneration/drug effects
16.
ACS Appl Mater Interfaces ; 12(9): 10156-10169, 2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32027477

ABSTRACT

Burn infection is one of the commonest causes of death in severely burned patients. Developing multifunctional biological nanomaterials has a great significance for the comprehensive treatment of burn infection. In this paper, we developed a hydrogel-based nanodelivery system with antibacterial activity and skin regeneration function, which was used for photodynamic antimicrobial chemotherapy (PACT) in the treatment of burns. The treatment system is mainly composed of porphyrin photosensitizer sinoporphyrin sodium (DVDMS) and poly(lactic-co-glycolic acid) (PLGA)-encapsulated basic fibroblast growth factor (bFGF) nanospheres that are embedded in carboxymethyl chitosan (CMCS)-sodium alginate to form CSDP hybrid hydrogel. We systematically evaluated the inherent antibacterial performance, rheological properties, fluorescence imaging, and biocompatibility of the CSDP nanosystem. Under mild photoirradiation (30 J/cm2, 5 min), 10 µg/mL CSDP showed excellent antibacterial and anti-biofilm activities, which eradicated almost 99.99% of Staphylococcus aureus and multidrug-resistant (MDR) S. aureus in vitro. KEGG analysis identified that multiple signaling pathways were changed in MDR S. aureus after PACT. In the burn-infection model, CSDP-PACT successfully inhibited bacteria growth and concurrently promoted wound healing. Moreover, several regenerative factors were increased and some proinflammatory factors were reduced in the burn wounds of CSDP hydrogel treatment. These results suggest that the multifunctional CSDP hydrogel is a portable, light-triggered, antibacterial theranostic-platform and CSDP-PACT provides a promising strategy or the mechanically based synergistic treatment of burn infections.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Burns/drug therapy , Fibroblast Growth Factor 2/administration & dosage , Hydrogels/administration & dosage , Photosensitizing Agents/administration & dosage , Animals , Anti-Bacterial Agents/chemistry , Burns/microbiology , Female , Fibroblast Growth Factor 2/chemistry , Humans , Hydrogels/chemistry , Mice , Mice, Inbred BALB C , Photochemotherapy , Photosensitizing Agents/chemistry , Porphyrins/administration & dosage , Porphyrins/chemistry , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcal Infections/physiopathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/physiology , Wound Healing/drug effects
17.
Laryngoscope ; 130(1): 159-165, 2020 01.
Article in English | MEDLINE | ID: mdl-30810241

ABSTRACT

OBJECTIVES/HYPOTHESIS: Unilateral vocal fold paralysis (UVFP) induces hoarseness due to progressive atrophy of the denervated thyroarytenoid (TA) muscle. Therefore, treatments aimed at regenerating the atrophied TA muscle are required. Basic fibroblast growth factor (bFGF) is involved in muscle development and regeneration. This study aimed to elucidate the effects of bFGF injection on atrophied TA muscle. STUDY DESIGN: Animal research. METHODS: A recurrent laryngeal nerve-paralysis rat model was established, and low- (200 ng) or high-dose (2,000 ng) bFGF or saline (control) was injected into the TA muscle 28 days later. The larynges were excised on day 1, 3, 7, 14, and 28 after treatment. The cross-sectional area of the TA muscle in normal and paralyzed sides was compared, and the Ki67-positive (Ki67+ ) dividing cells, paired box 7-positive (Pax7+ ) satellite cells (SCs), and myogenic differentiation-positive (MyoD+ ) myoblasts were counted. RESULTS: The TA muscle area of animals administered high-dose bFGF increased with time and was significantly larger than that of the saline-injected controls 28 days after treatment (P < .05). The counts of Ki67+ and Pax7+ cells were the highest on day 1, whereas the MyoD+ myoblast count was highest on day 7. These results suggest that bFGF administration into the denervated TA muscles compensated for the atrophied TA muscles by inducing proliferation of SCs and their differentiation to myoblasts. CONCLUSIONS: A single injection of high-dose bFGF augmented regeneration and differentiation of the atrophied TA muscle by enhancing proliferation and differentiation of muscle SCs, suggesting its possible clinical application in humans with UVFP. LEVEL OF EVIDENCE: NA Laryngoscope, 130:159-165, 2020.


Subject(s)
Fibroblast Growth Factor 2/administration & dosage , Laryngeal Muscles , Muscular Atrophy/drug therapy , Muscular Atrophy/etiology , Vocal Cord Paralysis/complications , Animals , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Injections, Intralesional , Laryngeal Muscles/cytology , Laryngeal Muscles/physiology , Male , Rats , Rats, Sprague-Dawley , Regeneration/drug effects
18.
J Cell Mol Med ; 24(1): 1128-1140, 2020 01.
Article in English | MEDLINE | ID: mdl-31755222

ABSTRACT

Increased immature neovessels contribute to plaque growth and instability. Here, we investigated a method to establish functional and stable neovessel networks to increase plaque stability. Rabbits underwent aortic balloon injury and were divided into six groups: sham, vector and lentiviral transfection with vascular endothelial growth factor-A (VEGF)-A, fibroblast growth factor (FGF)-2, platelet-derived growth factor (PDGF)-BB and FGF-2 + PDGF-BB. Lentivirus was percutaneously injected into the media-adventitia of the abdominal aorta by intravascular ultrasound guidance, and plaque-rupture rate, plaque-vulnerability index and plaque neovessel density at the injection site were evaluated. Confocal microscopy, Prussian Blue assay, Evans Blue, immunofluorescence and transmission electron microscopy were used to assess neovessel function and pericyte coverage. To evaluate the effect of FGF-2/PDGF-BB on pericyte migration, we used the mesenchymal progenitor cell line 10T1/2 as an in vitro model. VEGF-A- and FGF-2-overexpression increased the number of immature neovessels, which caused intraplaque haemorrhage and inflammatory cell infiltration, eventually resulting in the plaque vulnerability; however, FGF-2/PDGF-BB induced mature and functional neovessels, through increased neovessel pericyte coverage. Additionally, in vitro analysis of 10T1/2 cells revealed that FGF-2/PDGF-BB induced epsin-2 expression and enhanced the VEGF receptor-2 degradation, which negatively regulated pericyte function consistent with the in vivo data. These results showed that the combination of FGF-2 and PDGF-BB promoted the function and maturation of plaque neovessels, thereby representing a novel potential treatment strategy for vulnerable plaques.


Subject(s)
Becaplermin/administration & dosage , Fibroblast Growth Factor 2/administration & dosage , Genetic Vectors/administration & dosage , Lentivirus/genetics , Neovascularization, Pathologic/prevention & control , Plaque, Atherosclerotic/therapy , Adaptor Proteins, Vesicular Transport , Animals , Becaplermin/genetics , Becaplermin/metabolism , Cell Movement , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Genetic Vectors/genetics , Male , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Phosphorylation , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Rabbits
19.
Microsurgery ; 40(3): 377-386, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31868964

ABSTRACT

BACKGROUND: The development of drug delivery systems has enabled the release of multiple bioactive molecules. The efficacy of nerve conduits coated with dual controlled release of stromal cell-derived factor-1 (SDF-1) and basic fibroblast growth factor (bFGF) for peripheral nerve regeneration was investigated. MATERIALS AND METHODS: Sixty-two C57BL6 mice were used for peripheral nerve regeneration with a nerve conduit (inner diameter, 1 mm, and length, 7 mm) and an autograft. The mice were randomized into five groups based on the different repairs of nerve defects. In the group of repair with conduits alone (n = 9), a 5-mm sciatic nerve defect was repaired by the nerve conduit. In the group of repair with conduits coated with bFGF (n = 10), SDF-1 (n = 10), and SDF-1/bFGF (n = 10), it was repaired by the nerve conduit with bFGF gelatin, SDF-1 gelatin, and SDF-1/bFGF gelatin, respectively. In the group of repair with autografts (n = 10), it was repaired by the resected nerve itself. The functional recovery, nerve regeneration, angiogenesis, and TGF-ß1 gene expression were assessed. RESULTS: In the conduits coated with SDF-1/bFGF group, the mean sciatic functional index value (-88.68 ± 10.64, p = .034) and the axon number (218.8 ± 111.1, p = .049) were significantly higher than the conduit alone group, followed by the autograft group; in addition, numerous CD34-positive cells and micro vessels were observed. TGF-ß1 gene expression relative values in the conduits with SDF-1/bFGF group at 3 days (7.99 ± 5.14, p = .049) significantly increased more than the conduits alone group. CONCLUSION: Nerve conduits coated with dual controlled release of SDF-1 and bFGF promoted peripheral nerve regeneration.


Subject(s)
Chemokine CXCL12/administration & dosage , Coated Materials, Biocompatible , Fibroblast Growth Factor 2/administration & dosage , Guided Tissue Regeneration/instrumentation , Nerve Regeneration , Peripheral Nerves/surgery , Tissue Scaffolds , Animals , Male , Mice , Mice, Inbred C57BL , Random Allocation
20.
Biomater Sci ; 7(12): 5438-5450, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31642823

ABSTRACT

It is well known that the basic fibroblast growth factor (bFGF) promotes angiogenesis after myocardial infarction (MI), but its biological functions decrease in the event of diffusion, enzymolysis, and weak binding with co-receptors in vivo. Heparan sulfate proteoglycans (HSPG) are a major component of extracellular matrices and have been shown to regulate a wide range of cellular functions and bioprocesses by acting as a co-receptor for bFGF and affecting its bioactivities. However, the influence of HSPG on the function of bFGF after myocardial infarction is unknown. Here, exogenous HSPG along with bFGF was injected into the hearts of rats to deliver the angiogenic growth factor for ischemic heart repair following induced MI. The specific binding of HSPG with bFGF protein was demonstrated, which was about 6-fold stronger than the binding of bFGF with heparin. The biological mechanisms of HSPG binding with bFGF were further studied by cell adhesion assay, and assays of bFGF and matrix metalloproteinase 2 (MMP2) activities demonstrated that HSPG enhances cell adhesion, promotes the bioactivity of bFGF in angiogenesis, and protects bFGF from enzymolysis. Our results indicate that HSPG has potential clinical utility as a delivery agent for heparin-binding growth factors. Additionally, HSPG shows high binding affinities with different ECM proteins which also help to anchor bFGF to heart tissue. Therefore, extracellular proteins that mimic the bio-scaffold of the extracellular matrix could promote the activities of bFGF to facilitate ischemic heart repair.


Subject(s)
Fibroblast Growth Factor 2/administration & dosage , Heparan Sulfate Proteoglycans/administration & dosage , Matrix Metalloproteinase 2/genetics , Myocardial Infarction/drug therapy , Animals , BALB 3T3 Cells , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Fibroblast Growth Factor 2/chemistry , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation/drug effects , Heparan Sulfate Proteoglycans/pharmacology , Mice , Myocardial Infarction/genetics , Proteolysis , Rats , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...