Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Arch Plast Surg ; 51(3): 268-274, 2024 May.
Article in English | MEDLINE | ID: mdl-38737849

ABSTRACT

Most Asians have a nose with a short columella and a low dorsum; augmentation rhinoplasty using implants is commonly performed in Asian countries to achieve a taller and more well-defined nasal dorsum. However, the current knowledge is insufficient to fully understand the various subjective desires of patients, reflect on them during surgery, or to objectively analyze the results after surgery. Advances in digital imaging technologies, such as 3D printing and 3D scanning, have transformed the medical system from hospital-centric to patient-centric throughout the medical field. In this study, we applied these techniques to rhinoplasty. First, we used virtual 3D plastic surgery software to enable surgical planning through objectified numerical calculations based on the visualized data of the patient's medical images rather than simple virtual plastic surgery. Second, the customized nasal implant was manufactured by reflecting the patient's anatomical shape and virtual 3D plastic surgery data. Taken together, we describe the surgical results of applying these rhinoplasty solutions in four patients. Our experience indicates that high fidelity and patient satisfaction can be achieved by applying these techniques.

2.
Biomater Sci ; 11(24): 7884-7896, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37906468

ABSTRACT

Traumatic spinal cord injury (SCI) leads to Wallerian degeneration and the accompanying disruption of vasculature leads to ischemia, which damages motor and sensory function. Therefore, understanding the biological environment during regeneration is essential to promote neuronal regeneration and overcome this phenomenon. The band of Büngner is a structure of an aligned Schwann cell (SC) band that guides axon elongation providing a natural recovery environment. During axon elongation, SCs promote axon elongation while migrating along neovessels (endothelial cells [ECs]). To model this, we used extrusion 3D bioprinting to develop a multi-channel conduit (MCC) using collagen for the matrix region and sacrificial alginate to make the channel. The MCC was fabricated with a structure in which SCs and ECs were longitudinally aligned to mimic the sophisticated recovering SCI conditions. Also, we produced an MCC with different numbers of channels. The aligned SCs and ECs in the 9-channel conduit (9MCC-SE) were more biocompatible and led to more proliferation than the 5-channel conduit (5MCC-SE) in vitro. Also, the 9MCC-SE resulted in a greater healing effect than the 5MCC-SE with respect to neuronal regeneration, remyelination, inflammation, and angiogenesis in vivo. The above tissue recovery results led to motor function repair. Our results show that our 9MCC-SE model represents a new therapeutic strategy for SCI.


Subject(s)
Nerve Regeneration , Spinal Cord Injuries , Humans , Endothelial Cells , Schwann Cells , Spinal Cord Injuries/therapy , Collagen , Spinal Cord
3.
Drug Chem Toxicol ; 43(4): 361-368, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31305169

ABSTRACT

A skin irritation test using in vitro reconstructed human epidermis (RhE) models was established for hazard identification of irritant chemicals in accordance with UN Globally Harmonized System of Classification and Labelling of Chemicals (GHS) category. In this study, EpiDerm™ was used to assess skin irritation by oxybenzone and N,N-diethyl-m-toluamide (DEET), which are widely used sunscreen and insect repellent components, respectively. EpiDerm™ was applied with oxybenzone and DEET, combined and sequentially with each single dose. Epidermal morphology and differentiation/proliferation were examined microscopically. Oxybenzone and sequential administration groups were determined as nonirritant with cell viability >50% and the morphology was comparable to the human epidermis. Contrastingly, the DEET and coadministration groups exhibited cell viability <50% and poor epidermal morphology. Interleukin (IL)-1α release from substance-treated EpiDerm™ increased inversely to cell viability, suggesting the pro-inflammatory reaction was initiated by DEET. CK-10, E-cadherin, Ki-67, laminin, and ceramide were identified as relevant markers to assess oxybenzone- or DEET-induced epidermal injury. In conclusion, these results may indicate to be aware of the possible skin irritation by indiscriminate use of oxybenzone and DEET without animal testing.


Subject(s)
Benzophenones/toxicity , DEET/toxicity , Epidermis/drug effects , Insect Repellents/toxicity , Irritants/toxicity , Skin Irritancy Tests , Sunscreening Agents/toxicity , Cell Survival , Dermatitis, Irritant/etiology , Epidermis/pathology , Humans
4.
Artif Cells Nanomed Biotechnol ; 47(1): 644-649, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30873886

ABSTRACT

Upon bioprinting, cells are mixed with a biomaterial to fabricate a living tissue, thus emphasizing the importance of biomaterials. The biomaterial used in this study was a bio-ink prepared using skin decellularized extracellular matrix (dECM). Skin dECM was extracted by treating the dermis with chemicals and enzymes; the basic structural and functional proteins of the ECM, including collagen, glycosaminoglycans (GAGs), bioreactive materials and growth factors, were preserved, whereas the resident cells that might cause immune rejection or inflammatory responses were removed. The bio-ink based on dECM powder, together with human dermal fibroblasts (HDFs), was loaded into the nozzle of the 3D bioprinter to create the 3D construct. This construct underwent gelation with changing temperature while its shape was maintained for 7 days. The cells showed over 90% viability and proliferation. By analysing the gene expression pattern in the cells of the construct, the skin regenerative mechanism of the bio-ink was verified. Microarray results confirmed that the gene expression related to skin morphology and development had been enhanced because the bioreactive molecules and growth factors, in addition to residual ECM in dECM, provided an optimal condition for the HDFs.


Subject(s)
Acellular Dermis , Bioprinting/methods , Extracellular Matrix/metabolism , Skin, Artificial , Tissue Engineering/methods , Animals , Cell Proliferation , Cell Survival , Extracellular Matrix/chemistry , Fibroblasts/cytology , Gene Expression Profiling , Humans , Swine
5.
Biomed Mater ; 13(1): 015014, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29155411

ABSTRACT

The appropriate porosity and pore size of barrier membranes were associated with the transportation of biomolecules required for new bone formation and angiogenesis. In this study, we fabricated three-dimensional (3D)-printed resorbable polycaprolactone (PCL) membranes with different porosities (30%, 50%, and 70%) to evaluate the effective pore size for guided bone regeneration (GBR) membranes. To analyze mechanical properties and cytocompatibility, PCL membranes prepared using extrusion-based 3D printing technology were compared in dry and wet conditions and tested in vitro. The proliferation rates and pattern of fibroblasts and preosteoblasts on PCL membranes with different porosities were determined using a cell counting kit-8 assay and scanning electron microscopy. PCL membrane porosity did not affect cell proliferation, but osteogenic differentiation and mechanical properties were increased with lower porosity (30%) on day 14 (p < 0.001). Similar results were found in an in vivo calvarial defect model; new bone formation was significantly higher in PCL membranes with lower porosity (p < 0.001). These results indicate that 3D-printed PCL with 30% porosity (130 µm pore size) is an excellent pore size for GBR membranes.


Subject(s)
Biocompatible Materials/pharmacology , Bone Regeneration/drug effects , Osteogenesis/drug effects , Polyesters/chemistry , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Durapatite/pharmacology , Male , Mice , Microscopy, Electron, Scanning , NIH 3T3 Cells , Porosity , Printing, Three-Dimensional , Rabbits , Stress, Mechanical , Tissue Engineering/methods , Tissue Scaffolds , X-Ray Microtomography
6.
Sci Rep ; 7(1): 8624, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819137

ABSTRACT

Three-dimensional (3D) cell printing systems allow the controlled and precise deposition of multiple cells in 3D constructs. Hydrogel materials have been used extensively as printable bioinks owing to their ability to safely encapsulate living cells. However, hydrogel-based bioinks have drawbacks for cell printing, e.g. inappropriate crosslinking and liquid-like rheological properties, which hinder precise 3D shaping. Therefore, in this study, we investigated the influence of various factors (e.g. bioink concentration, viscosity, and extent of crosslinking) on cell printing and established a new 3D cell printing system equipped with heating modules for the precise stacking of decellularized extracellular matrix (dECM)-based 3D cell-laden constructs. Because the pH-adjusted bioink isolated from native tissue is safely gelled at 37 °C, our heating system facilitated the precise stacking of dECM bioinks by enabling simultaneous gelation during printing. We observed greater printability compared with that of a non-heating system. These results were confirmed by mechanical testing and 3D construct stacking analyses. We also confirmed that our heating system did not elicit negative effects, such as cell death, in the printed cells. Conclusively, these results hold promise for the application of 3D bioprinting to tissue engineering and drug development.


Subject(s)
Bioengineering/methods , Extracellular Matrix/metabolism , Hot Temperature , Printing, Three-Dimensional , Animals , Cell Survival , Heating , Hydrogen-Ion Concentration , Microscopy, Electron, Scanning , Rheology , Skin/cytology , Skin/metabolism , Skin/ultrastructure , Swine
7.
Int J Mol Sci ; 18(5)2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28441338

ABSTRACT

This study was conducted to compare 3D-printed polycaprolactone (PCL) and polycaprolactone/ß-tricalcium phosphate (PCL/ß-TCP) membranes with a conventional commercial collagen membrane in terms of their abilities to facilitate guided bone regeneration (GBR). Fabricated membranes were tested for dry and wet mechanical properties. Fibroblasts and preosteoblasts were seeded into the membranes and rates and patterns of proliferation were analyzed using a kit-8 assay and by scanning electron microscopy. Osteogenic differentiation was verified by alizarin red S and alkaline phosphatase (ALP) staining. An in vivo experiment was performed using an alveolar bone defect beagle model, in which defects in three dogs were covered with different membranes. CT and histological analyses at eight weeks after surgery revealed that 3D-printed PCL/ß-TCP membranes were more effective than 3D-printed PCL, and substantially better than conventional collagen membranes in terms of biocompatibility and bone regeneration and, thus, at facilitating GBR.


Subject(s)
Biocompatible Materials/chemistry , Bone Regeneration/physiology , Bone and Bones/physiology , Calcium Phosphates/chemistry , Polyesters/chemistry , Printing, Three-Dimensional , Animals , Biocompatible Materials/pharmacology , Biocompatible Materials/therapeutic use , Bone Regeneration/drug effects , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Collagen/chemistry , Dogs , Fractures, Bone/pathology , Fractures, Bone/therapy , Membranes, Artificial , Mice , Microscopy, Electron, Scanning , Osteogenesis/drug effects , X-Ray Microtomography
8.
Laryngoscope ; 127(5): 1036-1043, 2017 05.
Article in English | MEDLINE | ID: mdl-28150412

ABSTRACT

OBJECTIVES: Polycaprolactone (PCL) is an U.S. Food and Drug Administration-approved synthetic biodegradable polymer and is easily fabricated into three-dimensional (3D) structures. In this study, the 3D-printed PCL implant for nasal augmentation was further evaluated for its suitability for nasal surgeries such as septoplasty and rhinoplasty. METHODS: Ten New Zealand White rabbits were included and divided into study and sham groups (7 and 3, respectively). A lateral incision was made on the nasal dorsum and a pocket formed in the subperichondrial plane between the upper lateral cartilage and nasal septum. Polycaprolactone was fabricated based on 3D printing technology into a 0.8 × 0.8-cm rectangular shape for use as a nasal implant. The material was inserted as a septal extension graft and sutured with alar cartilage for nasal reshaping. The implants were harvested 4, 8, and 12 weeks after implantation and evaluated by gross morphological assessment and histological examination. RESULTS: The initial shape of the implant was unchanged in all cases, and no definitive postoperative complications were seen over the 3-month period. Gross morphological evaluation confirmed that implants remained in their initial location without migration or extrusion. Histologic evaluations showed that the implant architectures were maintained with excellent fibrovascular ingrowth and minimal inflammatory reactions. CONCLUSION: Polycaprolactone can be used for nasal reconstruction such as nasal augmentation. Polycaprolactone is easy to work with and will avoid the increased operative time and morbidity associated with autograft harvesting. Therefore, PCL implants designed by 3D printing can serve as clinically biocompatible materials in craniofacial reconstruction in the future. LEVEL OF EVIDENCE: NA. Laryngoscope, 127:1036-1043, 2017.


Subject(s)
Polyesters/pharmacology , Printing, Three-Dimensional , Prostheses and Implants , Rhinoplasty/instrumentation , Animals , Models, Animal , Nasal Septum/surgery , Prosthesis Design , Rabbits
9.
J Mol Cell Cardiol ; 97: 266-77, 2016 08.
Article in English | MEDLINE | ID: mdl-27266389

ABSTRACT

BACKGROUND: Peroxisome proliferator-activated receptor (PPAR)-δ is a nuclear receptor regulating cell metabolism. The role of PPAR-δ in late endothelial progenitor cells (EPCs) has not been fully elucidated. We aim to understand the effects of PPAR-δ activation on late EPC and to reveal the underlying mechanism. METHODS AND RESULTS: Treatment with a highly selective PPAR-δ agonist (GW501516) induced proliferation of late EPCs and enhanced their vasculogenic potential. Search for the target molecule of PPAR-δ activation revealed endothelial differentiation gene (Edg)-2. Chromatin immunoprecipitation and promoter assays demonstrated that Edg-2 gene was specifically induced by PPAR-δ through direct transcriptional activation. Lysophosphatidic acid (LPA), an Edg ligand, mimicked the pro-vasculogenic effects of GW501516 in late EPCs whereas Edg antagonist (Ki16425) blocked these effects. Edg-2 is a membrane receptor for LPA which is a major growth factor from activated platelets. Thus, the interaction between platelets and late EPCs via the LPA-Edg-2 axis was assessed. Platelet supernatant boosted the pro-vasculogenic effects of GW501516, which was reversed by antagonist to PPAR-δ (GSK0660) or Edg (Ki16425). Both of in vivo Matrigel plug model and mouse skin punch-wound model demonstrated that the combination of platelets and PPAR-δ-activated late EPCs synergistically enhanced vascular regeneration. CONCLUSIONS: There exists a synergistic interaction between human platelets and late EPCs leading to vascular regeneration. This interaction consists of LPA from platelets and its receptor Edg-2 on the surface of EPCs and can be potentiated by PPAR-δ activation in EPCs. A PPAR-δ agonist is a good candidate to achieve vasculogenesis for ischemic vascular disease.


Subject(s)
Blood Platelets/metabolism , Endothelial Progenitor Cells/metabolism , Lysophospholipids/metabolism , PPAR delta/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Base Sequence , Binding Sites , Cell Communication , Consensus Sequence , Gene Expression Regulation , Humans , Lysophospholipids/pharmacology , Neovascularization, Physiologic , Protein Binding , Receptors, Lysophosphatidic Acid/chemistry , Receptors, Lysophosphatidic Acid/genetics , Transcriptional Activation , Wound Healing
10.
Sci Rep ; 5: 16523, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26563778

ABSTRACT

Skeletal muscle regeneration occurs continuously to repair muscle damage incurred during normal activity and in chronic disease or injury. Herein, we report that A-kinase anchoring protein 6 (AKAP6) is important for skeletal myoblast differentiation and muscle regeneration. Compared with unstimulated skeletal myoblasts that underwent proliferation, differentiated cells show significant stimulation of AKAP6 expression. AKAP6 knockdown with siRNA effectively halts the formation of myotubes and decreases the expression of the differentiation markers myogenin and myosin heavy chain. When shAKAP6-lentivirus is delivered to mice with cardiotoxin (CTX)-induced muscle injury, muscle regeneration is impaired compared with that of mice injected with control shMock-lentivirus. The motor functions of mice infected with shAKAP6-lentivirus (CTX+shAK6) are significantly worse than those of mice infected with shMock-lentivirus (CTX+shMock). Mechanistic analysis showed that AKAP6 promotes myogenin expression through myocyte enhancer factor 2A (MEF2A). Notably, myogenin increases AKAP6 expression as well. The results of chromatin immunoprecipitation and luciferase assays showed that myogenin binds to an E-box site on the AKAP6 promoter. Taken together, our findings demonstrate a novel interplay between AKAP6 and myogenin, and we suggest that AKAP6 is an important regulator of myoblast differentiation, myotube formation, and muscle regeneration.


Subject(s)
A Kinase Anchor Proteins/genetics , Cell Differentiation/genetics , Muscle Development/genetics , Myoblasts/metabolism , Myogenin/genetics , Regeneration/genetics , A Kinase Anchor Proteins/metabolism , Animals , Cell Line , Cell Proliferation/genetics , Cells, Cultured , HEK293 Cells , Humans , Immunoblotting , Mice, Inbred C57BL , Microscopy, Confocal , Muscle Development/physiology , Myoblasts/cytology , Myogenin/metabolism , RNA Interference , Regeneration/physiology , Reverse Transcriptase Polymerase Chain Reaction
11.
Mol Ther ; 21(9): 1767-77, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23760445

ABSTRACT

Ischemia/reperfusion (I/R) injury to myocardium induces death of cardiomyocytes and destroys the vasculature, leading to cardiac fibrosis that is mainly mediated by the transdifferentiation of fibroblasts to myofibroblasts and the collagen deposition. Snail involvement in fibrosis is well known; however, the contribution of Snail to cardiac fibrosis during I/R injury and its underlying mechanisms have not been defined. We showed that I/R injury to mouse hearts significantly increases the expression of Snail. An in vitro hypoxia/reoxygenation (Hy/Reoxy) experiment showed that the cell source of Snail induction is endothelial cells rather than cardiac fibroblasts (cFibroblasts) or cardiomyoblasts. When Snail was overexpressed in endothelial cells, they underwent endothelial-to-mesenchymal transition (EndMT) but showed very poor capacity for collagen synthesis. Instead, reoxygenation- or Snail overexpression-mediated EndMT-like cells noticeably stimulated transdifferentiation of fibroblasts to myofibroblasts via secretion of connective tissue growth factor (CTGF). The injection of a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist, a selective Snail inhibitor, remarkably suppressed collagen deposition and cardiac fibrosis in mouse I/R injury, and significantly improved cardiac function and reduced Snail and CTGF expression in vivo. Our findings suggested a new mechanism of cell-to-cell communication between EndMT-like cells and fibroblasts for fibrosis induction and implicated Snail as a potential target molecule in cardiac fibrosis after I/R injury.


Subject(s)
Connective Tissue Growth Factor/metabolism , Endothelial Cells/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/therapy , Myocardium/metabolism , Myocardium/pathology , Myofibroblasts/metabolism , Transcription Factors/metabolism , Animals , Cell Transdifferentiation , Cells, Cultured , Fibrosis , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/pathology , PPAR gamma/agonists , Rats , Snail Family Transcription Factors
12.
Mol Ther ; 20(9): 1750-66, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22713697

ABSTRACT

Loss of cardiomyocytes impairs cardiac function after myocardial infarction (MI). Recent studies suggest that cardiac stem/progenitor cells could repair the damaged heart. However, cardiac progenitor cells are difficult to maintain in terms of purity and multipotency when propagated in two-dimensional culture systems. Here, we investigated a new strategy that enhances potency and enriches progenitor cells. We applied the repeated sphere formation strategy (cardiac explant → primary cardiosphere (CS) formation → sphere-derived cells (SDCs) in adherent culture condition → secondary CS formation by three-dimensional culture). Cells in secondary CS showed higher differentiation potentials than SDCs. When transplanted into the infarcted myocardium, secondary CSs engrafted robustly, improved left ventricular (LV) dysfunction, and reduced infarct sizes more than SDCs did. In addition to the cardiovascular differentiation of transplanted secondary CSs, robust vascular endothelial growth factor (VEGF) synthesis and secretion enhanced neovascularization in the infarcted myocardium. Microarray pathway analysis and blocking experiments using E-selectin knock-out hearts, specific chemicals, and small interfering RNAs (siRNAs) for each pathway revealed that E-selectin was indispensable to sphere initiation and ERK/Sp1/VEGF autoparacrine loop was responsible for sphere maturation. These results provide a simple strategy for enhancing cellular potency for cardiac repair. Furthermore, this strategy may be implemented to other types of stem/progenitor cell-based therapy.


Subject(s)
Cell Culture Techniques , Myocardial Infarction/therapy , Stem Cell Transplantation , Stem Cells/cytology , Ventricular Dysfunction, Left/therapy , Animals , Cell Differentiation , E-Selectin/genetics , E-Selectin/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression , Humans , Immunoglobulins/genetics , Immunoglobulins/metabolism , Male , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/genetics , Signal Transduction , Stem Cells/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
13.
Mol Med ; 17(9-10): 1095-106, 2011.
Article in English | MEDLINE | ID: mdl-21738954

ABSTRACT

Early reperfusion after myocardial ischemia that is essential for tissue salvage also causes myocardial and vascular injury. Cardioprotection during reperfusion therapy is an essential aspect of treating myocardial infarction. Angiopoietin-1 is an endothelial-specific angiogenic factor. The potential effects of angiopoietin-1 on cardiomyocytes and vascular cells undergoing reperfusion have not been investigated. We propose a protective mechanism whereby angiopoietin-1 increases the integrity of the endothelial lining and exerts a direct survival effect on cardiomyocytes under myocardial ischemia followed by reperfusion. First, we found that angiopoietin-1 prevents vascular leakage through regulating vascular endothelial (VE)-cadherin phosphorylation. The membrane expression of VE-cadherin was markedly decreased on hypoxia/reoxygenation but was restored by angiopoietin-1 treatment. Interestingly, these effects were mediated by the facilitated binding between SH2 domain-containing tyrosine phosphatase (SHP2) or receptor protein tyrosine phosphatase µ (PTPµ) and VE-cadherin, leading to dephosphorylation of VE-cadherin. siRNA against SHP2 or PTPµ abolished the effect of angiopoietin-1 on VE-cadherin dephosphorylation and thereby decreased levels of membrane-localized VE-cadherin. Second, we found that angiopoietin-1 prevented cardiomyocyte death, although cardiomyocytes lack the angiopoietin-1 receptor Tie2. Angiopoietin-1 increased cardiomyocyte survival through integrin-ß1-mediated extracellular signal-regulated kinase (ERK) phosphorylation, which inhibited caspase-9 through phosphorylation at Thr¹²5 and subsequently reduced active caspase-3. Neutralizing antibody against integrin-ß1 blocked these protective effects. In a mouse myocardial ischemia/reperfusion model, angiopoietin-1 enhanced cardiac function and reduction in left ventricular-end systolic dimension (LV-ESD) and left ventricular-end diastolic dimension (LV-EDD) with an increase in ejection fraction (EF) and fractional shortening (FS). Our findings suggest the novel cardioprotective mechanisms of angiopoietin-1 that are achieved by reducing both vascular leakage and cardiomyocyte death after ischemia/reperfusion injury.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Caspase 9/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Integrin beta1/metabolism , Myocardial Reperfusion Injury/prevention & control , Recombinant Fusion Proteins/pharmacology , Animals , Animals, Newborn , Antigens, CD/genetics , Blotting, Western , Cadherins/genetics , Capillary Permeability/drug effects , Cell Hypoxia , Cells, Cultured , Heart/drug effects , Heart/physiopathology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Oxygen/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , RNA Interference , Rats , Rats, Inbred F344
14.
Cell Transplant ; 18(10): 1123-35, 2009.
Article in English | MEDLINE | ID: mdl-19650969

ABSTRACT

Transplantation of autologous skeletal myoblasts (SMBs) is a potential therapeutic approach for myocardial infarction. However, their clinical efficacy and safety is still controversial. Electrical coupling through gap junction between SMBs and host myocardium is essential for synchronized contraction and electrical stability. Here, we investigated the effect of heart beat-simulating environment, oscillating pressure, on the expression of connexin43 in two types of SMBs from rat and mouse. We found that connexin43 is markedly decreased under ischemia-mimicking conditions such as serum starvation and hypoxia (1% O(2)) in rat primary cultured SMBs and mouse C2C12 SMB cell line. Interestingly, the decrease of connexin43 expression under serum starvation was attenuated by oscillating pressure. Oscillating pressure treatment increased the expression of connexin43 twofold through AP-1 stimulation, which was blocked by PD98059, ERK inhibitor. In coculture of cardiomyocytes and C2C12, pressure-treated C2C12 and cardiomyocytes were able to form functional gap junction, which was demonstrated by both calcein-AM dye transfer assay and measurement of simultaneous contraction. In rat myocardial infarction model, transplantation of SMBs pretreated with oscillating pressure resulted in lesser ventricular dilatation and better systolic function than transplantation of untreated SMBs and control group. These results suggested that application of oscillating pressure on SMBs before transplantation may be useful to promote therapeutic efficacy for myocardial infarction by enhancing gap junction formation between transplanted and host cells.


Subject(s)
Connexin 43/metabolism , Myoblasts, Skeletal/transplantation , Myocardial Infarction/therapy , Animals , Cell Hypoxia , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Gap Junctions/physiology , Mice , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Pressure , Rats , Transcription Factor AP-1/metabolism , Up-Regulation
15.
Stem Cells ; 26(7): 1901-12, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403756

ABSTRACT

Stem cell transplantation in acute myocardial infarction (AMI) has emerged as a promising therapeutic option. We evaluated the impact of AMI on mesenchymal stem cell (MSC) differentiation into cardiomyocyte lineage. Cord blood-derived human MSCs were exposed to in vitro conditions simulating in vivo environments of the beating heart with acute ischemia, as follows: (a) myocardial proteins or serum obtained from sham-operated rats, and (b) myocardial proteins or serum from AMI rats, with or without application of oscillating pressure. Expression of cardiac-specific markers on MSCs was greatly induced by the infarcted myocardial proteins, compared with the normal proteins. It was also induced by application of oscillating pressure to MSCs. Treatment of MSCs with infarcted myocardial proteins and oscillating pressure greatly augmented expression of cardiac-specific genes. Such expression was blocked by inhibitor of transforming growth factor beta(1) (TGF-beta(1)) or bone morphogenetic protein-2 (BMP-2). In vitro cellular and electrophysiologic experiments showed that these differentiated MSCs expressing cardiomyocyte-specific markers were able to make a coupling with cardiomyocytes but not to selfbeat. The pathophysiologic significance of in vitro results was confirmed using the rat AMI model. The protein amount of TGF-beta(1) and BMP-2 in myocardium of AMI was significantly higher than that in normal myocardium. When MSCs were transplanted to the heart and analyzed 8 weeks later, they expressed cardiomyocyte-specific markers, leading to improved cardiac function. These in vitro and in vivo results suggest that infarct-related biological and physical factors in AMI induce commitment of MSCs to cardiomyocyte-like cells through TGF-beta/BMP-2 pathways.


Subject(s)
Mesenchymal Stem Cells/cytology , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Stem Cells/cytology , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/metabolism , Calcium/metabolism , Cell Differentiation , Gap Junctions/metabolism , Humans , Oscillometry , Phenotype , Rats , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Umbilical Veins/metabolism
16.
Arterioscler Thromb Vasc Biol ; 28(2): 302-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18063811

ABSTRACT

BACKGROUND: The forkhead factor, FOXO3a, is known to induce apoptosis in endothelial cells (ECs). However, its effects on extracellular matrices (ECM), which are important in EC survival, remained unknown. Here, we evaluated the role of FOXO3a on EC-ECM interaction. METHODS AND RESULTS: Constitutively active FOXO3a was transduced to human umbilical vein endothelial cells by adenoviral vector (Ad-TM-FOXO3a). Ad-TM-FOXO3a transfection led to dehiscence of ECs from fibronectin-coated plates, resulting in anoikis, which was significantly reversed by matrix metalloproteinase (MMP) inhibitor, GM6001. FOXO3a increased the expression of MMP-3 (stromelysin-1) but decreased the expression of tissue inhibitors of metalloproteinases-1 (TIMP-1), which was associated with increased MMP enzymatic activity in zymography. Pathophysiologic conditions such as serum starvation or heat shock also induced activation of endogenous FOXO3a, leading to activation of MMP-3 and apoptosis, which was reversed by GM6001. Delivery of Ad-TM-FOXO3a to the intraluminal surface in vivo led to EC denudation, disrupted vascular integrity, and impaired endothelium-dependent vasorelaxation. CONCLUSIONS: Activation of MMPs and possible ECM disruption represent novel mechanisms of FOXO3a-mediated apoptosis in ECs.


Subject(s)
Apoptosis/physiology , Endothelial Cells/physiology , Forkhead Transcription Factors/physiology , Matrix Metalloproteinase 3/metabolism , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Anoikis/physiology , Carotid Arteries/physiology , Cell Adhesion/physiology , Cells, Cultured , Enzyme Activation , Extracellular Matrix/physiology , Forkhead Box Protein O3 , Humans , Rabbits , Umbilical Veins/cytology
17.
Biochem Biophys Res Commun ; 293(4): 1248-53, 2002 May 17.
Article in English | MEDLINE | ID: mdl-12054510

ABSTRACT

Cellular response to oxidative stress is a complex process that is often connected to cell cycle regulation. The present study examines the effect of H(2)O(2) on cell cycle regulation and involvement of reactive oxygen species (ROS) in these H(2)O(2)-induced responses in a p53-deficient human lung carcinoma cell line, H1299. Treatment of the cells with H(2)O(2) caused a G2/M phase arrest. Among the redox-sensitive transcription factors, NF-kappaB and AP-1, we found that only AP-1 was activated by 200 microM H(2)O(2) in human lung cells. Furthermore, electrophoretic mobility shift assays revealed that H(2)O(2) enhanced the DNA binding of AP-1 to a putative AP-1 binding element (TGAGGAA) in the p21(WAF1/CIP1) promoter region (between -2203 and -2197 nucleotides upstream of the transcription initiation site). An increase in c-Jun phosphorylation by ERK was also found to accompany the increased AP-1 activity as detected by Western blot. PD98059, a specific inhibitor of MEK, diminished H(2)O(2)-induced phosphorylation of c-Jun and DNA binding activity of AP-1, decreased expression of p21(WAF1/CIP1), and released the cells from G2/M arrest. Taken together, these results revealed a novel AP-1 binding site in the promoter region of p21(WAF1/CIP1) and a possible cell cycle regulation mechanism mediated by activation of a redox-dependent ERK signaling pathway.


Subject(s)
Cyclins/metabolism , G2 Phase/drug effects , Genes, p53 , Hydrogen Peroxide/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mitosis/drug effects , Transcription Factor AP-1/metabolism , Binding Sites , Blotting, Western , Cell Cycle , Cell Nucleus/metabolism , Cell Separation , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Flow Cytometry , Humans , Luciferases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins c-jun/metabolism , Reactive Oxygen Species , Signal Transduction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...