Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Cytotherapy ; 25(9): 946-955, 2023 09.
Article in English | MEDLINE | ID: mdl-37354151

ABSTRACT

BACKGROUND AIMS: While distraction osteogenesis (DO) achieves substantial bone regeneration, prolonged fixation may lead to infections. Existing stem cell and physical therapies have limitations, requiring the development of novel therapeutic approaches. Here, we evaluated high-mobility group box 1 (HMGB1) as a novel therapeutic target for DO treatment. METHODS: Micro-computed tomography (Micro-CT) analysis and histological staining of samples obtained from tibial DO model mice was performed. Transwell migration, wound healing, and proliferation assays were also performed on cultured human mesenchymal stem cells (hMSCs) and human umbilival vein endothelial cells (HUVECs). Tube formation assay was performed on HUVECs, whereas osteogenic differentiation assay was performed on hMSCs. RESULTS: Micro-CT analysis and histological staining of mouse samples revealed that HMGB1 promotes bone regeneration during DO via the recruitment of PDGFRα and Sca-1 positve (PαS+) cells and endothelial progenitor cells. Furthermore, HMGB1 accelerated angiogenesis during DO, promoted the migration and osteogenic differentiation of hMSCs as well as the proliferation, migration and angiogenesis of HUVECs in vitro. CONCLUSIONS: Our findings suggest that HMGB1 has a positive influence on endogenous stem/progenitor cells, representing a novel therapeutic target for the acceleration of DO-driven bone regeneration.


Subject(s)
HMGB1 Protein , Mesenchymal Stem Cells , Osteogenesis, Distraction , Humans , Mice , Animals , Osteogenesis , Osteogenesis, Distraction/methods , X-Ray Microtomography , Wound Healing , Cell Differentiation , Bone Regeneration , Stem Cells , Human Umbilical Vein Endothelial Cells , Cells, Cultured
2.
Nagoya J Med Sci ; 85(1): 70-78, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36923624

ABSTRACT

This study was conducted to evaluate the postsurgical stability of Le Fort I osteotomy using zygomatic buttress internal fixation alone with no piriform aperture internal fixation. Patients with maxillary retrognathia and mandibular prognathism underwent the Le Fort I osteotomy with a bilateral sagittal split ramus osteotomy. In group I, fixation was accomplished using titanium plate and screws placed at the piriform aperture and the zygomatic buttress (4 plates). In group II, fixation was accomplished using titanium plate and screws placed at the zygomatic buttress (2 plates). Lateral cephalometric radiographs were taken preoperatively (T1), immediately after surgery (T2), and at 6 months to 1 year (T3) to evaluate skeletal movement. In total, 32 patients were included in this study. None of the patients had wound infection, dehiscence, bone fragment instability, and long-term malocclusion. Regarding point A and the posterior nasal spine (PNS), vertical and horizontal relapse in groups I and II did not differ significantly. In most hospitals, the maxilla was fixed using four plates (piriform aperture and zygomatic buttress); however, within the limitations of the study, the choice of the number of plates for osteosynthesis following Le Fort I osteotomy and repositioning of the maxilla can be left to the discretion of the surgeon without putting the patients at risk for increased relapse by careful intraoperative management.


Subject(s)
Osteotomy, Le Fort , Titanium , Humans , Bone Plates , Maxilla/diagnostic imaging , Maxilla/surgery , Recurrence
3.
Cytotherapy ; 22(10): 543-551, 2020 10.
Article in English | MEDLINE | ID: mdl-32798177

ABSTRACT

BACKGROUND AIMS: When cells are exposed to stresses such as mechanical stimuli, they release growth factors and adapt to the surrounding environment H ere, we demonstrated that mechanical stimulation during culture affects the production of osteogenic and angiogenic factors. METHODS: Human bone marrow derived mesenchymal stromal cells (hMSCs) and human periodontal ligament fibroblasts (HPLFs ) were cultured under cyclic stretch stimulation for 24 h. Collected of the cells and conditioned media (CM), the gene and protein expression levels of osteogenic and angiogenic factors were evaluated. CM was also evaluated for angiogenic activity and calc ification ability. In in vivo study, CM was administered to a mouse calvarial defect model and histologically and radiologically evaluated. RESULTS: Quantitative real time polymerase chain reaction results showed that the expression of bone morphogenetic pro tein 2, 4 (BMP 2, 4), vascular endothelial growth factor A (VEGF A), and platelet derived growth factor AA (PDGF AA) was upregulated in the cyclic stretch stimulation group in comparison with the non stretch group in each cell type. Enzyme linked immunosor bent assay results revealed that the expression of BMP 2,4, VEGF A was upregulated in the cyclic stretch group in comparison with the non stretch group in each cell type. Only HPLFs showed significant difference in PDGF AA expression between the cyclic str etch and the non stretch group. Tube formation assay and Alizarin Red S staining results showed that angiogenic activity and calcification ability of CM was upregulated in the cyclic stretch stimulation group in comparison with the non stretch group in eac h cell type. CM was administered to the mouse calvarial defect model. Histological and radiological examination showed that the bone healing was promoted by CM from the cyclic stretch culture group. Immunohistological staining revealed that CM from cyclic stretch group have greater angiogenic effect than CM from the non stretch group. CONCLUSIONS: These results indicate that osteogenesis was promoted by CM obtained under cyclic stretch stimulation through the increase of angiogenesis in the mouse calvarial defect model.


Subject(s)
Culture Media, Conditioned/pharmacology , Fibroblasts/metabolism , Mesenchymal Stem Cells/metabolism , Periodontal Ligament/cytology , Skull/pathology , Stress, Mechanical , Wound Healing/drug effects , Animals , Bone Morphogenetic Protein 2/metabolism , Calcification, Physiologic/drug effects , Calcification, Physiologic/genetics , Cell Differentiation/drug effects , Fibroblasts/drug effects , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mesenchymal Stem Cells/drug effects , Mice, Inbred ICR , Neovascularization, Physiologic/genetics , Osteogenesis/drug effects , Osteogenesis/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
J Biomed Mater Res A ; 108(6): 1369-1379, 2020 06.
Article in English | MEDLINE | ID: mdl-32107841

ABSTRACT

Collagen 1 (COL1) and fibronectin (FN) are extracellular matrix proteins that contribute in cell activity and involve in regulating dental pulp cells (DPCs). The purpose of this study was to investigate the effect of COL1 and FN on the behavior of DPCs. Here, DPCs were grown under three different conditions: COL1 coating, FN coating, and control group without coating. The proliferation and differentiation of DPCs were investigated. DPCs in osteogenic media were able to differentiate into osteoblastic phenotype. The morphological analysis revealed no obvious difference on the shape of cells. Cells had spread well on both coated and noncoated culture plates with slightly more spreading in the coated plates after 24 hr. The MTT analysis did not demonstrate a significant difference at 1 and 3 hr among the groups, but interestingly, the analysis disclosed more cells on the coated plates after longer cultures, which indicated a higher proliferative capacity in response to COL1 and FN. RT-PCR, Western Blotting and mineralization assays did not reveal significant differences between the coated and noncoated surfaces in relation to osteogenic differential potential. Our data suggested that the surface coating of COL1 and FN were able to promote cellular proliferation and the osteogenic differentiation tendency of DPCs was also observed in vitro.


Subject(s)
Coated Materials, Biocompatible/pharmacology , Collagen Type I/pharmacology , Dental Pulp/drug effects , Fibronectins/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dental Pulp/cytology , Humans , Osteogenesis/drug effects
5.
J Vis Exp ; (141)2018 11 14.
Article in English | MEDLINE | ID: mdl-30507900

ABSTRACT

Distraction osteogenesis (DO) is a surgical procedure that involves skeletal tissue regeneration without cell transplantation. A DO model consists of the following three phases: the latency phase after osteotomy and placement of the external distractor; the distraction phase, wherein the separated bone ends are gradually and continuously distracted; and the consolidation phase. This custom-made distractor used for DO is comprised of two incomplete acrylic resin rings and an expansion screw. The process was initiated by making a mold with silicone impression material and then creating the custom-made distractor. Dental resin was poured into the formwork made of silicone impression material, and it was allowed to polymerize to create the incomplete resin rings required for the custom-made distractor. These rings were fixed with an expansion screw using transparent resin. The custom-made distractor created via this approach was attached to the tibia of mice. The tibia was fixed to the device using one pair of 25 G needles proximally, one pair of 27 G needles distally, and acrylic resin. After a latency period of 5 days, distraction was initiated at a rate of 0.2 mm/12 h. The lengthening was continued for 8 days, resulting in a total gap of 3.2 mm. The mice were sacrificed 4 weeks after distraction. Bone formation in the distraction gap was confirmed using both radiography and histology.


Subject(s)
Osteogenesis, Distraction/methods , Tibia/surgery , Animals , Bone Screws , Disease Models, Animal , Male , Mice , Osteogenesis , Osteogenesis, Distraction/instrumentation , Osteotomy , Radiography , Tibia/diagnostic imaging , Tibia/physiology
6.
Bone ; 105: 42-49, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28802681

ABSTRACT

Achondroplasia (ACH) is one of the most common short-limbed skeletal dysplasias caused by gain-of-function mutations in the fibroblast growth factor receptors 3 (FGFR3) gene. Distraction osteogenesis (DO) is a treatment option for short stature in ACH in some countries. Although the patients with ACH usually show faster healing in DO, details of the newly formed bone have not been examined. We have developed a mouse model of DO and analyzed new bone regenerates of the transgenic mice with ACH (Fgfr3ach mice) histologically and morphologically. We established two kinds of DO protocols, the short-DO consisted of 5days of latency period followed by 5days of distraction with a rate of 0.4mm per 24h, and the long-DO consisted of the same latency period followed by 7days of distraction with a rate of 0.3mm per 12h. The callus formation was evaluated radiologically by bone fill score and quantified by micro-CT scan in both protocols. The histomorphometric analysis was performed in the short-DO protocol by various stainings, including Villanueva Goldner, Safranin-O/Fast green, tartrate-resistant acid phosphatase, and type X collagen. Bone fill scores were significantly higher in Fgfr3ach mice than in wild-type mice in both protocols. The individual bone parameters, including bone volume and bone volume/tissue volume, were also significantly higher in Fgfr3ach mice than in wild-type mice in both protocols. The numbers of osteoblasts, as well as osteoclasts, around the trabecular bone were increased in Fgfr3ach mice. Cartilaginous tissues of the distraction region rapidly disappeared in Fgfr3ach mice compared to wild-type mice during the consolidation phase. Similarly, type X collagen-positive cells were markedly decreased in Fgfr3ach mice during the same period. Fgfr3ach mice exhibited accelerated bone regeneration after DO. Accelerated endochondral ossification could contribute to faster healing in Fgfr3ach mice.


Subject(s)
Osteogenesis, Distraction , Osteogenesis , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Animals , Bone and Bones/metabolism , Bony Callus/pathology , Cell Count , Female , Imaging, Three-Dimensional , Mice , Models, Animal , Organ Size , Osteoblasts/metabolism , Osteoclasts/metabolism , Tomography, X-Ray Computed
7.
J Tissue Eng Regen Med ; 11(7): 2116-2126, 2017 07.
Article in English | MEDLINE | ID: mdl-26612624

ABSTRACT

Distraction osteogenesis (DO) is a surgical procedure used to correct various skeletal disorders. Improving the technique by reducing the healing time would be of clinical relevance. The aim of this study was to determine the angiogenic and regenerative potential of conditioned media (CMs) collected from human dental pulp cells (hDPCs) grown under different culture conditions. CM collected from cells under hypoxia was used to improve bone healing and the DO procedure in vivo. The angiogenic potentials of CMs collected from hDPCs grown under normoxic (-Nor) and hypoxic (-Hyp) conditions were evaluated by quantitative PCR (VEGF-A, angiopoietin-1, angiopoietin-2, interleukin-6 (IL-6) and CXCL12), ELISA assays (VEGF-A, Ang-2), tube-formation and wound-healing assays, using human umbilical vein endothelial cells. The results demonstrated that hypoxic CM had significantly higher angiogenic potential than normoxic CM. Human fetal osteoblasts (hFOBs) were exposed to CM, followed by alizarin red staining, to assess the osteogenic potential. It was found that CM did not enhance the mineralization capacity of hFOBs. DO was performed in the tibiae of 30 mice, followed by a local injection of 20 µl CM (CM-Nor and CM-Hyp groups) or serum-free DMEM (control group) into the distraction zone every second day. The mice were sacrificed at days 13 and 27. The CM-Hyp treatment revealed a higher X-ray density than the control group (p < 0.05). Our study suggests that the angiogenic effect promoted by hypoxic culture conditions is dependent on VEGF-A and Ang-2 released from hDPCs. Furthermore, CM-Hyp treatment may thus improve the DO procedure, accelerating bone healing. © 2015 The Authors. Journal of Tissue Engineering and Regenerative Medicine published by John Wiley & Sons, Ltd.


Subject(s)
Dental Pulp/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Osteogenesis, Distraction , Tibia , Animals , Cell Hypoxia , Culture Media, Conditioned/pharmacology , Dental Pulp/pathology , Female , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Tibia/injuries , Tibia/metabolism , Tibia/pathology , Wound Healing/drug effects
8.
Biochem Biophys Rep ; 7: 316-322, 2016 Sep.
Article in English | MEDLINE | ID: mdl-28955921

ABSTRACT

Osseointegration is the structural and functional connection between bone tissues and implants such as titanium dioxide (TiO2). The bone-TiO2 interface is thought to contain proteoglycans. However, exhaustive analysis of the proteins in this layer has not been performed. In this study, we evaluated the bone protein adhered on the surface of TiO2 comprehensively. Pig bone protein was extracted by sequential elutions with guanidine, 0.1 M EDTA, and again with guanidine. The proteins obtained from these extractions were allowed to adhere to an HPLC column packed with TiO2 and were eluted with 0.2 M NaOH. The eluted proteins were identified by LC/MS/MS and included not only proteoglycans but also other proteins such as extracellular matrix proteins, enzymes, and growth factors. Calcium depositions were observed on TiO2 particles incubated with bone proteins, guanidine-extracted proteins adhered to TiO2 displayed significantly high amounts of calcium depositions.

9.
Int J Oral Maxillofac Implants ; 30(5): 1175-86, 2015.
Article in English | MEDLINE | ID: mdl-26394357

ABSTRACT

PURPOSE: To investigate whether bone regeneration can be accelerated by using a conditioned medium (CM) and guided bone regeneration (GBR) technique. MATERIALS AND METHODS: CM was harvested from rat bone marrow stromal cells (BMSCs). The components of CM were immobilized using a polylactide-co-glycolide (PLGA) membrane treated with and without 0.5 mol/L sodium hydroxide (NaOH) to elevate the hydrophilicity. Four experimental groups were prepared: PLGA membrane treated with (1) phosphate-buffered saline (PBS; PBS-M), (2) PBS and 0.5 mol/L NaOH (hydrophilic treatment; PBS-HM), (3) CM (CM-M), and (4) CM and 0.5 mol/L NaOH (CM-HM). These experimental membranes were observed using scanning electron microscopy. Proteins derived from BMSCs immobilized on the PLGA membrane were detected with liquid chromatography-tandem mass spectrometry (LC/MS/MS). Cell proliferation and alkaline phosphatase (ALP) activity were measured to analyze the effect of CM on the BMSCs. Experimental membranes were transplanted into a rat calvarial bone defect model. Microcomputed tomography and histologic analyses were performed 4 and 8 weeks after transplantation. RESULTS: The CM derived from BMSCs can be immobilized on the PLGA membrane. Hydrophilic treatment of the PLGA membrane enhanced the amount of CM immobilization. LC/MS/MS analysis showed that the immobilized proteins on the surface of PLGA membrane were extracellular matrix, such as collagen, decorin, and fibronectin. The proteins in the CM, which were released from the PLGA membrane, enhanced cell proliferation and ALP activity in rat BMSCs. Newly formed bone area at the bone defects that had been treated with CM-HM was significantly high compared with those at bone defects treated with the other membranes. CONCLUSION: The PLGA membrane treated with 0.5 mol/L NaOH and CM promoted bone regeneration in this rat calvarial defect model.


Subject(s)
Bone Regeneration/drug effects , Culture Media, Conditioned/chemistry , Guided Tissue Regeneration/methods , Immobilized Proteins/pharmacology , Lactic Acid/chemistry , Membranes, Artificial , Mesenchymal Stem Cells/physiology , Polyglycolic Acid/chemistry , Alkaline Phosphatase/analysis , Animals , Bone Diseases/pathology , Bone Diseases/surgery , Cell Proliferation/drug effects , Chromatography, Liquid/methods , Collagen/analysis , Collagen/pharmacology , Decorin/analysis , Decorin/pharmacology , Fibronectins/analysis , Fibronectins/pharmacology , Hydrophobic and Hydrophilic Interactions , Immobilized Proteins/analysis , Male , Microscopy, Electron, Scanning , Osteogenesis/drug effects , Polylactic Acid-Polyglycolic Acid Copolymer , Rats , Rats, Sprague-Dawley , Skull/pathology , Skull/surgery , Sodium Hydroxide/chemistry , Tandem Mass Spectrometry/methods , Time Factors , X-Ray Microtomography/methods
10.
Tissue Eng Part A ; 20(21-22): 3063-72, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24813721

ABSTRACT

Growth factors play an important role in osteo/odontogenic differentiation of human dental pulp cells (hDPCs). The aim of this in vitro study was to compare the biological effects of recombinant human growth differentiation factor 5 (rhGDF-5) alone and a cocktail of soluble growth factors (conditioned medium) released from human bone marrow mesenchymal stem cells (hBMMSCs) on the morphology, proliferation and osteo/odontogenic differentiation potential of hDPCs. Passage 4 hDPCs were harvested for culture in four different media: (a) DMEM with 10% FBS, (b) odontogenic induction medium (OM), (c) OM plus 500 ng/mL rhGDF-5, and (d) OM plus conditioned medium (CM). Morphological changes at 48 and 120 h were determined by crystal violet staining. The proliferation rates at 3, 24, 48, and 120 h were assayed by MTT. Using real-time reverse transcription-polymerase chain reaction (RT-PCR), the mRNA levels of dentin sialophosphoprotein (DSPP), dentin matrix protein 1 (DMP1), collagen type I (Col 1), Runt-related transcription factor 2 (Cbfa1/Runx2), alkaline phosphatase (ALP), osteocalcin (OC), ß3 tubulin (TUBB3), glial cell-derived neurotrophic factor (GDNF), angiopoietin-1 (Ang1), and vascular endothelial growth factor A (VEGFA), were determined at 2, 5, and 9 days. Protein expression of dental sialoprotein (DSP), DMP1, OC, and TUBB3 was recorded at 5 days, using western blot and immunocytochemistry. The effect of the different culture media on mineralization was determined by ALP staining at day 5 and Alizarin red S staining at days 7 and 14. In response to the different culture media, the shape of the hDPCs varied from spindled to polygonal and cuboidal. CM inhibited the cellular proliferation rate, while rhGDF-5 had no effect at early time points, but promoted cellular proliferation at 120 h of culture. In the CM group, the mRNA levels of Cbfa1/Runx2, Col 1, ALP, VEGFA, Ang1, and TUBB3 decreased and the levels of GDNF and OC increased. The mRNA levels of DSPP and DMP1 were inconsistent at the time points evaluated. The staining assays also demonstrated that compared with the other groups, the CM group exhibited lower expression of ALP and higher mineralization levels. Protein expression of DSP, DMP1, OC, and TUBB3 was pronounced by the CM-treated cells. It is concluded that under these in vitro conditions, CM released from hBMMSCs have a greater osteo/odontogenic inductive effect on hDPCs than rhGDF-5.


Subject(s)
Bone Marrow Cells/metabolism , Dental Pulp/cytology , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/metabolism , Odontogenesis/physiology , Osteogenesis/physiology , Paracrine Communication/physiology , Adolescent , Adult , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Dental Pulp/drug effects , Female , Humans , Male , Odontogenesis/drug effects , Osteogenesis/drug effects , Tissue Engineering/methods , Young Adult
11.
Bone ; 61: 82-90, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24389414

ABSTRACT

Distraction osteogenesis (DO) successfully induces large-scale skeletal tissue regeneration, but it involves an undesirably long treatment period. A high-speed DO mouse model (H-DO) with a distraction speed twice that of a control DO model failed to generate new bone callus in the distraction gap. Here we demonstrate that the local administration of serum-free conditioned medium from human mesenchymal stem cells (MSC-CM) accelerated callus formation in the mouse H-DO model. Secretomic analysis identified factors contained in MSC-CM that recruit murine bone marrow stromal cells (mBMSCs) and endothelial cells/endothelial progenitor cells (EC/EPCs), inhibit inflammation and apoptosis, and promote osteoblast differentiation, angiogenesis, and cell proliferation. Functional assays identified MCP-1/-3 and IL-3/-6 as essential factors in recruiting mBMSCs and EC/EPCs. IL-3/-6 also enhanced the osteogenic differentiation of mBMSCs. MSC-CM that had been depleted of MCP-1/-3 failed to recruit mBMSCs, and consequently failed to promote callus formation. Taken together, our data suggest that MSCs produce a broad repertoire of trophic factors with tissue-regenerative activities that accelerate healing in the DO process.


Subject(s)
Bone and Bones/metabolism , Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cells/metabolism , Osteogenesis, Distraction/methods , Osteogenesis/drug effects , Animals , Bone and Bones/drug effects , Cell Differentiation/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cell Transplantation , Mice , Mice, Inbred ICR , Osteoblasts/cytology , Osteoblasts/drug effects
12.
Bone ; 49(4): 693-700, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21741502

ABSTRACT

Distraction osteogenesis (DO) is a unique therapy that induces skeletal tissue regeneration without stem/progenitor cell transplantation. Although the self-regeneration property of DO provides many clinical benefits, the long treatment period required is a major drawback. A high-speed DO mouse model (H-DO), in which the distraction was done two times faster than in control DO (C-DO) mice, failed to generate new bone callus in the DO gap. We found that this was caused by the unsuccessful recruitment of bone marrow endothelial cells (BM-ECs)/endothelial progenitor cells (EPCs) into the gap. We then tested the ability of a local application of stromal cell-derived factor-1 (SDF-1), a major chemo-attractant for BM-ECs/EPCs, to accelerate the bone regeneration in H-DO. Our data showed that, in H-DO, SDF-1 induced callus formation in the gap through the recruitment of BM-ECs/EPCs, the maturation of neo-blood vessels, and increased blood flow. These results indicate that the active recruitment of endogenous BM-ECs/EPCs may provide a substantial clinical benefit for shortening the treatment period of DO.


Subject(s)
Bone Regeneration , Chemokine CXCL12/metabolism , Endothelial Cells/cytology , Osteogenesis, Distraction , Stem Cells/cytology , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Regeneration/drug effects , Bone and Bones/blood supply , Bone and Bones/metabolism , Bone and Bones/pathology , Bony Callus/drug effects , Bony Callus/pathology , Chemokine CXCL12/administration & dosage , Chemokine CXCL12/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Humans , Ischemia/pathology , Mice , Mice, Inbred ICR , Models, Animal , Neovascularization, Physiologic/drug effects , Receptors, CXCR/metabolism , Receptors, CXCR4/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Wound Healing/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL