Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Biochem Biophys Res Commun ; 558: 231-238, 2021 06 18.
Article in English | MEDLINE | ID: mdl-32113685

ABSTRACT

Several groups have developed in vitro expansion cultures for mouse metanephric nephron progenitor cells (NPCs) using cocktails of small molecules and growth factors including BMP7. However, the detailed mechanisms by which BMP7 acts in the NPC expansion remain to be elucidated. Here, by performing chemical screening for BMP substitutes, we identified a small molecule, TCS21311, that can replace BMP7 and revealed a novel inhibitory role of BMP7 in JAK3-STAT3 signaling in NPC expansion culture. Further, we found that TCS21311 facilitates the proliferation of mouse embryonic NPCs and human induced pluripotent stem cell-derived NPCs when added to the expansion culture. These results will contribute to understanding the mechanisms of action of BMP7 in NPC proliferation in vitro and in vivo and to the stable supply of NPCs for regenerative therapy, disease modeling and drug discovery for kidney diseases.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Janus Kinase Inhibitors/pharmacology , Nephrons/cytology , Nephrons/drug effects , Animals , Bone Morphogenetic Protein 7/administration & dosage , Cell Proliferation/drug effects , Cell Proliferation/physiology , Culture Media , Drug Evaluation, Preclinical , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Janus Kinase 3/antagonists & inhibitors , Mice , Mice, 129 Strain , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Nephrons/metabolism , Signal Transduction/drug effects , Small Molecule Libraries
2.
J Cell Mol Med ; 24(22): 13507-13522, 2020 11.
Article in English | MEDLINE | ID: mdl-33079436

ABSTRACT

While the bone morphogenetic protein-7 (BMP-7) is a well-known therapeutic growth factor reverting many fibrotic diseases, including peritoneal fibrosis by peritoneal dialysis (PD), soluble growth factors are largely limited in clinical applications owing to their short half-life in clinical settings. Recently, we developed a novel drug delivery model using protein transduction domains (PTD) overcoming limitation of soluble recombinant proteins, including bone morphogenetic protein-7 (BMP-7). This study aims at evaluating the therapeutic effects of PTD-BMP-7 consisted of PTD and full-length BMP-7 on epithelial-mesenchymal transition (EMT)-related fibrosis. Human peritoneal mesothelial cells (HPMCs) were then treated with TGF-ß1 or TGF-ß1 + PTD-BMP-7. Peritoneal dialysis (PD) catheters were inserted into Sprague-Dawley rats, and these rats were infused intra-peritoneally with saline, peritoneal dialysis fluid (PDF) or PDF + PTD-BMP-7. In vitro, TGF-ß1 treatment significantly increased fibronectin, type I collagen, α-SMA and Snail expression, while reducing E-cadherin expression in HPMCs (P < .001). PTD-BMP-7 treatment ameliorated TGF-ß1-induced fibronectin, type I collagen, α-SMA and Snail expression, and restored E-cadherin expression in HPMCs (P < .001). In vivo, the expressions of EMT-related molecules and the thickness of the sub-mesothelial layer were significantly increased in the peritoneum of rats treated with PDF, and these changes were significantly abrogated by the intra-peritoneal administration of PTD-BMP-7. PTD-BMP-7 treatment significantly inhibited the progression of established PD fibrosis. These findings suggest that PTD-BMP-7, as a prodrug of BMP-7, can be an effective therapeutic agent for peritoneal fibrosis in PD patients.


Subject(s)
Bone Morphogenetic Protein 7/administration & dosage , Drug Delivery Systems , Peritoneal Fibrosis/drug therapy , Animals , Biomarkers , Bone Morphogenetic Protein 7/chemistry , Disease Models, Animal , Drug Design , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Immunohistochemistry , Intravital Microscopy , Male , Mice , Peritoneal Fibrosis/etiology , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Rats , Recombinant Proteins , Treatment Outcome
3.
Hand Surg Rehabil ; 39(5): 383-388, 2020 10.
Article in English | MEDLINE | ID: mdl-32540417

ABSTRACT

The scaphoid is the most common non-union site in the wrist. Fixation with vascularised or non-vascularised autograft is the gold standard when it comes to treating these non-unions. But, what can we offer if the autograft fails? Using osteoinductive proteins in difficult cases of long bone non-union yields good results. However, only a few studies have been published on their use for scaphoid non-union. In our study, five patients with an average age of 32 years (ranging from 21 to 44 years) with old non-union (more than 24 months) of the scaphoid were treated after autograft treatment had failed. The procedure consisted of reaming the non-union site, then adding bone autograft combined with BMP-7 (Osigraft®) in the defect and fixing it all with a screw or K-wire. Postoperative immobilisation was prescribed. Only one patient achieved bone union (20%) despite an average follow-up of 10 years (80-143 months). The average flexion-extension loss was 16.6° (0-30) relative to the contralateral side. The average strength deficit was 450 grams (0-2000) for pinch and 12.1kg (0-29) for grip compared to the contralateral side. Self-assessment questionnaires had an average PRWE at 28.9 (10.5-49) and an average QuickDASH at 28.6 (9.09-61.36). Our study could not demonstrate any real benefit of using BMP-7 for treating old scaphoid non-union despite an elevated cost. Further research is needed to look at other treatment approaches, for instance, the use of new scaffolds combining VEGF and BMP.


Subject(s)
Bone Morphogenetic Protein 7/administration & dosage , Fractures, Ununited/surgery , Scaphoid Bone/surgery , Adult , Autografts , Cancellous Bone/transplantation , Disability Evaluation , Follow-Up Studies , Fracture Fixation, Internal , Fracture Healing , Hand Strength , Humans , Retrospective Studies , Young Adult
4.
Biomater Sci ; 8(3): 871-883, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-31820744

ABSTRACT

The choice of grafts for anterior cruciate ligament (ACL) reconstruction is a critical issue in sports medicine. Previous studies have revealed that scaffolds prepared from a single material could not achieve complete integration between the graft and autogenous bone tunnel. To solve this problem, we hypothesize that combining degradable scaffolds with nondegradable scaffolds can produce a novel hybrid ligament with the advantages of both types of scaffolds. In this study, a bone morphogenetic protein 7 (BMP-7)-loaded polycaprolactone (PCL) nanofibrous membrane was first manufactured as the degradable part of the hybrid ligament by using layer-by-layer (LbL) self-assembly. Then, we fabricated a multifunctional novel hybrid ligament by rolling up this nanofibrous membrane and polyethylene terephthalate (PET) mesh fabric (nondegradable part) into a "swiss roll" structure. The in vitro experimental results showed that this hybrid ligament could significantly improve the biocompatibility of pure PET ligament and further promote cell mineralization. The in vivo experimental results showed that this unique structure significantly promoted the integration of hybrid ligaments and bone tunnels, thereby achieving real "ligamentization" after ACL reconstruction surgery. These results suggest that this novel hybrid biomimetic artificial ligament scaffold provides a new direction for graft selection for ACL reconstruction.


Subject(s)
Anterior Cruciate Ligament/surgery , Bone and Bones/surgery , Tendons/physiopathology , Animals , Anterior Cruciate Ligament/physiopathology , Anterior Cruciate Ligament Reconstruction , Bone Morphogenetic Protein 7/administration & dosage , Bone Morphogenetic Protein 7/metabolism , Bone and Bones/physiopathology , Humans , Ligaments/injuries , Ligaments/surgery , Nanofibers/administration & dosage , Nanofibers/chemistry , Polyesters/chemistry , Rabbits , Tendon Injuries/physiopathology , Tendon Injuries/surgery , Tendons/surgery , Tissue Scaffolds/chemistry , Wound Healing
5.
J Orthop Surg Res ; 14(1): 297, 2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31488155

ABSTRACT

BACKGROUND: Due to our aging population, an increase in proximal femur fractures can be expected, which is associated with impaired activities of daily living and a high risk of mortality. These patients are also at a high risk to suffer a secondary osteoporosis-related fracture on the contralateral hip. In this context, growth factors could open the field for regenerative approaches, as it is known that, i.e., the growth factor BMP-7 (bone morphogenetic protein 7) is a potent stimulator of osteogenesis. Local prophylactic augmentation of the proximal femur with a BMP-7 loaded thermoresponsive hydrogel during index surgery of an osteoporotic fracture could be suitable to reduce the risk of further osteoporosis-associated secondary fractures. The present study therefore aims to test the hypothesis if a BMP-7 augmented hydrogel is an applicable carrier for the augmentation of non-fractured proximal femurs. Furthermore, it needs to be shown that the minimally invasive injection of a hydrogel into the mouse femur is technically feasible. METHODS: In this study, male C57BL/6 mice (n = 36) received a unilateral femoral intramedullary injection of either 100 µl saline, 100 µl 1,4 Butan-Diisocyanat (BDI)-hydrogel, or 100 µl hydrogel loaded with 1 µg of bone morphogenetic protein 7. Mice were sacrificed 4 and 12 weeks later. The femora were submitted to high-resolution X-ray tomography and subsequent histological examination. RESULTS: Analysis of normalized CtBMD (Cortical bone mineral density) as obtained by X-ray micro-computed tomography analysis revealed significant differences depending on the duration of treatment (4 vs 12 weeks; p < 0.05). Furthermore, within different anatomically defined regions of interest, significant associations between normalized TbN (trabecular number) and BV/TV (percent bone volume) were noted. Histology indicated no signs of inflammation and no signs of necrosis and there were no cartilage damages, no new bone formations, or new cartilage tissues, while BMP-7 was readily detectable in all of the samples. CONCLUSIONS: In conclusion, the murine femoral intramedullary injection model appears to be feasible and worth to be used in subsequent studies that are directed to examine the therapeutic potential of BMP-7 loaded BDI-hydrogel. Although we were unable to detect any significant osseous effects arising from the mode or duration of treatment in the present trial, the effect of different concentrations and duration of treatment in an osteoporotic model appears of interest for further experiments to reach translation into clinic and open new strategies of growth factor-mediated augmentation.


Subject(s)
Bone Morphogenetic Protein 7/administration & dosage , Femoral Fractures/prevention & control , Femur/drug effects , Hydrogels/administration & dosage , Animals , Bone Morphogenetic Protein 7/analysis , Drug Evaluation, Preclinical/methods , Femoral Fractures/pathology , Femur/chemistry , Femur/pathology , Fracture Fixation, Intramedullary/methods , Hydrogels/analysis , Male , Mice , Mice, Inbred C57BL
6.
World J Gastroenterol ; 25(30): 4222-4234, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31435175

ABSTRACT

BACKGROUND: Liver fibrosis is a refractory disease whose persistence can eventually induce cirrhosis or even liver cancer. Early liver fibrosis is reversible by intervention. As a member of the transforming growth factor-beta (TGF-ß) superfamily, bone morphogenetic protein 7 (BMP7) has anti-liver fibrosis functions. However, little is known about BMP7 expression changes and its potential regulatory mechanism as well as the relationship between BMP7 and TGF-ß during liver fibrosis. In addition, the mechanism underlying the anti-liver fibrosis function of BMP7 needs to be further explored. AIM: To investigate changes in the dynamic expression of BMP7 during liver fibrosis, interactions between BMP7 and TGF-ß1, and possible mechanisms underlying the anti-liver fibrosis function of BMP7. METHODS: Changes in BMP7 expression during liver fibrosis and the interaction between BMP7 and TGF-ß1 in mice were observed. Exogenous BMP7 was used to treat mouse primary hepatic stellate cells (HSCs) to observe its effect on activation, migration, and proliferation of HSCs and explore the possible mechanism underlying the anti-liver fibrosis function of BMP7. Mice with liver fibrosis received exogenous BMP7 intervention to observe improvement of liver fibrosis by using Masson's trichrome staining and detecting the expression of the HSC activation indicator alpha-smooth muscle actin (α-SMA) and the collagen formation associated protein type I collagen (Col I). Changes in the dynamic expression of BMP7 during liver fibrosis in the human body were further observed. RESULTS: In the process of liver fibrosis induced by carbon tetrachloride (CCl4) in mice, BMP7 protein expression first increased, followed by a decrease; there was a similar trend in the human body. This process was accompanied by a sustained increase in TGF-ß1 protein expression. In vitro experiment results showed that TGF-ß1 inhibited BMP7 expression in a time- and dose-dependent manner. In contrast, high doses of exogenous BMP7 inhibited TGF-ß1-induced activation, migration, and proliferation of HSCs; this inhibitory effect was associated with upregulation of pSmad1/5/8 and downregulation of phosphorylation of Smad3 and p38 by BMP7. In vivo experiment results showed that exogenous BMP7 improved liver fibrosis in mice. CONCLUSION: During liver fibrosis, BMP7 protein expression first increases and then decreases. This changing trend is associated with inhibition of BMP7 expression by sustained upregulation of TGF-ß1 in a time- and dose-dependent manner. Exogenous BMP7 could selectively regulate TGF-ß/Smad pathway-associated factors to inhibit activation, migration, and proliferation of HSCs and exert anti-liver fibrosis functions. Exogenous BMP7 has the potential to be used as an anti-liver fibrosis drug.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/pathology , Liver/pathology , Administration, Oral , Animals , Bone Morphogenetic Protein 7/administration & dosage , Carbon Tetrachloride/toxicity , Cells, Cultured , Down-Regulation , Hepatic Stellate Cells/drug effects , Humans , Liver/cytology , Liver/drug effects , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Mice , Phosphorylation , Primary Cell Culture , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Up-Regulation
7.
BMC Musculoskelet Disord ; 19(1): 261, 2018 Jul 27.
Article in English | MEDLINE | ID: mdl-30049273

ABSTRACT

BACKGROUND: Bone infections due to trauma and subsequent delayed or impaired fracture healing represent a great challenge in orthopedics and trauma surgery. The prevalence of such bacterial infection-related types of delayed non-union is high in complex fractures, particularly in open fractures with additional extensive soft-tissue damage. The aim of this study was to establish a rat model of delayed osseous union secondary to bacterial osteitis and investigate the impact of rhBMP-7 and rhBMP-2 on fracture healing in the situation of an ongoing infection. METHODS: After randomization to four groups 72 Sprague-Dawley rats underwent a transverse fracture of the midshaft tibia stabilized by intramedullary titanium K-wires. Three groups received an intramedullary inoculation with Staphylococcus aureus (103 colony-forming units) before stabilization and the group without bacteria inoculation served as healing control. After 5 weeks, a second surgery was performed with irrigation of the medullary canal and local rhBMP-7 and rhBMP-2 treatment whereas control group and infected control group received sterile saline. After further 5 weeks rats were sacrificed and underwent biomechanical testing to assess the mechanical stability of the fractured bone. Additional micro-CT analysis, histological, and histomorphometric analysis were done to evaluate bone consolidation or delayed union, respectively, and to quantify callus formation and the mineralized area of the callus. RESULTS: Biomechanical testing showed a significantly higher fracture torque in the non-infected control group and the infected rhBMP-7- and rhBMP-2 group compared with the infected control group (p < 0.001). RhBMP-7 and rhBMP-2 groups did not show statistically significant differences (p = 0.57). Histological findings supported improved bone-healing after rhBMP treatment but quantitative micro-CT and histomorphometric results still showed significantly more hypertrophic callus tissue in all three infected groups compared to the non-infected group. Results from a semiquantitative bone-healing-score revealed best bone-healing in the non-infected control group. The expected chronic infection was confirmed in all infected groups. CONCLUSIONS: In delayed bone healing secondary to infection rhBMP treatment promotes bone healing with no significant differences in the healing efficacy of rhBMP-2 and rhBMP-7 being noted. Further new therapeutic bone substitutes should be analyzed with the present rat model for delayed osseous union secondary to bacterial osteitis.


Subject(s)
Bone Morphogenetic Protein 2/administration & dosage , Bone Morphogenetic Protein 7/administration & dosage , Fracture Healing/physiology , Fractures, Bone/drug therapy , Osteitis/drug therapy , Staphylococcal Infections/drug therapy , Animals , Disease Models, Animal , Female , Fractures, Bone/diagnostic imaging , Osteitis/diagnostic imaging , Random Allocation , Rats , Rats, Sprague-Dawley , Staphylococcal Infections/diagnostic imaging , Staphylococcus aureus/drug effects , Time Factors , Treatment Outcome
8.
J Oral Maxillofac Surg ; 76(5): 1092.e1-1092.e10, 2018 May.
Article in English | MEDLINE | ID: mdl-29425753

ABSTRACT

PURPOSE: The effects of a recombinant human bone morphogenetic protein-2/7 (rhBMP-2/7) heterodimer and a RADA16 (Ac-RADARADARADARADA-CONH2) hydrogel scaffold on bone formation during distraction osteogenesis were evaluated. MATERIALS AND METHODS: Forty New Zealand white rabbits, which underwent mandibular lengthening, were randomly divided into 5 groups. One group served as the control group. The others received 2 µg of rhBMP-2 homodimer, 2 µg of rhBMP-2/7 heterodimer, 100 µL of RADA16, or 100 µL of RADA16 plus 2 µg of rhBMP-2/7 heterodimer in the mandibular distraction gap at the beginning of distraction. Fluorine-18-labeled fluoride positron emission tomography was used to assess osteogenesis both after distraction and at the end of consolidation. Dual-energy x-ray absorptiometry (DEXA) examination and bone histologic findings were also evaluated. RESULTS: At the end of distraction, the radioactivity concentration in the distracted area was significantly greater in the RADA16 plus rhBMP-2/7 heterodimer group than in the other groups (P < .01). The differences among the other 4 groups were also statistically significant in the following order: rhBMP-2/7 heterodimer group greater than the rhBMP-2 homodimer group, which was greater than the RADA16 group (or control group; P < .05). However, the radioactivity concentration of the RADA16 group was slightly greater than that of the control group with a nonsignificant difference (P > .05). By the end of consolidation, the activity in the control group, RADA16 group, rhBMP-2 homodimer group, and rhBMP-2/7 heterodimer group had significantly diminished (P < .05). However, the activity in the RADA16 plus rhBMP-2/7 heterodimer group remained at the same level (P > .05). The DEXA results and bone histologic findings indicated that more callus regeneration was noted in the RADA16 plus rhBMP-2/7 heterodimer group than in any other group. CONCLUSIONS: The use of rhBMP-2/7 heterodimer and RADA16 hydrogel scaffold significantly promoted mandibular distraction osteogenesis.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 7/pharmacology , Mandible/drug effects , Osteogenesis, Distraction/methods , Osteogenesis/drug effects , Peptides/pharmacology , Tissue Scaffolds , Transforming Growth Factor beta/pharmacology , Animals , Bone Density/drug effects , Bone Morphogenetic Protein 2/administration & dosage , Bone Morphogenetic Protein 7/administration & dosage , Bone Regeneration/drug effects , Humans , Hydrogels , Male , Mandible/physiology , Peptides/administration & dosage , Rabbits , Random Allocation , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Transforming Growth Factor beta/administration & dosage
9.
Sci Rep ; 8(1): 1591, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29371668

ABSTRACT

Osteoporosis, a prevalent metabolic bone disorder, predisposes individuals to increased susceptibility to fractures. It is also, somewhat controversially, thought to delay or impair the regenerative response. Using high-resolution Fourier-transform infrared spectroscopy and small/wide-angle X-ray scattering we sought to answer the following questions: Does the molecular composition and the nano-structure in the newly regenerated bone differ between healthy and osteoporotic environments? And how do pharmacological treatments, such as bone morphogenetic protein 7 (BMP-7) alone or synergistically combined with zoledronate (ZA), alter callus composition and nano-structure in such environments? Cumulatively, on the basis of compositional and nano-structural characterizations of newly formed bone in an open-osteotomy rat model, the healing response in untreated healthy and ovariectomy-induced osteoporotic environments was fundamentally the same. However, the BMP-7 induced osteogenic response resulted in greater heterogeneity in the nano-structural crystal dimensions and this effect was more pronounced with osteoporosis. ZA mitigated the effects of the upregulated catabolism induced by both BMP-7 and an osteoporotic bone environment. The findings contribute to our understanding of how the repair processes in healthy and osteoporotic bone differ in both untreated and treated contexts and the data presented represents the most comprehensive study of fracture healing at the nanoscale undertaken to date.


Subject(s)
Bony Callus/chemistry , Fracture Healing , Fractures, Bone/pathology , Osteoporosis/pathology , Animals , Bone Density Conservation Agents/administration & dosage , Bone Morphogenetic Protein 7/administration & dosage , Diphosphonates/administration & dosage , Disease Models, Animal , Imidazoles/administration & dosage , Rats , Scattering, Small Angle , Spectroscopy, Fourier Transform Infrared , Zoledronic Acid
10.
Physiol Rep ; 5(16)2017 Aug.
Article in English | MEDLINE | ID: mdl-28867673

ABSTRACT

BMP7 is expressed in ureteric buds and cap mesenchyme of the fetal kidney, mediating branching morphogenesis and survival and priming of metanephric mesenchyme. Although dose-dependent effects of BMP7 in collecting duct cells have been reported, studies in metanephric mesenchymal cells are lacking. We examined the effects of BMP7 on MAP kinase activation, proliferation, and expression of cadherins in a metanephric mesenchymal cell line MS7 by thymidine incorporation, immunoblot analysis, and quantitative real-time PCR The levels of phosphorylated ERK (P-ERK) and phosphorylated p38 (P-p38) were not altered at 10 min, 1 h, and 6 h with low-dose BMP7 (0.25 nmol/L), but were increased at 24 h. At 24 h, P-ERK was increased with low-dose BMP7, but not by intermediate- (1 nmol/L) or high-dose (10 nmol/L) BMP7, whereas p38 was activated by intermediate-dose BMP7. Cell proliferation of MS7 was significantly increased by low- and intermediate-dose BMP7 and decreased by high-dose BMP7. A p38 inhibitor SB203580 5 µmol/L or a MEK inhibitor PD98059 5 µmol/L abolished BMP7-stimulated proliferation. Expression of cadherin-11, an adhesion molecule known to promote cell migration and compaction, was upregulated by intermediate-dose BMP7. BMP7-induced cadherin-11 expression was inhibited by cotreatment with SB203580 and PD98059. Finally, in metanephroi cultured with siRNA for cadherin-11, the number and thickness of cap mesenchyme were reduced. In conclusion, BMP7 exerts differential effects depending on the concentration; it may expand mesenchymal cells in the stroma where BMP7 concentration is low and may upregulate cadherin-11 promoting condensation around the tip of ureteric buds.


Subject(s)
Bone Morphogenetic Protein 7/metabolism , Cadherins/metabolism , Cell Proliferation , Kidney/physiology , Mitogen-Activated Protein Kinase 3/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bone Morphogenetic Protein 7/administration & dosage , Cell Line , Mice , Organ Culture Techniques
11.
J Recept Signal Transduct Res ; 37(5): 515-521, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28812969

ABSTRACT

BACKGROUND: Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-ß (TGF-ß) superfamily. Recently, BMP7 has been demonstrated to be produced by salivary glands and contribute to embryonic branching in mice. The BMP7 in saliva is thought to be delivered to the oral cavity and is expected to contact with stratified squamous epithelial cells which line the surface of oral mucosa. In this study, we attempted to investigate the effects of BMP7 on oral epithelial cells. METHODS: The expression of BMP receptors was examined by reverse transcriptase-polymerase chain reaction (RT-PCR). OSCCs were stimulated with human recombinant BMP7 (hrBMP7) and the phosphorylation status of Smad1/5/8 was examined by western blotting. For microarray analysis, Ca9-22 cells were stimulated with 100 ng/mL of hrBMP7 and total RNA was extracted and subjected to real-time PCR. The 5'-untranslated region (5'-UTR) of IL-17 F gene was cloned to pGL4-basic vector and used for luciferase assay. Ca9-22 cells were pre-incubated with DM3189, a specific inhibitor of Smad1/5/8, for inhibition assay. RESULTS: All isoforms of type I and type II BMP receptors were expressed in both Ca9-22 and HSC3 cells and BMP7 stimulation resulted in the phosphorylation of Smad1/5/8 in both cell lines. The microarray analysis revealed the induction of interleukin-17 F (IL-17 F), netrin G2 (NTNG2) and hyaluronan synthase 1 (HAS1). Luciferase assay using the 5'-UTR of the IL-17 F gene revealed transcriptional regulation. Induced IL-17 F production was further confirmed at the protein level by ELISA. Smad1/5/8 inhibitor pretreatment decreased IL-17 F expression levels in the cells.


Subject(s)
Bone Morphogenetic Protein 7/genetics , Carcinoma, Squamous Cell/genetics , Interleukin-17/genetics , Mouth Neoplasms/genetics , Recombinant Proteins/genetics , Bone Morphogenetic Protein 7/administration & dosage , Bone Morphogenetic Protein 7/metabolism , Carcinoma, Squamous Cell/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyaluronan Synthases/genetics , Mouth Neoplasms/pathology , Netrins/genetics , Phosphorylation , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Smad Proteins/antagonists & inhibitors , Smad Proteins/genetics , Transforming Growth Factor beta/genetics
12.
Eur J Pharm Biopharm ; 114: 69-78, 2017 May.
Article in English | MEDLINE | ID: mdl-28087378

ABSTRACT

The regeneration of articular cartilage remains an unresolved question despite the current access to a variety of tissue scaffolds activated with growth factors relevant to this application. Further advances might result from combining more than one of these factors; here, we propose a scaffold composition optimized for the dual delivery of BMP-7 and TGF-ß3, two proteins with described chondrogenic activity. First, we tested in a mesenchymal stem cell micromass culture with TGF-ß3 whether the exposure to microspheres loaded with BMP-7 would improve cartilage formation. Histology and qRT-PCR data confirmed that the sustained release of BMP-7 cooperates with TGF-ß3 towards chondrogenic differentiation. Then, we optimized a scaffold prototype for tissue culture and dual encapsulation of BMP-7 and TGF-ß3. The scaffolds were prepared from poly(lactic-co-glycolic acid), and BMP-7/TGF-ß3 were loaded as nanocomplexes with heparin and Tetronic 1107. The scaffolds showed the sustained release of both proteins over four weeks, with minimal burst effect. We finally cultured human mesenchymal stem cells on these scaffolds, in the absence of exogenous chondrogenic factor supplementation. The cells cultured on the scaffolds loaded with BMP-7 and TGF-ß3 showed clear signs of cartilage formation macroscopically and histologically. RT-PCR studies confirmed a clear upregulation of cartilage markers SOX9 and Aggrecan. In summary, scaffolds encapsulating BMP-7 and TGF-ß3 can efficiently deliver a cooperative growth factor combination that drives efficient cartilage formation in human mesenchymal stem cell cultures. These results open attractive perspectives towards in vivo translation of this technology in cartilage regeneration experiments.


Subject(s)
Bone Morphogenetic Protein 7/administration & dosage , Bone Morphogenetic Protein 7/pharmacology , Chondrocytes/drug effects , Chondrogenesis/drug effects , Tissue Scaffolds/chemistry , Transforming Growth Factor beta3/administration & dosage , Transforming Growth Factor beta3/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Drug Combinations , Drug Compounding , Humans , Lactic Acid , Mesenchymal Stem Cells/drug effects , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Regeneration/drug effects , Tissue Engineering/methods
13.
J Orthop Res ; 35(6): 1311-1322, 2017 06.
Article in English | MEDLINE | ID: mdl-26218641

ABSTRACT

The main reason for intervertebral disc (IVD) degeneration is the decrease in the quantity and activity of IVD cells with subsequent reduction of the extracellular matrix (ECM). In this study, we investigated a cell-based repair strategy by injecting nucleus pulposus cells (NPCs) transduced with human bone morphogenetic protein (hBMP7) by adeno-associated virus-2 into the canine degenerative IVD to determine whether NPCs expressing hBMP7 could delay the degeneration of the IVD. Fourteen canines received annular punctures to induce disc degeneration. Eight weeks later, saline (group A), allogeneic NPCs (group B), or allogeneic NPCs transduced with hBMP7 (group C) were injected into the degenerative discs. Twelve weeks after the injection, MRI scan showed that the degeneration process of groups C was slower and less severe compared with that of groups B and C. The IVD stability in group C was superior to that in groups A and B in left-right bending and rotation. HE, safranin-O staining, and ELISA indicated that the degenerative degree of the IVD in group C was significantly milder than that in groups A and B. The study demonstrated that the implantation of NPCs-hBMP7 could effectively maintained the structural integrity, ECM, and biomechanical properties of the canine degenerated discs. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1311-1322, 2017.


Subject(s)
Bone Morphogenetic Protein 7/administration & dosage , Cell- and Tissue-Based Therapy/methods , Genetic Therapy/methods , Intervertebral Disc Degeneration/therapy , Animals , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Dogs , Female , Gene Transfer Techniques , Humans , Intervertebral Disc Degeneration/diagnostic imaging , Magnetic Resonance Imaging , Nucleus Pulposus/cytology , Radiography
14.
Int Orthop ; 40(11): 2339-2345, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27520739

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the clinical and radiological outcome following compression plate fixation in combination with autologous bone grafting, with and without additional application of recombinant human bone morphogenetic protein (rhBMP) for treatment of aseptic clavicle non-union. METHODS: Between April 2004 and April 2015, 82 patients were treated for clavicle fracture and had developed aseptic clavicle non-union. Seventy-three out of 82 patients were available for follow-up at least one year after revision surgery; among them, 27 women and 46 men, with a median age of 49 (range, 19-86) years. Forty-five patients received compression plate osteosynthesis with autologous bone grafting, and 28 patients obtained compression plate fixation with autologous bone grafting and additional application of rhBMP-2 (3/28 patients) or rhBMP-7 (25/28 patients). RESULTS: Seventy out of 73 non-unions (96 %) healed within 12 months after revision surgery. Functional outcome according to the DASH Outcome Measure (with rhBMP, 33.16 ± 1.17 points; without rhBMP, 30.58 ± 2.12 points [mean ± SEM]; p = 0.81), non-union healing (p = 0.86), time interval between revision surgery and bone healing (p = 0.37), as well as post-operative complications, did not demonstrate relevant differences between the treatment groups and were not age-dependent. DISCUSSION: Functional and radiological results demonstrate that successful healing of aseptic clavicle non-union is dependent on radical resection of non-union tissue, restoration of length of the shoulder girdle and application of stable locking-plate osteosynthesis in combination with autologous bone grafting, but not dependent on application of additional rhBMP.


Subject(s)
Bone Morphogenetic Protein 2/administration & dosage , Bone Morphogenetic Protein 7/administration & dosage , Clavicle/surgery , Fracture Fixation, Internal/instrumentation , Fracture Healing/physiology , Fractures, Ununited/surgery , Transforming Growth Factor beta/administration & dosage , Administration, Topical , Adult , Aged , Aged, 80 and over , Bone Plates , Bone Transplantation , Clavicle/injuries , Female , Humans , Male , Middle Aged , Recombinant Proteins/administration & dosage , Reoperation , Retrospective Studies , Transplantation, Autologous , Treatment Outcome , Young Adult
15.
J Biomed Nanotechnol ; 12(4): 602-18, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27301188

ABSTRACT

Enhancing the biocompatibility and osteogenic activity of nano-apatite for applications in bone graft substitutes and bone tissue engineering have been the current challenge in regeneration of lost bone. Inspired by mussels, here we have developed facile biomimetic approaches for preparation of two types of peptide-conjugated apatite nanocompsoties assisted by polydopamine (pDA). We exploited polydopamine chemistry for the modification of nano-apatite crystals: polydopamine coated apatite (HA-c-pDA) and polydopamine template-mediated apatite (HA-t-pDA), on which bone forming peptide was subsequently immobilized under weakly basic conditions to obtain peptide-conjugated apatite nanocomposites (HA-c-pep and HA-t-pep, respectively). TEM images revealed that HA-c-pDA displayed typically rod-like morphology, while HA-t-pDA was sponge-like structure where pDA sheets were decorated by needle-like apatite crystals with low degree of crystallinity. In the cell culture experiments, HA-t-pep nanocomposite exhibited higher cell proliferation, spreading, and alkaline phosphatase activity as well as calcium nodule-formation, compared with pristine nano-HA and HA-c-pep nanocomposite. We then implanted the peptide-decorated apatite into rabbit calvarial defects and analyzed bone formation after 2 months. The data revealed that HA-t-pep group exhibited remarkably enhanced bioactivity and bone formation in vivo. Based on these results, our biomimetic approach could be a promising tool to develop peptide-conjugated apatites for bone regeneration. Meanwhile, the excellent biocompatibility and high osteogenesis of the peptide-conjugated apatite nanocomposite might confer its great potentials in bone repair, bone augmentation, as well as coating of biomedical implants.


Subject(s)
Bone Morphogenetic Protein 7/administration & dosage , Durapatite/chemistry , Indoles/chemistry , Nanocomposites/chemistry , Osteoblasts/cytology , Osteogenesis/physiology , Polymers/chemistry , 3T3 Cells , Animals , Bone Morphogenetic Protein 7/chemistry , Cell Line , Cell Survival/drug effects , Coated Materials, Biocompatible/chemical synthesis , Coated Materials, Biocompatible/toxicity , Durapatite/toxicity , Mice , Nanocomposites/toxicity , Nanocomposites/ultrastructure , Osteoblasts/drug effects , Osteoblasts/physiology , Osteogenesis/drug effects
16.
Mater Sci Eng C Mater Biol Appl ; 63: 596-608, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27040255

ABSTRACT

Injectable chitosan microparticles were prepared using a simple coacervation method under physiologically friendly conditions by eliminating oil or toxic chemical, and employing low temperature and pressure for growth factor stability. Amount of 200 ng of bone morphogenetic protein-7 (BMP-7) was incorporated in the chitosan microparticles by two methods: encapsulating and coating techniques. These microparticles were tested in vivo to determine the biological response in a rat femoral bone defect at 6 and 12 weeks. Four groups (n=10) were tested which include two groups for BMP-7 incorporated microparticles (by two techniques), microparticles without BMP-7, and defect itself (negative control). Healthy bone formation was observed around the microparticles, which were only confined to the defect site and did not disperse. Histology indicated minor inflammatory response around the microparticles at 6 weeks, which reduced by 12 weeks. Micro-CT analysis of bone surface density and porosity was found to be significantly more (p<0.05) for microparticles containing groups, in comparison with controls, which suggests that the new bone formed in the presence of microparticles is more interconnected and porous. Collagen fibrils analysis conducted using multiphoton microscopy showed significant improvement in the formation of bundled collagen area (%) in microparticles containing groups in comparison with controls, indicating higher cross-linking between the fibrils. Microparticles were biocompatible and did not degrade in the 12 week implant period.


Subject(s)
Bone Morphogenetic Protein 7/pharmacology , Bone Regeneration/drug effects , Chitosan/chemistry , Drug Carriers/chemistry , Animals , Bone Density , Bone Diseases/drug therapy , Bone Morphogenetic Protein 7/administration & dosage , Bone Morphogenetic Protein 7/chemistry , Bone and Bones/diagnostic imaging , Bone and Bones/pathology , Microscopy, Fluorescence, Multiphoton , Porosity , Rats , X-Ray Microtomography
17.
Cardiovasc Res ; 110(3): 331-45, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27068510

ABSTRACT

AIMS: TGF-ß regulates tissue fibrosis: TGF-ß promotes fibrosis, whereas bone morphogenetic protein (BMP)-7 is antifibrotic. To demonstrate that (i) left ventricular (LV) remodelling after pressure overload is associated with disequilibrium in the signalling mediated by these cytokines, and (ii) BMP-7 exerts beneficial effects on LV remodelling and reverse remodelling. METHODS AND RESULTS: We studied patients with aortic stenosis (AS) and mice subjected to transverse aortic constriction (TAC) and TAC release (de-TAC). LV morphology and function were assessed by echocardiography. LV biopsies were analysed by qPCR, immunoblotting, and histology. Pressure overload reduced BMP-7 and pSmad1/5/8 and increased TGF-ß and pSmad2/3 in AS patients and TAC mice. BMP-7 correlated inversely with collagen, fibronectin, and ß-MHC expressions, and with hypertrophy and diastolic dysfunction, and directly with the systolic function. Multiple linear regression disclosed BMP-7 and TGF-ß as hypertrophy predictors, negative and positive, respectively. BMP-7 prevented TGF-ß-elicited hypertrophic program in cardiomyocytes, and Col1A1 promoter activity in NIH-3T3 fibroblasts. The treatment of TAC mice with rBMP-7 attenuated the development of structural damage and dysfunction, and halted ongoing remodelling. The reverse remodelling after pressure overload release was facilitated by rBMP-7, and hampered by disrupting BMP-7 function using a neutralizing antibody or genetic deletion. CONCLUSION: The disequilibrium between BMP-7 and TGF-ß signals plays a relevant role in the LV remodelling response to haemodynamic stress in TAC mice and AS patients. Our observations may provide new important insights aimed at developing novel therapies designed to prevent, halt, or reverse LV pathological remodelling in pressure overload cardiomyopathy.


Subject(s)
Bone Morphogenetic Protein 7/analysis , Bone Morphogenetic Protein 7/metabolism , Hypertrophy, Left Ventricular/prevention & control , Myocytes, Cardiac/metabolism , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left , Ventricular Remodeling , Aged , Aged, 80 and over , Animals , Aortic Valve Stenosis/complications , Bone Morphogenetic Protein 7/administration & dosage , Bone Morphogenetic Protein 7/deficiency , Bone Morphogenetic Protein 7/genetics , Case-Control Studies , Collagen/metabolism , Disease Models, Animal , Female , Fibronectins/metabolism , Fibrosis , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Myosin Heavy Chains/metabolism , NIH 3T3 Cells , Rats, Wistar , Recombinant Proteins/administration & dosage , Signal Transduction , Smad Proteins/metabolism , Time Factors , Transforming Growth Factor beta1/metabolism , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
18.
J Cell Physiol ; 231(7): 1601-10, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26574905

ABSTRACT

Endothelial and epithelial cell transition to a mesenchymal phenotype was identified as cellular paradigms implicated in the appearance of fibroblasts and development of reactive fibrosis in interstitial lung disease. The intermediate filament protein nestin was highly expressed in fibrotic tissue, detected in fibroblasts and participated in proliferation and migration. The present study tested the hypothesis that the transition of endothelial and epithelial cells to a mesenchymal phenotype was delineated by nestin expression. Three weeks following hypobaric hypoxia, adult male Sprague-Dawley rats characterized by alveolar and perivascular lung fibrosis were associated with increased nestin protein and mRNA levels and marked appearance of nestin/collagen type I((+))-fibroblasts. In the perivascular region of hypobaric hypoxic rats, displaced CD31((+))-endothelial cells were detected, exhibited a mesenchymal phenotype and co-expressed nestin. Likewise, epithelial cells in the lungs of hypobaric hypoxic rats transitioned to a mesenchymal phenotype distinguished by the co-expression of E-cadherin and collagen. Following the removal of FBS from primary passage rat alveolar epithelial cells, TGF-ß1 was detected in the media and a subpopulation acquired a mesenchymal phenotype characterized by E-cadherin downregulation and concomitant induction of collagen and nestin. Bone morphogenic protein-7 treatment of alveolar epithelial cells prevented E-cadherin downregulation, suppressed collagen induction but partially inhibited nestin expression. These data support the premise that the transition of endothelial and epithelial cells to a mesenchymal cell may have contributed in part to the appearance nestin/collagen type I((+))-fibroblasts and the reactive fibrotic response in the lungs of hypobaric hypoxic rats.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Hypoxia/genetics , Nestin/biosynthesis , Pulmonary Fibrosis/genetics , Animals , Bone Morphogenetic Protein 7/administration & dosage , Cadherins/biosynthesis , Cell Differentiation/genetics , Cell Line , Collagen Type I/biosynthesis , Collagen Type I/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts , Gene Expression Regulation/drug effects , Hypoxia/pathology , Nestin/genetics , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/pathology , RNA, Messenger/biosynthesis , Rats , Transforming Growth Factor beta1
19.
Hum Exp Toxicol ; 35(1): 69-77, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25733726

ABSTRACT

The epithelial-mesenchymal transition (EMT) is a critical process in the pulmonary fibrosis. It has been reported that bone morphogenetic protein 7 (BMP-7) was able to reverse EMT in proximal tubular cells. Therefore, we test the hypothesis that EMT contributes to silica-induced pulmonary fibrosis and BMP-7 inhibits EMT in silica-induced pulmonary fibrosis. Progressive silica-induced pulmonary fibrosis in the rat was used as a model of silicosis. Epithelial and mesenchymal markers were measured from rat fibrotic lungs. Then the effects of BMP-7 on the EMT were further confirmed in A549 cells. There are increases of vimentin as a mesenchymal marker and decreases of E-cadherin as an epithelial marker in the silica-exposed rat lungs, which is in agreement with the A549 cells data. However, BMP-7 treatment significantly reduced expression of vimentin in the rat pulmonary fibrosis model and in A549 cells. In conclusion, EMT contributes to silica-induced pulmonary fibrosis. Meanwhile, the treatment of BMP-7 can inhibit silica-induced EMT in vitro and in vivo.


Subject(s)
Bone Morphogenetic Protein 7/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Pulmonary Fibrosis/chemically induced , Silicon Dioxide/toxicity , Silicosis/prevention & control , Animals , Bone Morphogenetic Protein 7/administration & dosage , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Lung/drug effects , Lung/pathology , Male , Rats , Rats, Wistar , Recombinant Proteins , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Vimentin/genetics , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL