Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Tissue Eng Part C Methods ; 30(2): 53-62, 2024 02.
Article in English | MEDLINE | ID: mdl-38019085

ABSTRACT

The effect and mechanism of type III recombinant humanized collagen (hCOLIII) on human vascular endothelial EA.hy926 cells at the cellular and molecular levels were investigated. The impact of hCOLIII on the proliferation of EA.hy926 cells was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromid assay, the effect of hCOLIII on cell migration was investigated by scratch assay, the impact of hCOLIII on cell cycle and apoptosis was detected by flow cytometry, the ability of hCOLIII to induce angiogenesis of EA.hy926 cells was evaluated by angiogenesis assay, and the effect of hCOLIII on vascular endothelial growth factor (VEGF) expression was detected by real-time reverse transcription-polymerase chain reaction analysis. The hCOLIII at concentrations of 0.5, 0.25, and 0.125 mg/mL all showed specific effects on the proliferation and migration of human vascular endothelial cells. It could also affect the cell cycle, increase the proliferation index, and increase the expression level of VEGF in human vascular endothelial cells. In the meantime, hCOLIII at the concentration of 0.5 mg/mL also showed a promoting effect on vessel formation. hCOLIII can potentially promote the endothelization process of blood vessels, mainly by affecting the proliferation, migration, and vascular-like structure of human endothelial cells. At the same time, hCOLIII can promote the expression of VEGF. This collagen demonstrated its potential as a raw material for cardiovascular implants.


Subject(s)
Endothelial Cells , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Collagen Type III/metabolism , Collagen Type III/pharmacology , Collagen/pharmacology , Collagen/metabolism , Cell Movement , Cell Proliferation
2.
Zhonghua Nei Ke Za Zhi ; 62(7): 841-849, 2023 Jul 01.
Article in Chinese | MEDLINE | ID: mdl-37394854

ABSTRACT

Objective: JWH133, a cannabinoid type 2 receptor agonist, was tested for its ability to protect mice from bleomycin-induced pulmonary fibrosis. Methods: By using a random number generator, 24 C57BL/6J male mice were randomly divided into the control group, model group, JWH133 intervention group, and JWH133+a cannabinoid type-2 receptor antagonist (AM630) inhibitor group, with 6 mice in each group. A mouse pulmonary fibrosis model was established by tracheal instillation of bleomycin (5 mg/kg). Starting from the first day after modeling, the control group mice were intraperitoneally injected with 0.1 ml of 0.9% sodium chloride solution, and the model group mice were intraperitoneally injected with 0.1 ml of 0.9% sodium chloride solution. The JWH133 intervention group mice were intraperitoneally injected with 0.1 ml of JWH133 (2.5 mg/kg, dissolved in physiological saline), and the JWH133+AM630 antagonistic group mice were intraperitoneally injected with 0.1 ml of JWH133 (2.5 mg/kg) and AM630 (2.5 mg/kg). After 28 days, all mice were killed; the lung tissue was obtained, pathological changes were observed, and alveolar inflammation scores and Ashcroft scores were calculated. The content of type Ⅰ collagen in the lung tissue of the four groups of mice was measured using immunohistochemistry. The levels of interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) in the serum of the four groups of mice were measured using enzyme-linked immunosorbent assay (ELISA), and the content of hydroxyproline (HYP) in the lung tissue of the four groups of mice was measured. Western blotting was used to measure the protein expression levels of type Ⅲ collagen, α-smooth muscle actin (α-SMA), extracellular signal regulated kinase (ERK1/2), phosphorylated P-ERK1/2 (P-ERK1/2), and phosphorylated ribosome S6 kinase type 1 (P-p90RSK) in the lung tissue of mice in the four groups. Real-time quantitative polymerase chain reaction was used to measure the expression levels of collagen Ⅰ, collagen Ⅲ, and α-SMA mRNA in the lung tissue of the four groups of mice. Results: Compared with the control group, the pathological changes in the lung tissue of the model group mice worsened, with an increase in alveolar inflammation score (3.833±0.408 vs. 0.833±0.408, P<0.05), an increase in Ashcroft score (7.333±0.516 vs. 2.000±0.633, P<0.05), an increase in type Ⅰ collagen absorbance value (0.065±0.008 vs. 0.018±0.006, P<0.05), an increase in inflammatory cell infiltration, and an increase in hydroxyproline levels [(1.551±0.051) µg/mg vs. (0.974±0.060) µg/mg, P<0.05]. Compared with the model group, the JWH133 intervention group showed reduced pathological changes in lung tissue, decreased alveolar inflammation score (1.833±0.408, P<0.05), decreased Ashcroft score (4.167±0.753, P<0.05), decreased type Ⅰ collagen absorbance value (0.032±0.004, P<0.05), reduced inflammatory cell infiltration, and decreased hydroxyproline levels [(1.148±0.055) µg/mg, P<0.05]. Compared with the JWH133 intervention group, the JWH133+AM630 antagonistic group showed more severe pathological changes in the lung tissue of mice, increased alveolar inflammation score and Ashcroft score, increased type Ⅰ collagen absorbance value, increased inflammatory cell infiltration, and increased hydroxyproline levels. Compared with the control group, the expression of α-SMA, type Ⅲ collagen, P-ERK1/2, and P-p90RSK proteins in the lung tissue of the model group mice increased, while the expression of type Ⅰ collagen, type Ⅲ collagen, and α-SMA mRNA increased. Compared with the model group, the protein expression of α-SMA (relative expression 0.60±0.17 vs. 1.34±0.19, P<0.05), type Ⅲ collagen (relative expression 0.52±0.09 vs. 1.35±0.14, P<0.05), P-ERK1/2 (relative expression 0.32±0.11 vs. 1.14±0.14, P<0.05), and P-p90RSK (relative expression 0.43±0.14 vs. 1.15±0.07, P<0.05) decreased in the JWH133 intervention group. The type Ⅰ collagen mRNA (2.190±0.362 vs. 5.078±0.792, P<0.05), type Ⅲ collagen mRNA (1.750±0.290 vs. 4.935±0.456, P<0.05), and α-SMA mRNA (1.588±0.060 vs. 5.192±0.506, P<0.05) decreased. Compared with the JWH133 intervention group, the JWH133+AM630 antagonistic group increased the expression of α-SMA, type Ⅲ collagen, P-ERK1/2, and P-p90RSK protein in the lung tissue of mice, and increased the expression of type Ⅲ collagen and α-SMA mRNA. Conclusion: In mice with bleomycin-induced pulmonary fibrosis, the cannabinoid type-2 receptor agonist JWH133 inhibited inflammation and improved extracellular matrix deposition, which alleviated lung fibrosis. The underlying mechanism of action may be related to the activation of the ERK1/2-RSK1 signaling pathway.


Subject(s)
Cannabinoids , Pulmonary Fibrosis , Mice , Male , Animals , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Cannabinoid Receptor Agonists/adverse effects , Cannabinoid Receptor Agonists/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I/pharmacology , Collagen Type III/metabolism , Collagen Type III/pharmacology , Hydroxyproline/analysis , Hydroxyproline/metabolism , Hydroxyproline/pharmacology , Sodium Chloride/adverse effects , Sodium Chloride/metabolism , Mice, Inbred C57BL , Lung/pathology , Cannabinoids/adverse effects , Bleomycin/adverse effects , Bleomycin/metabolism , Collagen/adverse effects , Collagen/metabolism , Inflammation/pathology , RNA, Messenger/metabolism
3.
Int J Biol Macromol ; 243: 125130, 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37263336

ABSTRACT

Breast cancer (BC) has become the most common cancer in the world and lacks safe and efficient treatment. The novel biomaterial recombinant humanized collagen type III (rhCOLIII) has been reported to have various biological functions, such as promoting skin extracellular matrix regeneration and improving the cell microenvironment, but its role in breast cancer is unclear. In this study, we first found that rhCOLIII inhibited the proliferation, migration, and invasion of breast cancer cells (BCCs) but had no effect on the survival of normal breast epithelial cells. In addition, rhCOLIII not only promoted apoptosis and dormancy of BCCs but also inhibited autophagy within BCCs. Subsequently, RNA-Seq analysis suggested that DDR1 may be a key target for rhCOLIII to exert antitumor effects, and we validated that inhibition of DDR1 eliminated the effects of rhCOLIII on the proliferation, migration, apoptosis, dormancy and autophagy of BCCs. Moreover, rhCOLIII treatment was found to reduce the tumorigenic activity of BCCs in animal experiments and to upregulate DDR1 protein expression while inhibiting autophagy at the tissue level. Therefore, rhCOLIII may serve as a potential treatment method for BC patients and is expected to improve the prognosis of patients.


Subject(s)
Collagen Type III , Neoplasms , Animals , Cell Line, Tumor , Collagen Type III/pharmacology , Cell Proliferation , Apoptosis , Autophagy , Cell Movement
4.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-985994

ABSTRACT

Objective: JWH133, a cannabinoid type 2 receptor agonist, was tested for its ability to protect mice from bleomycin-induced pulmonary fibrosis. Methods: By using a random number generator, 24 C57BL/6J male mice were randomly divided into the control group, model group, JWH133 intervention group, and JWH133+a cannabinoid type-2 receptor antagonist (AM630) inhibitor group, with 6 mice in each group. A mouse pulmonary fibrosis model was established by tracheal instillation of bleomycin (5 mg/kg). Starting from the first day after modeling, the control group mice were intraperitoneally injected with 0.1 ml of 0.9% sodium chloride solution, and the model group mice were intraperitoneally injected with 0.1 ml of 0.9% sodium chloride solution. The JWH133 intervention group mice were intraperitoneally injected with 0.1 ml of JWH133 (2.5 mg/kg, dissolved in physiological saline), and the JWH133+AM630 antagonistic group mice were intraperitoneally injected with 0.1 ml of JWH133 (2.5 mg/kg) and AM630 (2.5 mg/kg). After 28 days, all mice were killed; the lung tissue was obtained, pathological changes were observed, and alveolar inflammation scores and Ashcroft scores were calculated. The content of type Ⅰ collagen in the lung tissue of the four groups of mice was measured using immunohistochemistry. The levels of interleukin 6 (IL-6) and tumor necrosis factor α (TNF-α) in the serum of the four groups of mice were measured using enzyme-linked immunosorbent assay (ELISA), and the content of hydroxyproline (HYP) in the lung tissue of the four groups of mice was measured. Western blotting was used to measure the protein expression levels of type Ⅲ collagen, α-smooth muscle actin (α-SMA), extracellular signal regulated kinase (ERK1/2), phosphorylated P-ERK1/2 (P-ERK1/2), and phosphorylated ribosome S6 kinase type 1 (P-p90RSK) in the lung tissue of mice in the four groups. Real-time quantitative polymerase chain reaction was used to measure the expression levels of collagen Ⅰ, collagen Ⅲ, and α-SMA mRNA in the lung tissue of the four groups of mice. Results: Compared with the control group, the pathological changes in the lung tissue of the model group mice worsened, with an increase in alveolar inflammation score (3.833±0.408 vs. 0.833±0.408, P<0.05), an increase in Ashcroft score (7.333±0.516 vs. 2.000±0.633, P<0.05), an increase in type Ⅰ collagen absorbance value (0.065±0.008 vs. 0.018±0.006, P<0.05), an increase in inflammatory cell infiltration, and an increase in hydroxyproline levels [(1.551±0.051) μg/mg vs. (0.974±0.060) μg/mg, P<0.05]. Compared with the model group, the JWH133 intervention group showed reduced pathological changes in lung tissue, decreased alveolar inflammation score (1.833±0.408, P<0.05), decreased Ashcroft score (4.167±0.753, P<0.05), decreased type Ⅰ collagen absorbance value (0.032±0.004, P<0.05), reduced inflammatory cell infiltration, and decreased hydroxyproline levels [(1.148±0.055) μg/mg, P<0.05]. Compared with the JWH133 intervention group, the JWH133+AM630 antagonistic group showed more severe pathological changes in the lung tissue of mice, increased alveolar inflammation score and Ashcroft score, increased type Ⅰ collagen absorbance value, increased inflammatory cell infiltration, and increased hydroxyproline levels. Compared with the control group, the expression of α-SMA, type Ⅲ collagen, P-ERK1/2, and P-p90RSK proteins in the lung tissue of the model group mice increased, while the expression of type Ⅰ collagen, type Ⅲ collagen, and α-SMA mRNA increased. Compared with the model group, the protein expression of α-SMA (relative expression 0.60±0.17 vs. 1.34±0.19, P<0.05), type Ⅲ collagen (relative expression 0.52±0.09 vs. 1.35±0.14, P<0.05), P-ERK1/2 (relative expression 0.32±0.11 vs. 1.14±0.14, P<0.05), and P-p90RSK (relative expression 0.43±0.14 vs. 1.15±0.07, P<0.05) decreased in the JWH133 intervention group. The type Ⅰ collagen mRNA (2.190±0.362 vs. 5.078±0.792, P<0.05), type Ⅲ collagen mRNA (1.750±0.290 vs. 4.935±0.456, P<0.05), and α-SMA mRNA (1.588±0.060 vs. 5.192±0.506, P<0.05) decreased. Compared with the JWH133 intervention group, the JWH133+AM630 antagonistic group increased the expression of α-SMA, type Ⅲ collagen, P-ERK1/2, and P-p90RSK protein in the lung tissue of mice, and increased the expression of type Ⅲ collagen and α-SMA mRNA. Conclusion: In mice with bleomycin-induced pulmonary fibrosis, the cannabinoid type-2 receptor agonist JWH133 inhibited inflammation and improved extracellular matrix deposition, which alleviated lung fibrosis. The underlying mechanism of action may be related to the activation of the ERK1/2-RSK1 signaling pathway.


Subject(s)
Mice , Male , Animals , Pulmonary Fibrosis/pathology , Cannabinoid Receptor Agonists/metabolism , Collagen Type I/pharmacology , Collagen Type III/pharmacology , Hydroxyproline/pharmacology , Sodium Chloride/metabolism , Mice, Inbred C57BL , Lung/pathology , Cannabinoids/adverse effects , Bleomycin/metabolism , Collagen/metabolism , Inflammation/pathology , RNA, Messenger/metabolism
6.
J Diabetes Investig ; 13(6): 955-964, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35098679

ABSTRACT

AIMS/INTRODUCTION: Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have been shown to display excellent renoprotective effects in diabetic kidney disease with macroalbuminuria/proteinuria. Regarding the renoprotective mechanism of SGLT2i, a sophisticated hypothesis was made by explaining the suppression of glomerular hypertension/hyperfiltration through the adenosine/adenosine type 1 receptor (A1R) signaling-mediated restoration of the tubuloglomerular feedback mechanism; however, how such A1R signaling is relevant for renoprotection by SGLT2i in diabetic kidney disease with proteinuria has not been elucidated. MATERIALS AND METHODS: Streptozotocin-induced diabetic CD-1 mice were injected with bovine serum albumin (BSA) and treated with SGLT2i in the presence/absence of A1R inhibitor administration. RESULTS: We found that the influences of SGLT2i are essentially independent of the activation of A1R signaling in the kidney of BSA-overloaded streptozotocin-induced diabetic mice. BSA-overloaded diabetic mice showed the trend of kidney damage with higher glomerular filtration rate (GFR) and the significant induction of fibrogenic genes, such as transforming growth factor-ß2 and collagen type III. SGLT2i TA-1887 suppressed diabetes-induced GFR in BSA-overloaded diabetic mice was associated with the significant suppression of transforming growth factor-ß2 and collagen type III; A1R-specific inhibitor 8-cyclopentyl-1,3-dipropylxanthine did not cancel the effects of TA-1887 on either GFR or associated gene levels. Both TA-1887 and 8-cyclopentyl-1,3-dipropylxanthine-treated BSA-overloaded diabetic mice showed suppressed glycated hemoglobin levels associated with the increased food intake. When analyzing the association among histological evaluation, GFR and potential fibrogenic gene levels, each group of mice showed distinct correlation patterns. CONCLUSIONS: A1R signaling activation was not the dominant mechanism on the influence of SGLT2i in the kidney of BSA-overloaded diabetic mice.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Nephropathies , Receptors, Purinergic P1/metabolism , Sodium-Glucose Transporter 2 Inhibitors , Adenosine/metabolism , Adenosine/pharmacology , Animals , Collagen Type III/metabolism , Collagen Type III/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Glucose/metabolism , Humans , Kidney , Mice , Proteinuria/metabolism , Serum Albumin, Bovine/metabolism , Serum Albumin, Bovine/pharmacology , Signal Transduction , Sodium/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Streptozocin , Transforming Growth Factors/metabolism , Transforming Growth Factors/pharmacology
7.
Nat Commun ; 10(1): 4866, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31653830

ABSTRACT

Despite the success of current therapies for acute myocardial infarction (MI), many patients still develop adverse cardiac remodeling and heart failure. With the growing prevalence of heart failure, a new therapy is needed that can prevent remodeling and support tissue repair. Herein, we report on injectable recombinant human collagen type I (rHCI) and type III (rHCIII) matrices for treating MI. Injecting rHCI or rHCIII matrices in mice during the late proliferative phase post-MI restores the myocardium's mechanical properties and reduces scar size, but only the rHCI matrix maintains remote wall thickness and prevents heart enlargement. rHCI treatment increases cardiomyocyte and capillary numbers in the border zone and the presence of pro-wound healing macrophages in the ischemic area, while reducing the overall recruitment of bone marrow monocytes. Our findings show functional recovery post-MI using rHCI by promoting a healing environment, cardiomyocyte survival, and less pathological remodeling of the myocardium.


Subject(s)
Collagen Type III/pharmacology , Collagen Type I/pharmacology , Heart/drug effects , Myocardial Infarction/pathology , Recombinant Proteins/pharmacology , Ventricular Function/drug effects , Ventricular Remodeling/drug effects , Animals , Capillaries/drug effects , Carbodiimides/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cicatrix/pathology , Coronary Vessels/drug effects , Cross-Linking Reagents/pharmacology , Dimethylamines/pharmacology , Humans , Macrophages/drug effects , Mice , Monocytes/drug effects , Myocardial Infarction/physiopathology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Succinimides/pharmacology
8.
Acta Cir Bras ; 33(2): 144-155, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29513813

ABSTRACT

PURPOSE: To evaluate the efficacy of the application of the human amniotic membrane (HAM) on the inflammatory process, fibroblast proliferation, formation of collagenand reduction of skin wound areas in rats. METHODS: Thirty six rats were submitted to a surgical injury induction and divided into two groups (n = 18): group C (control) and T (treated with the HAM). The macroscopic evolution in the wound area and the histological characteristics of the skin samples were evaluated. RESULTS: The regression of the wound area was greater in group T. The histological analysis revealed a significant reduction (p < 0.05) in the inflammatory infiltrate in group T at all experimental periods compared with that in the control group. Furthermore, the group T presented a significant increase in the proliferation of fibroblasts at 14 and 21 days compared with group C (p < 0.05). Regarding the deposition of mature collagen fibers, there was an increase in the replacement of type III collagen by type I collagen in group T (p < 0.05). CONCLUSION: Treatment with the HAM reduced the healing time as well as the inflammatory responses, increased the proliferation of fibroblasts, and induced a higher concentration of mature collagen fibers.


Subject(s)
Amnion/transplantation , Biological Dressings , Collagen/pharmacology , Skin/injuries , Wound Healing/physiology , Amnion/chemistry , Animals , Collagen Type I/metabolism , Collagen Type I/pharmacology , Collagen Type III/metabolism , Collagen Type III/pharmacology , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Inflammation/metabolism , Male , Random Allocation , Rats , Rats, Wistar , Skin/pathology , Wound Healing/drug effects
9.
Acta cir. bras ; 33(2): 144-155, Feb. 2018. graf
Article in English | LILACS | ID: biblio-886256

ABSTRACT

Abstract Purpose: To evaluate the efficacy of the application of the human amniotic membrane (HAM) on the inflammatory process, fibroblast proliferation, formation of collagenand reduction of skin wound areas in rats. Methods: Thirty six rats were submitted to a surgical injury induction and divided into two groups (n = 18): group C (control) and T (treated with the HAM). The macroscopic evolution in the wound area and the histological characteristics of the skin samples were evaluated. Results: The regression of the wound area was greater in group T. The histological analysis revealed a significant reduction (p < 0.05) in the inflammatory infiltrate in group T at all experimental periods compared with that in the control group. Furthermore, the group T presented a significant increase in the proliferation of fibroblasts at 14 and 21 days compared with group C (p < 0.05). Regarding the deposition of mature collagen fibers, there was an increase in the replacement of type III collagen by type I collagen in group T (p < 0.05). Conclusion: Treatment with the HAM reduced the healing time as well as the inflammatory responses, increased the proliferation of fibroblasts, and induced a higher concentration of mature collagen fibers.


Subject(s)
Humans , Animals , Male , Rats , Skin/injuries , Wound Healing/physiology , Biological Dressings , Collagen/pharmacology , Amnion/transplantation , Skin/pathology , Wound Healing/drug effects , Random Allocation , Rats, Wistar , Collagen Type I/metabolism , Collagen Type I/pharmacology , Collagen Type III/metabolism , Collagen Type III/pharmacology , Disease Models, Animal , Fibroblasts/metabolism , Fibroblasts/pathology , Amnion/chemistry , Inflammation/metabolism
10.
Protein J ; 36(4): 322-331, 2017 08.
Article in English | MEDLINE | ID: mdl-28589291

ABSTRACT

High-level expression of recombinant collagen by genetic engineering is urgently required. Recombinant collagen is different from natural collagen in its hydroxyproline (Hyp) content and thermal stability. To obtain hydroxylated collagen for applications in biomedicine and biomaterials, the human collagen α1(III) chain was co-expressed with the viral prolyl 4-hydroxylase A085R in Escherichia coli. Unlike previous reports using human prolyl 4-hydroxylase, this study examined the hydroxylation of full-length human collagen α1(III) chain (COL3A1) by viral prolyl 4-hydroxylase. The genes encoding these two proteins were controlled by different promoters, Ptac and PRPL, on a recombinant pKK223-3 plasmid. The sequencing results verified that the target genes were successfully inserted into the recombinant vector. Based on quantitative PCR, SDS-PAGE, and western blotting, successful expression by E. coli BL21(DE3) was detected at the mRNA and protein levels for both loci. Liquid chromatography-mass spectrometry (LC-MS/MS) results suggested that the highest Hyp yield was obtained when the two proteins were induced with 0.5 mM IPTG and heat-shock treatment at 50 °C, corresponding to high enzyme expression and low human collagen α1(III) chain expression levels. A biological activity analysis indicated that the recombinant collagen with the highest hydroxylation level supported the growth of baby hamster kidney cells, similar to observations for native collagen. The production of hydroxylated collagen in this study establishes a new method for collagen hydroxylation and provides a basis for the application of recombinant collagen expressed in E. coli.


Subject(s)
Collagen Type III/metabolism , Escherichia coli/metabolism , Plasmids/metabolism , Prolyl Hydroxylases/metabolism , Recombinant Proteins/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Proliferation/drug effects , Collagen Type III/chemistry , Collagen Type III/genetics , Collagen Type III/pharmacology , Cricetinae , Escherichia coli/genetics , Gene Expression , Humans , Hydroxylation , Phycodnaviridae/chemistry , Phycodnaviridae/enzymology , Plasmids/chemistry , Prolyl Hydroxylases/genetics , Promoter Regions, Genetic , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Sequence Alignment , Transformation, Bacterial , Viral Proteins/genetics
11.
Ann Otol Rhinol Laryngol ; 125(2): 115-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26276144

ABSTRACT

OBJECTIVE: We developed an in situ regeneration-inducible artificial trachea composed of a porcine collagen sponge and polypropylene framework and used it for tracheal reconstruction. In the present study, collagen sponges with different structures were prepared from various concentrations of collagen solutions, and their effect on the regeneration of tracheal epithelium was examined. METHODS: Collagen sponges were prepared from type I and III collagen solutions. The structures of the sponges were analyzed using scanning electron microscopy (SEM). Artificial tracheae, which were formed using the collagen sponges with different structures, were implanted into rabbits, and regeneration of the tracheal epithelium on the artificial tracheae was evaluated by SEM analysis and histological examination. RESULTS: The SEM analysis showed that collagen sponges prepared from 0.5% and 1.0% collagen solutions had a porous structure. However, the sponges prepared from a 1.5% collagen solution had a nonporous structure. After implantation of artificial tracheae prepared from 0.5% and 1.0% collagen solutions, their luminal surfaces were mostly covered with epithelium within 14 days. However, epithelial reorganization occurred later on artificial tracheae prepared from the 1.5% collagen solution. CONCLUSION: Collagen sponges with a porous structure are suitable for regeneration of the tracheal epithelium in our artificial trachea.


Subject(s)
Artificial Organs , Collagen Type III/pharmacology , Collagen Type I/pharmacology , Guided Tissue Regeneration/methods , Plastic Surgery Procedures/methods , Respiratory Mucosa/surgery , Tissue Scaffolds , Trachea , Animals , Biocompatible Materials/pharmacology , Materials Testing/methods , Microscopy, Electron, Scanning , Models, Animal , Polypropylenes/pharmacology , Rabbits , Swine , Tissue Engineering/methods
12.
Osteoarthritis Cartilage ; 23(4): 648-60, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25575968

ABSTRACT

UNLABELLED: Autologous chondrocyte implantation (ACI) has improved outcome in long-term studies of joint repair in man. However, ACI requires sutured periosteal flaps to secure the cells, which precludes minimally-invasive implantation, and introduces complications with arthrofibrosis and graft hypertrophy. This study evaluated ACI on a collagen type I/III scaffold (matrix-induced autologous chondrocyte implantation; MACI(®)) in critical sized defects in the equine model. METHODS: Chondrocytes were isolated from horses, expanded and seeded onto a collagen I/III membrane (ACI-Maix™) and implanted into one of two 15-mm defects in the femoral trochlear ridge of six horses. Control defects remained empty as ungrafted debrided defects. The animals were examined daily, scored by second look arthroscopy at 12 weeks, and necropsy examination 6 months after implantation. Reaction to the implant was determined by lameness, and synovial fluid constituents and synovial membrane histology. Cartilage healing was assessed by arthroscopic scores, gross assessment, repair tissue histology and immunohistochemistry, cartilage glycosaminoglycan (GAG) and DNA assay, and mechanical testing. RESULTS: MACI(®) implanted defects had improved arthroscopic second-look, gross healing, and composite histologic scores, compared to spontaneously healing empty defects. Cartilage GAG and DNA content in the defects repaired by MACI implant were significantly improved compared to controls. Mechanical properties were improved but remained inferior to normal cartilage. There was minimal evidence of reaction to the implant in the synovial fluid, synovial membrane, subchondral bone, or cartilage. CONCLUSIONS: The MACI(®) implant appeared to improve cartilage healing in a critical sized defect in the equine model evaluated over 6 months.


Subject(s)
Cartilage, Articular/physiology , Cell Transplantation/methods , Chondrocytes/transplantation , Collagen Type III/pharmacology , Collagen Type I/pharmacology , Patellofemoral Joint/injuries , Wound Healing/drug effects , Animals , Arthroscopy , Biomechanical Phenomena/physiology , Biopsy , Cartilage, Articular/drug effects , Cell Survival , Cells, Cultured , Chondrocytes/pathology , Collagen Type I/administration & dosage , Collagen Type III/administration & dosage , Disease Models, Animal , Glycosaminoglycans/physiology , Horses , Humans , In Vitro Techniques , Patellofemoral Joint/physiopathology , Treatment Outcome , Wound Healing/physiology
13.
Biotechnol Appl Biochem ; 62(4): 467-75, 2015.
Article in English | MEDLINE | ID: mdl-25231012

ABSTRACT

Recombinant collagen and gelatin have been applied in biomedical materials field because of products from genetically engineered microorganisms with improved safety, traceability, reproducibility, and homogeneous quality. To obtain high-level secretory expression of single-chain full-length human collagen α1(III) chain (COL3A1) without the N and C telopeptides, the cDNA coding for the human COL3A1 gene was cloned into the secretory expression vector pPIC9K and integrated into Pichia pastoris GS115. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis and Western blotting analysis of culture supernatant from the recombinant methylotrophic yeast suggested that the unhydroxylated recombinant human COL3A1 (rhCOL3A1) was secreted into the culture medium, and exhibited an apparent molecular mass of approximately 130 kDa, which is 1.4 times higher than the theoretical one. Finally, the unhydroxylated rhCOL3A1 was purified to greater than 90% purity using a four-step approach. In addition, methylthiazolydiphenyl-tetrazolium bromide experiments indicated that low concentration of rhCOL3A1 could promote Baby hamster kidney cell (BHK21) proliferation effectively. The production and purification of rhCOL3A1 described in this study offer a new method for obtaining high level of rhCOL3A1 in relatively pure form, which is suitable for biomedical materials application.


Subject(s)
Collagen Type III/biosynthesis , Gene Expression , Pichia/metabolism , Animals , Cell Line , Cell Proliferation/drug effects , Cloning, Molecular , Collagen Type III/chemistry , Collagen Type III/pharmacology , Cricetinae , Genetic Vectors/genetics , Humans , Pichia/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology
14.
J Tissue Eng Regen Med ; 9(12): 1386-93, 2015 Dec.
Article in English | MEDLINE | ID: mdl-23362205

ABSTRACT

Complex skin wounds, such as chronic ulcers and deep burns, require lengthy treatments and cause extensive burdens on healthcare and the economy. Use of biomaterials and cell transplantation may improve traditional treatments and promote the healing of difficult-to-treat wounds. In this study, we investigated the use of recombinant human collagen III (rhCol-III) gel as a delivery vehicle for cultured autologous skin cells (keratinocytes only or keratinocyte-fibroblast mixtures). We examined its effect on the healing of full-thickness wounds in a porcine wound-healing model. Two Landrace pigs were used for the study. Fourteen deep dermal wounds were created on the back of each pig with an 8 mm biopsy punch. Syringes containing acellular rhCol-III gel (n = 8) or rhCol-III gel with autologous keratinocytes (n = 8) or rhCol-III gel with autologous keratinocytes and fibroblasts (n = 8) were applied into wounds. Untreated wounds were used as controls for the treatment groups (n = 4). We used rhCol-III gel to manufacture a cell-delivery syringe containing autologous skin cells. In a full-thickness wound-healing model, we observed that rhCol-III gel enhances early granulation tissue formation. Interestingly, we found cell type-dependent differences in the stability of rhCol-III in vivo. Fibroblast-containing gel was effectively removed from the wound, whereas gels without cells or with keratinocytes only remained intact. Our results demonstrate that the properties of rhCol-III gel for skin cell transplantation can be significantly altered in a cell type-dependent manner.


Subject(s)
Burns/therapy , Cell Transplantation/methods , Collagen Type III/pharmacology , Fibroblasts/transplantation , Keratinocytes/transplantation , Wound Healing/drug effects , Animals , Autografts , Burns/metabolism , Burns/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Recombinant Proteins/pharmacology , Swine
15.
PLoS One ; 9(6): e100043, 2014.
Article in English | MEDLINE | ID: mdl-24949629

ABSTRACT

GPR56 is a member of the adhesion G protein-coupled receptor (GPCR) family. Despite the importance of GPR56 in brain development, where mutations cause a devastating human brain malformation called bilateral frontoparietal polymicrogyria (BFPP), the signaling mechanism(s) remain largely unknown. Like many other adhesion GPCRs, GPR56 is cleaved via a GPCR autoproteolysis-inducing (GAIN) domain into N- and C-terminal fragments (GPR56N and GPR56C); however, the biological significance of this cleavage is elusive. Taking advantage of the recent identification of a GPR56 ligand and the presence of BFPP-associated mutations, we investigated the molecular mechanism of GPR56 signaling. We demonstrate that ligand binding releases GPR56N from the membrane-bound GPR56C and triggers the association of GPR56C with lipid rafts and RhoA activation. Furthermore, one of the BFPP-associated mutations, L640R, does not affect collagen III-induced lipid raft association of GPR56. Instead, it specifically abolishes collagen III-mediated RhoA activation. Together, these findings reveal a novel signaling mechanism that may apply to other members of the adhesion GPCR family.


Subject(s)
Collagen Type III/pharmacology , Receptors, G-Protein-Coupled/metabolism , rhoA GTP-Binding Protein/metabolism , Amino Acid Sequence , Animals , Conserved Sequence , Enzyme Activation/drug effects , Evolution, Molecular , HEK293 Cells , Humans , Ligands , Malformations of Cortical Development/genetics , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Protein Structure, Secondary , Protein Transport/drug effects , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects
16.
J Appl Oral Sci ; 22(1): 29-37, 2014.
Article in English | MEDLINE | ID: mdl-24626246

ABSTRACT

OBJECTIVE: To analyze Mucograft®(MG), a recently introduced collagen matrix, in vitro and in vivo, and compare it with BioGide®(BG), a well-established collagen membrane, as control. MATERIAL AND METHODS: A detailed analysis of the materials surface and ultra-structure was performed. Cellular growth patterns and proliferation rates of human fibroblasts on MG and BG were analyzed in vitro. In addition, the early tissue reaction of CD-1 mouse to these materials was analyzed by means of histological and histomorphometrical analysis. RESULTS: MG showed a three-fold higher thickness both in dry and wet conditions, when compared to BG. The spongy surface of BG significantly differed from that of MG. Cells showed a characteristic proliferation pattern on the different materials in vitro. Fibroblasts tended to proliferate on the compact layers of both collagens, with the highest values on the compact side of BG. In vivo, at day three both materials demonstrated good tissue integration, with a mononuclear cell sheet of fibroblasts on all surfaces, however, without penetrating into the materials. CONCLUSIONS: The findings of this study showed that MG and BG facilitate cell proliferation on both of their surfaces in vitro. In vivo, these two materials induce a comparable early tissue reaction, while serving as cell occlusive barriers.


Subject(s)
Biocompatible Materials/pharmacology , Cell Proliferation , Collagen Type III/pharmacology , Collagen Type I/pharmacology , Fibroblasts/cytology , Animals , Cell Survival , Cells, Cultured , Collagen/pharmacology , Female , Humans , Immunohistochemistry , Materials Testing , Mice , Random Allocation , Reproducibility of Results , Surface Properties , Time Factors
17.
J. appl. oral sci ; 22(1): 29-37, Jan-Feb/2014. graf
Article in English | LILACS, BBO - Dentistry | ID: lil-699912

ABSTRACT

Objective: To analyze Mucograft®(MG), a recently introduced collagen matrix, in vitro and in vivo, and compare it with BioGide®(BG), a well-established collagen membrane, as control. Material and Methods: A detailed analysis of the materials surface and ultra-structure was performed. Cellular growth patterns and proliferation rates of human fibroblasts on MG and BG were analyzed in vitro. In addition, the early tissue reaction of CD-1 mouse to these materials was analyzed by means of histological and histomorphometrical analysis. Results: MG showed a three-fold higher thickness both in dry and wet conditions, when compared to BG. The spongy surface of BG significantly differed from that of MG. Cells showed a characteristic proliferation pattern on the different materials in vitro. Fibroblasts tended to proliferate on the compact layers of both collagens, with the highest values on the compact side of BG. In vivo, at day three both materials demonstrated good tissue integration, with a mononuclear cell sheet of fibroblasts on all surfaces, however, without penetrating into the materials. Conclusions: The findings of this study showed that MG and BG facilitate cell proliferation on both of their surfaces in vitro. In vivo, these two materials induce a comparable early tissue reaction, while serving as cell occlusive barriers. .


Subject(s)
Humans , Animals , Female , Mice , Biocompatible Materials/pharmacology , Cell Proliferation , Collagen Type I/pharmacology , Collagen Type III/pharmacology , Fibroblasts/cytology , Cell Survival , Cells, Cultured , Collagen/pharmacology , Immunohistochemistry , Materials Testing , Random Allocation , Reproducibility of Results , Surface Properties , Time Factors
18.
PLoS One ; 8(3): e59386, 2013.
Article in English | MEDLINE | ID: mdl-23516631

ABSTRACT

Enterohemorrhagic Escherichia coli (EHEC) O157:H7 are responsible for repeated food-poisoning cases often caused by contaminated burgers. EHEC infection is predominantly a pediatric illness, which can lead to life-threatening diseases. Ruminants are the main natural reservoir for EHEC and food contamination almost always originates from faecal contamination. In beef meat products, primary bacterial contamination occurs at the dehiding stage of slaughtering. The extracellular matrix (ECM) is the most exposed part of the skeletal muscles in beef carcasses. Investigating the adhesion to the main muscle fibrous ECM proteins, insoluble fibronectin, collagen I, III and IV, laminin-α2 and elastin, results demonstrated that the preceding growth conditions had a great influence on subsequent bacterial attachment. In the tested experimental conditions, maximal adhesion to fibril-forming collagens I or III occurred at 25°C and pH 7. Once initially adhered, exposure to lower temperatures, as applied to meat during cutting and storage, or acidification, as in the course of post-mortem physiological modifications of muscle, had no effect on detachment, except at pHu. In addition, dense biofilm formation occurred on immobilized collagen I or III and was induced in growth medium supplemented with collagen I in solution. From this first comprehensive investigation of EHEC adhesion to ECM proteins with respect to muscle biology and meat processing, new research directions for the development of innovative practices to minimize the risk of meat contamination are further discussed.


Subject(s)
Collagen Type III/pharmacology , Collagen Type I/pharmacology , Extracellular Matrix/metabolism , Animals , Biofilms/drug effects , Cattle , Culture Media/pharmacology , Elastin/metabolism , Escherichia coli/metabolism , Fibronectins/metabolism , Hydrogen-Ion Concentration , Laminin/metabolism , Temperature
19.
Biochim Biophys Acta ; 1820(10): 1576-87, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22705941

ABSTRACT

BACKGROUND: Alternative splicing of EDA fibronectin (FN) region is a cell type- and development-regulated mechanism controlled by pathological processes, growth factors and extracellular matrix (ECM). Classic and vascular Ehlers-Danlos syndrome (cEDS and vEDS) are connective tissue disorders caused by COL5A1/COL5A2 and COL3A1 gene mutations, leading to an in vivo abnormal collagen fibrillogenesis and to an in vitro defective organisation in the ECM of type V (COLLV) and type III collagen (COLLIII). These defects induce the FN-ECM disarray and the decrease of COLLs and FN receptors, the α2ß1 and α5ß1 integrins. Purified COLLV and COLLIII restore the COLL-FN-ECMs in both EDS cell strains. METHODS: Real-time PCR, immunofluorescence microscopy, and Western blotting were used to investigate the effects of COLLs on FN1 gene expression, EDA region alternative splicing, EDA(+)-FN-ECM assembly, α5ß1 integrin and EDA(+)-FN-specific α9 integrin subunit organisation, α5ß1 integrin and FAK co-regulation in EDS fibroblasts. RESULTS: COLLV-treated cEDS and COLLIII-treated vEDS fibroblasts up-regulate the FN1 gene expression, modulate the EDA(+) mRNA maturation and increase the EDA(+)-FN levels, thus restoring a control-like FN-ECM, which elicits the EDA(+)-FN-specific α9ß1 integrin organisation, recruits the α5ß1 integrin and switches on the FAK binding and phosphorylation. CONCLUSION: COLLs regulate the EDA(+)-FN-ECM organisation at transcriptional and post-transcriptional level and activate the α5ß1-FAK complexes. COLLs also recruit the α9ß1 integrin involved in the assembly of the EDA(+)-FN-ECM in EDS cells. GENERAL SIGNIFICANCE: The knowledge of the COLLs-ECM role in FN isotype expression and in EDA(+)-FN-ECM-mediated signal transduction adds insights in the ECM remodelling mechanisms in EDS cells.


Subject(s)
Collagen Type III/pharmacology , Collagen Type V/pharmacology , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Case-Control Studies , Cells, Cultured , Ehlers-Danlos Syndrome/metabolism , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/physiology , Gene Expression Regulation/drug effects , Humans , Integrin alpha5beta1/metabolism , Integrins/metabolism , Male , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization/drug effects
20.
Acta Biomater ; 8(8): 3061-72, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22561669

ABSTRACT

There are conflicting reports concerning the tissue reaction of small animals to porcine-based, non-cross-linked collagen I-III membranes/matrices for use in guided tissue/bone regeneration. The fast degradation of these membranes/matrices combined with transmembrane vascularization within 4 weeks has been observed in rats compared with the slow vascularization and continuous integration observed in mice. The aim of the present study was to analyze the tissue reaction to a porcine-based non-cross-linked collagen I-III membrane in mice. Using a subcutaneous implantation model, the membrane was implanted subcutaneously in mice for up to 60 days. The extent of scaffold vascularization, tissue integration and scaffold thickness were assessed using general and specialized histological methods, together with a unique histomorphometrical analysis technique. A dense Bombyx mori-derived silk fibroin membrane was used as a positive control, whilst a polytetrafluoroethylene (PTFE) membrane served as a negative control. Within the observation period, the collagen membrane induced a mononuclear cellular tissue response, including anti-inflammatory macrophages and the absence of multinucleated giant cells within its implantation bed. Transmembrane scaffold vascularization was not observed, whereas a mild scaffold vascularization was generated through microvessels located at both scaffold surfaces. However, the silk fibroin induced a mononuclear and multinucleated cell-based tissue response, in which pro-inflammatory macrophages and multinucleated giant cells were associated with an increasing transmembrane scaffold vascularization and a breakdown of the membrane within the experimental period. The PTFE membrane remained as a stable barrier throughout the study, and visible cellular degradation was not observed. However, multinucleated giant cells were located on both interfaces. The present study demonstrated that the tested non-cross-linked collagen membrane remained as a stable barrier membrane throughout the study period. The membrane integrated into the subcutaneous connective tissue and exhibited only a mild peripheral vascularization without experiencing breakdown. The silk fibroin, in contrast, induced granulation tissue formation, which resulted in its high vascularization and the breakdown of the material over time. The presence of multinucleated giant cells at both interfaces of the PFTE membrane is a sign of its slow cellular biodegradation and might lead to adhesions between the membrane and its surrounding tissue. This hypothesis could explain the observed clinical complications associated with the retrieval of these materials after guided tissue regeneration.


Subject(s)
Collagen Type III/pharmacology , Collagen Type I/pharmacology , Cross-Linking Reagents/pharmacology , Neovascularization, Physiologic/drug effects , Prosthesis Implantation , Animals , Female , Fibroins/ultrastructure , Immunohistochemistry , Membranes/drug effects , Membranes, Artificial , Mice , Microscopy, Electron, Scanning , Polytetrafluoroethylene/pharmacology , Rats , Sus scrofa , Tissue Scaffolds
SELECTION OF CITATIONS
SEARCH DETAIL
...