Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 474
Filter
1.
Carbohydr Polym ; 334: 122061, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38553245

ABSTRACT

The galactomannan-based gel from Cassia grandis seeds was used to incorporate Penicillium sp. UCP 1286 and commercial collagenases. Experiments were carried out according to a 23-full factorial design to identify the most significant parameters for the incorporation process. The pH of the incorporation solution (pHi), stirring time (t), and initial protein concentration in the crude extract (PCi) were selected as the three independent variables, and the efficiency of collagenase incorporation (E) and collagenolytic activity (CA) after 360 min as the responses. pHi and PCi showed positive statistically significant effects on E, while CA was positively influenced by pHi and t, but negatively by PCi. The fungi collagenase was released from the gel following a pseudo-Fickian behavior. Additionally, no <76 % of collagenase was efficiently incorporated into the gel retaining a high CA (32.5-69.8 U/mL). The obtained results for the commercial collagenase (E = 93.88 %, CA = 65.8 U/mL, and n = 0.10) demonstrated a pseudo-Fickian behavior similar to the fungi-collagenase. The results confirm the biotechnological potential of the gel as an efficient matrix for the incorporation of catalytic compounds; additionally, the incorporation of collagenases was achieved by retaining the proteases CA and releasing them in a controlled manner.


Subject(s)
Cassia , Galactose/analogs & derivatives , Mannans , Cassia/chemistry , Collagenases/chemistry , Fungi/metabolism , Seeds/chemistry
2.
J Biol Chem ; 298(8): 102109, 2022 08.
Article in English | MEDLINE | ID: mdl-35679897

ABSTRACT

Collagenase from the gram-negative bacterium Grimontia hollisae strain 1706B (Ghcol) degrades collagen more efficiently even than clostridial collagenase, the most widely used industrial collagenase. However, the structural determinants facilitating this efficiency are unclear. Here, we report the crystal structures of ligand-free and Gly-Pro-hydroxyproline (Hyp)-complexed Ghcol at 2.2 and 2.4 Å resolution, respectively. These structures revealed that the activator and peptidase domains in Ghcol form a saddle-shaped structure with one zinc ion and four calcium ions. In addition, the activator domain comprises two homologous subdomains, whereas zinc-bound water was observed in the ligand-free Ghcol. In the ligand-complexed Ghcol, we found two Gly-Pro-Hyp molecules, each bind at the active site and at two surfaces on the duplicate subdomains of the activator domain facing the active site, and the nucleophilic water is replaced by the carboxyl oxygen of Hyp at the P1 position. Furthermore, all Gly-Pro-Hyp molecules bound to Ghcol have almost the same conformation as Pro-Pro-Gly motif in model collagen (Pro-Pro-Gly)10, suggesting these three sites contribute to the unwinding of the collagen triple helix. A comparison of activities revealed that Ghcol exhibits broader substrate specificity than clostridial collagenase at the P2 and P2' positions, which may be attributed to the larger space available for substrate binding at the S2 and S2' sites in Ghcol. Analysis of variants of three active-site Tyr residues revealed that mutation of Tyr564 affected catalysis, whereas mutation of Tyr476 or Tyr555 affected substrate recognition. These results provide insights into the substrate specificity and mechanism of G. hollisae collagenase.


Subject(s)
Bacterial Proteins , Collagen , Collagenases , Vibrionaceae , Bacterial Proteins/chemistry , Collagen/chemistry , Collagenases/chemistry , Hydroxyproline/chemistry , Substrate Specificity , Vibrionaceae/enzymology , Water/chemistry , Zinc/chemistry
3.
Nat Commun ; 13(1): 566, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35091565

ABSTRACT

The collagenases of Vibrio species, many of which are pathogens, have been regarded as an important virulence factor. However, there is little information on the structure and collagenolytic mechanism of Vibrio collagenase. Here, we report the crystal structure of the collagenase module (CM) of Vibrio collagenase VhaC and the conformation of VhaC in solution. Structural and biochemical analyses and molecular dynamics studies reveal that triple-helical collagen is initially recognized by the activator domain, followed by subsequent cleavage by the peptidase domain along with the closing movement of CM. This is different from the peptidolytic mode or the proposed collagenolysis of Clostridium collagenase. We propose a model for the integrated collagenolytic mechanism of VhaC, integrating the functions of VhaC accessory domains and its collagen degradation pattern. This study provides insight into the mechanism of bacterial collagenolysis and helps in structure-based drug design targeting of the Vibrio collagenase.


Subject(s)
Bacterial Proteins/chemistry , Collagen/metabolism , Collagenases/chemistry , Protein Conformation , Vibrio/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites/genetics , Biocatalysis , Chromatography, Liquid , Collagenases/genetics , Collagenases/metabolism , Crystallography, X-Ray , Mass Spectrometry , Microscopy, Atomic Force , Molecular Dynamics Simulation , Peptides/genetics , Peptides/metabolism , Protein Binding , Vibrio/enzymology , Vibrio/genetics
4.
Methods Mol Biol ; 2319: 143-152, 2021.
Article in English | MEDLINE | ID: mdl-34331252

ABSTRACT

Heart disease is one of the leading causes of death in the United States. Isolation and culture adult cardiomyocytes are important for studying cardiomyocyte contractility, heart hypertrophy, and cardiac failure. In contrast to neonatal cardiomyocyte isolation, adult mice cardiomyocytes isolation is challenging due to firm connections among cardiomyocytes through intercalated discs. The availability of newly generated genetically modified mouse lines requires to establish protocols to isolation and culture adult mouse cardiomyocyte for in vitro studies. In this manuscript, we described a straightforward method of isolating adult mouse cardiomyocytes using Langendorff perfusion apparatus. Briefly, the hearts were harvested from adult mice and the heart was mounted to Lagendorff apparatus. After perfusion with calcium depletion and collagenase digestion, the left ventricles were minced and filtered. Lastly, the separated cardiomyocytes were treated with CaCl2. The isolated cardiac myocytes can be utilized in a broad range of experiments including screening for drugs.


Subject(s)
Cell Culture Techniques/methods , Cell Separation/instrumentation , Cell Separation/methods , Isolated Heart Preparation/instrumentation , Isolated Heart Preparation/methods , Myocytes, Cardiac/cytology , Animals , Calcium/pharmacology , Calcium Chloride/pharmacology , Cells, Cultured , Collagenases/chemistry , Collagenases/pharmacology , Mice , Myocytes, Cardiac/metabolism , Perfusion/methods
5.
Mol Vis ; 27: 125-141, 2021.
Article in English | MEDLINE | ID: mdl-33907368

ABSTRACT

Purpose: Collagen is a key player contributing to vitreoelasticity and vitreoretinal adhesions. Molecular reorganization causes spontaneous weakening of these adhesions with age, resulting in the separation of the posterior hyaloid membrane (PHM) from the retina in what is called complete posterior vitreous detachment (PVD). Incomplete separation of the posterior hyaloid or tight adherence or both can lead to retinal detachment, vitreomacular traction syndrome, or epiretinal membrane formation, which requires surgical intervention. Pharmacological vitrectomy has the potential of avoiding surgical vitrectomy; it is also useful as an adjunct during retinal surgery to induce PVD. Previously studied enzymatic reagents, such as collagenase derived from Clostridium histolyticum, are nonspecific and potentially toxic. We studied a novel collagenase from Vibrio mimicus (VMC) which remains active (VMA), even after deletion of 51 C-terminal amino acids. To limit the activity of VMA to the vitreous cavity, a fusion construct (inhibitor of hyaluronic acid-VMA [iHA-VMA]) was made in which a 12-mer peptide (iHA, which binds to HA) was fused to the N-terminus of VMA. The construct was evaluated in the context of PVD. Methods: VMA and iHA-VMA were expressed in Escherichia coli, purified, and characterized with gelatin zymography, collagen degradation assay, fluorescamine-based assay, and cell-based assays. Two sets of experiments were performed in New Zealand albino rabbits. Group A (n = 10) received iHA-VMA, while group B (n = 5) received the equivalent dose of VMA. In both groups, saline was injected as a control in the contralateral eyes. Animals were monitored with indirect ophthalmoscopy, optical coherence tomography (OCT), and B-scan ultrasonography. Retinal toxicity was assessed with hematoxylin and eosin (H&E) staining of retinal tissue. Results: The activity of iHA-VMA and VMA was comparable and 65-fold lower than that of C. histolyticum collagenase Type IV. In the iHA-VMA group, all the rabbits (n = 10) developed PVD, with complete PVD seen in six animals. No statistically significant histomorphological changes were seen. In the VMA group, four of the five rabbits developed complete PVD; however, retinal morphological changes were seen in two animals. Conclusions: iHA-VMA displays targeted action confined to the vitreous and shows potential for safe pharmacologic vitreolysis.


Subject(s)
Collagenases/therapeutic use , Hyaluronic Acid/therapeutic use , Vibrio mimicus/enzymology , Vitrectomy/methods , Vitreous Body/drug effects , Vitreous Detachment/chemically induced , Animals , Cell Survival , Collagenases/chemistry , Collagenases/genetics , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Goats , Hyaluronic Acid/chemistry , Hyaluronic Acid/genetics , Intravitreal Injections , Microscopy, Electron, Scanning , Ophthalmoscopy , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use , Retina/drug effects , Retina/physiology , Vitreous Body/ultrastructure , Vitreous Detachment/diagnostic imaging
6.
Carbohydr Polym ; 263: 117964, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33858569

ABSTRACT

Components of the extracellular matrix (ECM) are overexpressed in fibrotic liver. Collagen is the main component of the liver fibrosis stroma. Here we demonstrate that chondroitin sulfate coated multilayered 50-nm nanoparticles encapsulating collagenase and silibinin (COL + SLB-MLPs) break down the dense collagen stroma, while silibinin inhibits activated hepatic stellate cells. The nanoparticles were taken up to a much greater extent by hepatic stellate cells than by normal hepatocytes, and they down-regulated production of type I collagen. In addition, chondroitin sulfate protected the collagenase from premature deactivation. COL + SLB-MLPs were delivered to the cirrhotic liver, and the collagenase and silibinin synergistically inhibited fibrosis in mice. Immunofluorescence staining of liver tissues revealed that CD44, mediated by chondroitin sulfate, delivered the nanoparticles to hepatic stellate cells. This strategy holds promise for degrading extracellular stroma and thereby facilitating drug penetration into fibrotic liver and related diseases such as liver cirrhosis and liver cancer.


Subject(s)
Chondroitin Sulfates/chemistry , Chondroitin Sulfates/metabolism , Collagenases/chemistry , Collagenases/pharmacology , Liver Cirrhosis/drug therapy , Nanoparticles/chemistry , Silybin/chemistry , Silybin/pharmacology , Animals , Capsules/chemistry , Cell Line , Chondroitin Sulfates/administration & dosage , Collagenases/administration & dosage , Disease Models, Animal , Hepatic Stellate Cells/drug effects , Humans , Hyaluronan Receptors/metabolism , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/pathology , Mice , Nanoparticles/therapeutic use , Silybin/administration & dosage
7.
Mater Sci Eng C Mater Biol Appl ; 112: 110902, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32409058

ABSTRACT

When damaged or fractured collagen-rich hard tissues are repaired by resin material, the collagen matrix may be used as a scaffold, after removal of the natural minerals, for resin monomers to penetrate and polymerize in-situ. Formation of a collagen-polymer hybrid biocomposite via mechanical hybridization provides a stable and strong link between endogenous tissue and the prosthesis for successful clinical integration. However, the heterogeneity between hydrophobic resin polymers and hydrophilic collagen presents a challenge to the quality of hybrid biocomposite. The objective of the present study was to evaluate the potential benefits of a collagen-reactive monomer (CRM, an isocyanate-terminated urethane-based methacrylate) with covalent affinity to collagen as "chemical link" to enhance in-situ resin hybridization within a collagen scaffold. Here, the CRM ligand with active isocyanate group may be chemically grafted onto the collagen receptor via covalent and hydrogen bonds. Dentin-derived collagen chemical modified by CRM shows improved mechanical property, thermostability and enzymatic stability. Moreover, CRM inhibited both exogenous and endogenous collagenase activities. The modification of collagen by chemical grafting of resin monomers improved its mechanical and physicochemical properties and demonstrated the potential of CRM for use in promoting chemical adhesion and creating a much stronger and durable bonding interface. Formation of a chemical bond between polymer and collagen scaffold in-situ improves the mechanical performance of collagen and may create a much stronger and durable collagen-polymer hybrid material. Addition of CRM into adhesives might effectively prolong the longevity of clinical resin-bonded restorations.


Subject(s)
Collagen/chemistry , Composite Resins/chemistry , Isocyanates/chemistry , Methacrylates/chemistry , Urethane/chemistry , Binding Sites , Collagen/metabolism , Collagenases/chemistry , Collagenases/metabolism , Composite Resins/metabolism , Dentin/chemistry , Dentin/metabolism , Materials Testing , Molecular Docking Simulation , Temperature , Tensile Strength
8.
J Biochem ; 168(4): 385-392, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32386303

ABSTRACT

Collagenase from the Grimontia hollisae strain 1706B (Ghcol) is a zinc metalloproteinase with the zinc-binding motif H492EXXH496. It exhibits higher collagen-degrading activity than the collagenase from Clostridium histolyticum, which is widely used in industry. We previously examined the pH and temperature dependencies of Ghcol activity; Glu493 was thought to contribute acidic pKa (pKe1), while no residue was assigned to contribute alkaline pKa (pKe2). In this study, we introduced nine single mutations at the His or Tyr residues in and near the active site. Our results showed that H412A, H485A, Y497A, H578A and H737A retained the activities to hydrolyze collagen and gelatin, while H426A, H492A, H496A and Y568A lacked them. Purification of active variants H412A, H485A, H578A and H737A, along with inactive variants H492A and H496A, were successful. H412A preferred (7-methoxycoumarin-4-yl)acetyl-L-Lys-L-Pro-L-Leu-Gly-L-Leu-[N3-(2,4-dinitrophenyl)-L-2,3-diaminopropionyl]-L-Ala-L-Arg-NH2 to collagen, while H485A preferred collagen to the peptide, suggesting that His412 and His485 are important for substrate specificity. Purification of the active variant Y497A and inactive variants H426A and Y568A were unsuccessful, suggesting that these three residues were important for stability. Based on the reported crystal structure of clostridial collagenase, Tyr568 of Ghcol is suggested to be involved in catalysis and may be the ionizable residue for pKe2.


Subject(s)
Collagenases/metabolism , Histidine/metabolism , Tyrosine/metabolism , Vibrionaceae/enzymology , Amino Acid Sequence , Catalysis , Catalytic Domain , Collagenases/chemistry , Collagenases/genetics , Collagenases/isolation & purification , Histidine/chemistry , Histidine/genetics , Mutagenesis, Site-Directed/methods , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Homology , Structure-Activity Relationship , Substrate Specificity , Tyrosine/chemistry , Tyrosine/genetics , Vibrionaceae/genetics
9.
Sci Rep ; 10(1): 6841, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32321979

ABSTRACT

The mammalian ovarian follicle is comprised of the germ cell or oocyte surrounded by the somatic cells, the granulosa and theca cells. The ovarian stroma, including the collagen-rich matrix that supports the three-dimensional disk-like follicular structure, impacts the integrity of the ovarian follicle and is essential for follicular development. Maintaining follicular integrity during cryopreservation has remained a limiting factor in preserving ovarian tissues for transplantation because a significant proportion of developed follicles in the frozen-thawed ovaries undergo atresia after transplantation. In this study, we show for the first time that during vitrification of the mouse ovary, the attachment of the oocyte to the granulosa cells was impaired by the loss of the cadherin adhesion molecules. Importantly, exposure to a high osmotic solution greatly decreased the ratio of oocyte diameter to the diameter of its follicle but did not alter the collagen-rich matrix surrounding the follicles. By treating ovaries briefly with collagenase before exposure to the hyper-osmotic solution the ratio of oocyte diameter to follicle diameter was maintained, and cadherin adhesion junctions were preserved. When frozen-thawed ovaries were transplanted to the bursa of recipient hosts, pretreatment with collagenase significantly increased serum levels of AMH, the number of intact follicles and the total number of viable offspring compared to frozen-thawed ovaries without collagenase pretreatment, even 6 months after transplantation. Thus, the collagenase pretreatment could provide a beneficial approach for maintaining the functions and viability of cryopreserved ovaries in other species and clinically relevant situations.


Subject(s)
Cryopreservation , Granulosa Cells/metabolism , Oocytes/metabolism , Animals , Cell Adhesion , Collagenases/chemistry , Female , Granulosa Cells/cytology , Granulosa Cells/transplantation , Mice , Oocytes/cytology , Vitrification
10.
Int J Biol Macromol ; 150: 1314-1322, 2020 May 01.
Article in English | MEDLINE | ID: mdl-31747567

ABSTRACT

In this study, silk fibroin (SF)/sodium alginate (SA) porous materials (PMs) with different blend ratios were generated using 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) as crosslinking agent by a simple freeze-dried method. Degradation experiment of SF/SA PMs have been systematically investigated up to 18 days in Collagenase IA solution at 37 °C, Phosphate buffer saline (PBS) solution without enzyme was used as a control. The results showed SF/SA 50/50 PMs exhibited a lowest rate of weight loss, about 68% of the weight retained within 18 d in Collagenase IA solution. SEM images indicated Collagenase IA can degrade fibroin leading to collapse of the pure SF PMs, while SF/SA 50/50 PMs still possessed integrity of pore structure during enzyme degradation with increasing exposure time. The crystalline structure of the SF in the SF/SA PMs changed to silk II after degradation for 18 d. Furthermore, the results of the in vivo degradation by subcutaneous implantation in rats showed that all PMs can be degraded at different levels, and exhibited good subcutaneous histocompatibility to the host animals. The degradability was strongly correlated to the blend ratios in a series of SF/SA composite PMs, and insights gained in this study can serve as a guide to match desired degradation behavior with specific applications for the SF/SA composite PMs.


Subject(s)
Alginates , Fibroins , Materials Testing , Alginates/chemistry , Alginates/pharmacokinetics , Alginates/pharmacology , Animals , Collagenases/chemistry , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Fibroins/chemistry , Fibroins/pharmacokinetics , Fibroins/pharmacology , Male , Porosity , Rats , Rats, Sprague-Dawley
11.
Bioelectromagnetics ; 41(2): 113-120, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31872912

ABSTRACT

Ultrasound has been applied for varied purposes as it provides additional mechanical energy to a system, and is still profitable and straightforward, which are advantages for industrial applications. In this work, ultrasonic treatments were applied to purified collagenase fractions from a fermented extract by Aspergillus terreus UCP 1276 aiming to evaluate the potential effect on collagen hydrolysis. The physical agent was evaluated as an inductor of collagen degradation and consequently as a producer of peptides with anticoagulant activity. The sodium dodecyl sulphate-polyacrylamide gel electrophoresis analyses were also carried out to compare the hydrolysis techniques. The ultrasound (40 kHz, 47.4 W/L) processing was conducted under the same conditions of pH and temperature at different times. The ultrasound-assisted reaction was accelerated in relation to conventional processing. Collagenolytic activity was enhanced and tested in the presence of phenylmethanesulfonyl fluoride inhibitor. Underexposure, the activity was enhanced, reaching more than 72.0% of improvement in relation to the non-exposed enzyme. A period of 30 min of incubation under ultrasound exposure was enough to efficiently produce peptides with biological activity, including anticoagulation and effect on prothrombin time at about 60%. The results indicate that low-frequency ultrasound is an enzymatic inducer with likely commercial applicability accelerating the enzymatic reaction. Bioelectromagnetics. 2020;41:113-120. © 2019 Bioelectromagnetics Society.


Subject(s)
Anticoagulants/pharmacology , Aspergillus/enzymology , Collagen/chemistry , Collagenases/metabolism , Peptides/chemistry , Anticoagulants/chemistry , Catalysis , Collagen/metabolism , Collagenases/chemistry , Collagenases/isolation & purification , Electrophoresis, Polyacrylamide Gel , Fermentation , Humans , Hydrolysis , Peptides/pharmacology , Phenylmethylsulfonyl Fluoride/chemistry , Phenylmethylsulfonyl Fluoride/pharmacology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protein Hydrolysates/chemistry , Ultrasonics/methods
12.
J Mech Behav Biomed Mater ; 100: 103406, 2019 12.
Article in English | MEDLINE | ID: mdl-31473438

ABSTRACT

The temporomandibular joint disc is a structure, characterized as heterogeneous fibrocartilage, and is composed of macromolecular biopolymers. Despite a large body of characterization studies, the contribution of matrix biopolymers on the dynamic viscoelastic behavior of the disc is poorly understood. Given the high permeability and low concentration of glycosaminoglycans in the disc, it has been suggested that poro-elastic behavior can be neglected and that the intrinsic viscoelastic nature of solid matrix plays a dominant role in governing its time-dependent behavior. This study attempts to quantify the contribution of collagen and elastin fibers to the viscoelastic properties of the disc. Using collagenase and elastase, we perturbed the collagen and elastin fibrillar network in porcine temporomandibular joint discs and investigated the changes of dynamic viscoelastic properties in five different regions of the disc. Following both treatments, the storage and loss moduli of these regions were reduced dramatically up to the point that the tissue was no longer mechanically heterogeneous. However, the proportion of changes in storage and loss moduli were different for each treatment, reflected in the decrease and increase of the loss tangent for collagenase and elastase treated discs, respectively. The reduction of storage and loss moduli of the disc correlated with a decrease of biopolymer length. The present study indicates that the compositional and structural changes of collagen and elastin fibers alter the viscoelastic properties of the disc consistent with polymer dynamics.


Subject(s)
Biopolymers/chemistry , Collagen/chemistry , Elastin/chemistry , Polymers/chemistry , Temporomandibular Joint Disc/physiology , Animals , Biomechanical Phenomena , Collagenases/chemistry , Elasticity , Glycosaminoglycans/chemistry , Microscopy, Electron, Transmission , Permeability , Stress, Mechanical , Swine , Temperature , Viscosity
13.
ACS Nano ; 13(10): 11008-11021, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31503443

ABSTRACT

Overexpressed extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDAC) limits drug penetration into the tumor and is associated with poor prognosis. Here, we demonstrate that a pretreatment based on a proteolytic-enzyme nanoparticle system disassembles the dense PDAC collagen stroma and increases drug penetration into the pancreatic tumor. More specifically, the collagozome, a 100 nm liposome encapsulating collagenase, was rationally designed to protect the collagenase from premature deactivation and prolonged its release rate at the target site. Collagen is the main component of the PDAC stroma, reaching 12.8 ± 2.3% vol in diseased mice pancreases, compared to 1.4 ± 0.4% in healthy mice. Upon intravenous injection of the collagozome, ∼1% of the injected dose reached the pancreas over 8 h, reducing the level of fibrotic tissue to 5.6 ± 0.8%. The collagozome pretreatment allowed increased drug penetration into the pancreas and improved PDAC treatment. PDAC tumors, pretreated with the collagozome followed by paclitaxel micelles, were 87% smaller than tumors pretreated with empty liposomes followed by paclitaxel micelles. Interestingly, degrading the ECM did not increase the number of circulating tumor cells or metastasis. This strategy holds promise for degrading the extracellular stroma in other diseases as well, such as liver fibrosis, enhancing tissue permeability before drug administration.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Collagenases/pharmacology , Nanoparticles/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Collagen/chemistry , Collagen/genetics , Collagenases/chemistry , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Fibrosis/drug therapy , Fibrosis/pathology , Fibrosis/prevention & control , Humans , Liposomes/chemistry , Liposomes/pharmacology , Mice , Nanoparticles/therapeutic use , Paclitaxel/chemistry , Paclitaxel/pharmacology , Pancreas/drug effects , Pancreas/pathology , Tumor Microenvironment/drug effects
14.
Food Chem ; 301: 125302, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31387034

ABSTRACT

The autolysis of sea cucumber is caused by depolymerisation of collagen fibres and unfolding of fibrils. In order to highlight the role of collagenase in sea cucumber autolysis, collagen fibres from sea cucumber were hydrolysed with collagenase type I. Electron microscopy (EM) results indicated the collagenase caused partial depolymerisation of collagen fibres into fibrils due to the fracture of proteoglycan interfibrillar bridges, as well as uncoiling of collagen fibrils. Chemical analysis and SDS-PAGE both indicated collagenase induced a time-dependent release of glycosaminoglycans (GAGs) and soluble proteins, which further demonstrated the degradation of proteoglycan interfibrillar bridges. Collagenase also degraded collagens by releasing soluble hydroxyproline (Hpy), with the dissolution rate of Hyp reaching 11.11% after 72 h. Fourier transform infrared analysis showed that collagenase caused the reduction of intermolecular interactions and structural order of collagen. Hence, collagenase participated in the autolysis of sea cucumber by deteriorating both macromolecular and monomeric collagens.


Subject(s)
Collagen/chemistry , Collagenases/chemistry , Stichopus/chemistry , Animals , Autolysis , Collagen/metabolism , Collagenases/metabolism , Electrophoresis, Polyacrylamide Gel , Glycosaminoglycans/metabolism , Hydrolysis , Hydroxyproline/metabolism , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Proteoglycans/chemistry , Proteoglycans/metabolism , Spectroscopy, Fourier Transform Infrared , Stichopus/anatomy & histology
15.
J Ethnopharmacol ; 245: 112168, 2019 Dec 05.
Article in English | MEDLINE | ID: mdl-31430525

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Scorzonera latifolia (Fisch. & Mey.) DC. (Asteraceae) grows naturally in Eastern Anatolia, northeastern Iran, and Caucasus. Latex of S. latifolia roots is used in Turkish folk medicine for its analgesic effects, externally to cure infertility in women, and internally as an antihelmintic. The milk obtained from the stem of the Scorzonera species is used for wound healing activity. Antinociceptive, anti-inflammatory, wound-healing, antioxidant, and antimicrobial activities have previously been reported for S. latifolia. AIM OF THE STUDY: A methanol extract of the aerial parts of Scorzonera latifolia that had been shown to possess wound-healing activity, was used to elucidate the possible mechanism of the wound-healing activity and to identify the compound(s) responsible for the effect by means of bioassay-guided fractionation. MATERIALS AND METHODS: The wound-healing activity potential of methanol extract of S. latifolia was detected by evaluating the inhibitory activity on the collagenase, hyaluronidase and elastase, which play important roles in the wound-healing process. Succesive fractionation of the methanol extract using petroleum ether, chloroform, ethyl acetate, respectively, and the residual wateryielded four respective fractions. The ethyl acetate part, which was determined as the most active fraction, was selected for further separation using chromatographic techniques. RESULTS: Ethylacetate fraction exhibited significant inhibitory activities on collagenase and elastase. Chromatographic separation of the ethylacetate extract yielded an active subfraction, from which was used to isolate quercetin-3-O-ß-apiofuranosyl-(1'''→2'')-ß-D-glucopyranoside (1), quercetin-3-O-α-rhamnopyranosyl-(1→6)-ß-D-galactopyranoside (2), isoorientin (3), and 7-methylisoorientin (4). Of the compounds tested, 7-methylisoorientin (4) exerted inhibitory activity on collagenase and elastase, while quercetin-3-O-ß-apiofuranosyl-(1'''→2'')-ß-glucopyranoside (1) inhibited collagenase only. None of the fractions, or isolated compounds showed any inhibitory effect on hyaluronidase. It must be mentioned, that in vitro tests showed that compounds 1-4 inhibit the collagenase and elastase and could help wound-healing process. However, the inhibititory effect of the methanol extract appears to be greater than that of both of the ethylacetate fraction, subfraction G and the isolated compounds, which suggest that a synergistic interaction of several compounds could be responsible for the wound-healing activity of the aerial parts of S. latifolia.


Subject(s)
Analgesics/chemistry , Anti-Inflammatory Agents/chemistry , Collagenases/chemistry , Hyaluronoglucosaminidase/antagonists & inhibitors , Matrix Metalloproteinase Inhibitors/chemistry , Pancreatic Elastase/antagonists & inhibitors , Plant Extracts/chemistry , Scorzonera , Medicine, Traditional , Plant Components, Aerial , Turkey , Wound Healing
16.
Colloids Surf B Biointerfaces ; 182: 110350, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31326622

ABSTRACT

Centaurea pumilio was the subject of phytochemical and biological studies, and its extract was used in the green synthesis of silver nanoparticles (AgNPs). Liquid chromatography/electrospray ionization mass spectrometry allowed the tentative identification of twenty-nine phytoconstituents of C. pumilio methanolic extract (CME), while column chromatography led to the identification of eight phenolic compounds. The neutral red uptake method showed the safety of CME and AgNPs on skin cells (HaCaT cell lines), while their high antioxidant potentials were demonstrated based on their oxygen radical absorbance capacity, and these results were confirmed in vivo. Additionally, CME and AgNPs had promising abilities to retard the ageing process and combat dark spots by potently inhibiting collagenase, elastase and tyrosinase, in addition to antimicrobial activity against skin infection-causing strains, especially Staphylococcus aureus, which was further confirmed by the significant phagocytic activity of neutrophils via engulfment. This study presents C. pumilio as a candidate for healthy skin.


Subject(s)
Anti-Infective Agents/chemistry , Antioxidants/chemistry , Centaurea/chemistry , Enzyme Inhibitors/chemistry , Flavonoids/chemistry , Glycosides/chemistry , Metal Nanoparticles/chemistry , Animals , Anti-Infective Agents/isolation & purification , Anti-Infective Agents/pharmacology , Antioxidants/isolation & purification , Antioxidants/pharmacology , Candida albicans/growth & development , Cell Line , Collagenases/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Epithelial Cells , Flavonoids/isolation & purification , Flavonoids/pharmacology , Glycosides/isolation & purification , Glycosides/pharmacology , Humans , Metal Nanoparticles/administration & dosage , Monophenol Monooxygenase/antagonists & inhibitors , Monophenol Monooxygenase/chemistry , Neutrophils/cytology , Neutrophils/drug effects , Pancreatic Elastase/antagonists & inhibitors , Pancreatic Elastase/chemistry , Phagocytosis/drug effects , Plant Components, Aerial/chemistry , Plant Extracts/chemistry , Primary Cell Culture , Rats , Rats, Wistar , Silver/chemistry , Skin/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
17.
Int J Mol Sci ; 20(12)2019 Jun 24.
Article in English | MEDLINE | ID: mdl-31238564

ABSTRACT

The physiological spaces (lateral ventricles, intrathecal space) or pathological cavities (stroke lesion, syringomyelia) may serve as an attractive gateway for minimally invasive deployment of stem cells. Embedding stem cells in injectable scaffolds is essential when transplanting into the body cavities as they secure favorable microenvironment and keep cells localized, thereby preventing sedimentation. However, the limited migration of transplanted cells from scaffold to the host tissue is still a major obstacle, which prevents this approach from wider implementation for the rapidly growing field of regenerative medicine. Hyaluronan, a naturally occurring polymer, is frequently used as a basis of injectable scaffolds. We hypothesized that supplementation of hyaluronan with activated proteolytic enzymes could be a viable approach for dissolving the connective tissue barrier on the interface between the scaffold and the host, such as pia mater or scar tissue, thus demarcating lesion cavity. In a proof-of-concept study, we have found that collagenase and trypsin immobilized in hyaluronan-based hydrogel retain 60% and 28% of their proteolytic activity compared to their non-immobilized forms, respectively. We have also shown that immobilized enzymes do not have a negative effect on the viability of stem cells (glial progenitors and mesenchymal stem cells) in vitro. In conclusion, proteolytic rafts composed of hyaluronan-based hydrogels and immobilized enzymes may be an attractive strategy to facilitate migration of stem cells from injectable scaffolds into the parenchyma of surrounding tissue.


Subject(s)
Hydrogels , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/physiology , Tissue Scaffolds , Animals , Cell Movement , Cell Survival , Cells, Immobilized , Collagenases/chemistry , Humans , Hyaluronic Acid , Hydrogels/chemistry , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice , Proteolysis , Stem Cell Transplantation/methods , Tissue Scaffolds/chemistry , Trypsin/chemistry
18.
Int Wound J ; 16(4): 1013-1023, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31148413

ABSTRACT

Bacterial collagenase from the aerobic non-pathogenic Vibrio alginolyticus chemovar iophagus is an extracellular metalloproteinase. This collagenase preparation is obtained through a fermentation process and is purified chromatographically, resulting in a highly purified 82-kDa single-band protein that does not contain non-specific proteases or other microbial impurities. V. alginolyticus collagenase was added to a hyaluronan (HA)-based device to develop a novel debriding agent to improve the treatment of ulcers, necrotic burns, and decubitus in the initial phase of wound bed preparation. In this study, an in vitro biochemical characterisation of V. alginolyticus collagenase versus a commercial preparation from a Clostridium histolyticum strain on various dermal extracellular matrix (ECM) substrates was performed. V. alginolyticus collagenase demonstrated its ability to carry out the enzymatic cleavage of the substrate, allowing a selective removal of necrotic tissues while sparing healthy tissue, as reported in clinical studies and through routine clinical experience. in vitro tests under physiological conditions (pH, presence of Ca++, etc.) have demonstrated that V. alginolyticus collagenase exhibits very poor/limited non-specific proteolytic activity, whereas the collagenase preparation from C. histolyticum is highly active both on collagen and on non-collagenic substrates. This finding implies that while the V. alginolyticus enzyme is fully active on the collagen filaments that anchor the necrotic tissue to the wound bed, it does not degrade other minor, but structurally important, components of the dermal ECM. This feature could explain why collagenase preparation from V. alginolyticus has been reported to be much gentler on perilesional, healthy skin.


Subject(s)
Collagenases/chemistry , Collagenases/therapeutic use , Microbial Collagenase/chemistry , Microbial Collagenase/therapeutic use , Substrate Specificity/drug effects , Wound Healing/drug effects , Wounds and Injuries/drug therapy , Clostridium histolyticum/chemistry , Humans , Vibrio alginolyticus/chemistry
19.
Carbohydr Polym ; 216: 45-53, 2019 Jul 15.
Article in English | MEDLINE | ID: mdl-31047081

ABSTRACT

Herein the construction of a strong gelatin hydrogel is presented by using pullulan dialdehyde (PDA) as a macromolecular crosslinker. The resultant PDA crosslinked gelatin hydrogels (G-PDA) exhibit extremely high mechanical strength, manifested in the achieved optimal compressive stress of 5.80 MPa at 80% strain, which is up to 152 times higher than pure gelatin hydrogel. The G-PDA were characterized by Fourier transform infrared (FTIR) spectroscopy and scanning electron microscopy (SEM). The extent of crosslinking was determined by ninhydrin assay. The results suggested that the synergistic effect of dual-crosslinking, which is composed of short- and long-range covalent crosslinking and thermoreversible physical crosslinking, may played a key role in enhancing the load-bearing capacity of ensuing hydrogels. The swelling and enzymatic degradation of G-PDA are gradually limited with increasing PDA concentration. The result from MTT assay demonstrated that G-PDA is non-cytotoxic against MC3T3 cells, regardless of the concentrations of PDA.


Subject(s)
Aldehydes/chemistry , Biocompatible Materials/chemistry , Cross-Linking Reagents/chemistry , Gelatin/chemistry , Glucans/chemistry , Hydrogels/chemistry , Aldehydes/chemical synthesis , Aldehydes/toxicity , Animals , Biocompatible Materials/chemical synthesis , Biocompatible Materials/toxicity , Cell Line , Collagenases/chemistry , Compressive Strength , Cross-Linking Reagents/chemical synthesis , Cross-Linking Reagents/toxicity , Elastic Modulus , Gelatin/chemical synthesis , Gelatin/toxicity , Glucans/chemical synthesis , Glucans/toxicity , Hydrogels/chemical synthesis , Hydrogels/toxicity , Hydrolysis , Mice , Oxidation-Reduction , Periodic Acid/chemistry , Porosity
20.
Macromol Rapid Commun ; 40(15): e1900127, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31136037

ABSTRACT

Injectable hydrogels are considered important to realize safe and effective minimally invasive therapy. Although animal-derived natural polymers are well studied, they typically lack injectability and fail to eliminate the potential risks of immunogenic reactions or unknown pathogen contamination. Despite extensive research activities to explore ideal injectable hydrogels, such state-of-the-art technology remains inaccessible to non-specialists. In this article, the design of a new injectable hydrogel platform that can be extemporaneously prepared from commercially available animal-component-free materials is described. The hydrogels can be prepared simply by mixing mutually reactive aqueous solutions without necessitating specialized knowledge or equipment. Their solidification time can be adjusted by choosing proper buffer conditions from immediate to an extended period of time, that is, few or several tens of minutes depending on the concentration of polymeric components, which not only provides injectability, but enables 3D encapsulation of cells. Mesenchymal stromal/stem cells can be encapsulated and cultured in the hydrogels at least for 2 weeks by traditional cell culture techniques, and retrieved by collagenase digestion with cell viability of approximately 80%. This hydrogel platform accelerates future cell-related research activities.


Subject(s)
Cell Culture Techniques , Collagen Type I/metabolism , Collagenases/metabolism , Hydrogels/metabolism , Mesenchymal Stem Cells/cytology , Animals , Cell Survival , Collagen Type I/chemistry , Collagenases/chemistry , Humans , Hydrogels/chemistry , Mesenchymal Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...