Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.784
Filter
1.
Sci Rep ; 14(1): 9469, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658583

ABSTRACT

Bovine mastitis caused by S. aureus has a major economic impact on the dairy sector. With the crucial need for new therapies, anti-virulence strategies have gained attention as alternatives to antibiotics. Here we aimed to identify novel compounds that inhibit the production/activity of hemolysins, a virulence factor of S. aureus associated with mastitis severity. We screened Bacillus strains obtained from diverse sources for compounds showing anti-hemolytic activity. Our results demonstrate that lipopeptides produced by Bacillus spp. completely prevented the hemolytic activity of S. aureus at certain concentrations. Following purification, both iturins, fengycins, and surfactins were able to reduce hemolysis caused by S. aureus, with iturins showing the highest anti-hemolytic activity (up to 76% reduction). The lipopeptides showed an effect at the post-translational level. Molecular docking simulations demonstrated that these compounds can bind to hemolysin, possibly interfering with enzyme action. Lastly, molecular dynamics analysis indicated general stability of important residues for hemolysin activity as well as the presence of hydrogen bonds between iturins and these residues, with longevous interactions. Our data reveals, for the first time, an anti-hemolytic activity of lipopeptides and highlights the potential application of iturins as an anti-virulence therapy to control bovine mastitis caused by S. aureus.


Subject(s)
Bacillus , Hemolysin Proteins , Hemolysis , Lipopeptides , Molecular Docking Simulation , Staphylococcus aureus , Bacillus/metabolism , Bacillus/chemistry , Staphylococcus aureus/drug effects , Hemolysis/drug effects , Animals , Cattle , Lipopeptides/pharmacology , Lipopeptides/chemistry , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Mastitis, Bovine/microbiology , Mastitis, Bovine/drug therapy , Female , Peptides, Cyclic/pharmacology , Peptides, Cyclic/chemistry , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Molecular Dynamics Simulation
2.
Mar Drugs ; 22(4)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38667805

ABSTRACT

Three Laminaria japonica polysaccharides (LJPs) extracted via water extraction (LJP-W), acid extraction (LJP-A), and enzymatic extraction (LJP-E) were used as raw materials to be cross-linked with chitosan and polyvinyl alcohol to prepare hydrogels. Compared with conventional hydrogel systems, all three types of LJP-based polysaccharide hydrogels exhibited better swelling properties (14 times their original weight) and the absorption ability of simulated body fluid (first 2 h: 6-10%). They also demonstrated better rigidity and mechanical strength. Young's modulus of LJP-E was 4 times that of the blank. In terms of hemostatic properties, all three polysaccharide hydrogels did not show significant cytotoxic and hemolytic properties. The enzyme- and acid-extracted hydrogels (LJP-Gel-A and LJP-Gel-E) demonstrated better whole-blood coagulant ability compared with the water-extracted hydrogel (LJP-Gel-W), as evidenced by the whole blood coagulation index being half that of LJP-Gel-W. Additionally, the lactate dehydrogenase viabilities of LJP-Gel-A and LJP-Gel-E were significantly higher, at about four and three times those of water extraction, respectively. The above results suggested that LJP-Gel-A and LJP-Gel-E exhibited better blood coagulation capabilities than LJP-Gel-W, due to their enhanced platelet enrichment and adhesion properties. Consequently, these hydrogels are more conducive to promoting coagulation and have good potential for wound hemostasis.


Subject(s)
Blood Coagulation , 60578 , Hemostatics , Hydrogels , Laminaria , Polysaccharides , Hydrogels/chemistry , Hydrogels/pharmacology , Laminaria/chemistry , Polysaccharides/chemistry , Polysaccharides/pharmacology , Polysaccharides/isolation & purification , Blood Coagulation/drug effects , Hemostatics/pharmacology , Hemostatics/chemistry , Hemostatics/isolation & purification , Humans , Animals , Chitosan/chemistry , Chitosan/pharmacology , Polyvinyl Alcohol/chemistry , Hemostasis/drug effects , Hemolysis/drug effects
4.
J Med Chem ; 67(8): 6365-6383, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38436574

ABSTRACT

Antimicrobial peptides (AMPs) have emerged as promising agents to combat the antibiotic resistance crisis due to their rapid bactericidal activity and low propensity for drug resistance. However, AMPs face challenges in terms of balancing enhanced antimicrobial efficacy with increased toxicity during modification processes. In this study, de novo d-type ß-hairpin AMPs are designed. The conformational transformation of self-assembling peptide W-4 in the environment of the bacterial membrane and the erythrocyte membrane affected its antibacterial activity and hemolytic activity and finally showed a high antibacterial effect and low toxicity. Furthermore, W-4 displays remarkable stability, minimal occurrence of drug resistance, and synergistic effects when combined with antibiotics. The in vivo studies confirm its high safety and potent wound-healing properties at the sites infected by bacteria. This study substantiates that nanostructured AMPs possess enhanced biocompatibility. These advances reveal the superiority of self-assembled AMPs and contribute to the development of nanoantibacterial materials.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Peptides , Hemolysis , Microbial Sensitivity Tests , Nanofibers , Tryptophan , Nanofibers/chemistry , Tryptophan/chemistry , Tryptophan/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemical synthesis , Hemolysis/drug effects , Animals , Humans , Mice
5.
Blood Adv ; 7(16): 4258-4268, 2023 08 22.
Article in English | MEDLINE | ID: mdl-36897252

ABSTRACT

Dysregulated activation of the complement system is implicated in the onset or progression of several diseases. Most clinical-stage complement inhibitors target the inactive complement proteins present at high concentrations in plasma, which increases target-mediated drug disposition and necessitates high drug levels to sustain therapeutic inhibition. Furthermore, many efforts are aimed at inhibiting only terminal pathway activity, which leaves opsonin-mediated effector functions intact. We describe the discovery of SAR443809, a specific inhibitor of the alternative pathway C3/C5 convertase (C3bBb). SAR443809 selectively binds to the activated form of factor B (factor Bb) and inhibits alternative pathway activity by blocking the cleavage of C3, leaving the initiation of classical and lectin complement pathways unaffected. Ex vivo experiments with patient-derived paroxysmal nocturnal hemoglobinuria erythrocytes show that, although terminal pathway inhibition via C5 blockade can effectively inhibit hemolysis, proximal complement inhibition with SAR443809 inhibits both hemolysis and C3b deposition, abrogating the propensity for extravascular hemolysis. Finally, intravenous and subcutaneous administration of the antibody in nonhuman primates demonstrated sustained inhibition of complement activity for several weeks after injection. Overall, SAR443809 shows strong potential for treatment of alternative pathway-mediated disorders.


Subject(s)
Complement Factor B , Complement Pathway, Alternative , Animals , Complement Factor B/antagonists & inhibitors , Erythrocytes/drug effects , Hemolysis/drug effects , Complement C3-C5 Convertases/antagonists & inhibitors , Complement Pathway, Alternative/drug effects , Immune System Diseases/drug therapy , Immune System Diseases/enzymology , Humans , Macaca fascicularis , Antibodies/administration & dosage , Proteolysis/drug effects
6.
Carbohydr Polym ; 309: 120702, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-36906367

ABSTRACT

The acidity of high tannic acid (TA) content solution can destroy the structure of protein, such as gelatin (G). This causes a big challenge to introduce abundant TA into the G-based hydrogels. Here, the G-based hydrogel system with abundant TA as hydrogen bonds provider was constructed by a "protective film" strategy. The protective film around the composite hydrogel was first formed by the chelation of sodium alginate (SA) and Ca2+. Subsequently, abundant TA and Ca2+ were successively introduced into the hydrogel system by immersing method. This strategy effectively protected the structure of the designed hydrogel. After treatment with 0.3 w/v TA and 0.06 w/v Ca2+ solutions, the tensile modulus, elongation at break and toughness of G/SA hydrogel increased about 4-, 2-, and 6-fold, respectively. Besides, G/SA-TA/Ca2+ hydrogels exhibited good water retention, anti-freezing, antioxidant, antibacterial properties and low hemolysis ratio. Cell experiments showed that G/SA-TA/Ca2+ hydrogels possessed good biocompatibility and could promote cell migration. Therefore, G/SA-TA/Ca2+ hydrogels are expected to be used in the field of biomedical engineering. The strategy proposed in this work also provides a new idea for improving the properties of other protein-based hydrogels.


Subject(s)
Alginates , Anti-Bacterial Agents , Antioxidants , Biocompatible Materials , Gelatin , Hydrogels , Gelatin/chemistry , Alginates/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Polyphenols , Tensile Strength , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Movement/drug effects , Calcium/chemistry , Cations, Divalent/chemistry , Solutions , Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Animals , Rabbits , Hemolysis/drug effects , L Cells , Mice
7.
Wien Med Wochenschr ; 173(5-6): 152-157, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36178637

ABSTRACT

BI2536 is potent inhibitor of polo-like kinases PLK1, 2, and 3. The inhibition of PLKs in nucleated cells induces apoptosis by perturbing the cell cycle with consequent engagement of mitotic catastrophe. BI2536 is being tested as chemotherapy in various phase I/II/III clinical trials. Erythrocytes do not have a nucleus; however, they may undergo programmed suicide with characteristic hallmarks including cell shrinkage and phosphatidylserine translocation to the cell surface. This particular death is baptized eryptosis. Our study explored whether BI2536 induces eryptosis. We used flow cytometry to access death in red blood cells. We analyzed the cellular volume, the intracellular calcium concentration, the cell surface phosphatidylserine exposure, and the ceramide abundance. In addition, we analyzed the effect of BI2536 on hemolysis. Our investigation showed that after 48 h of incubation with PLK inhibitor BI2536, erythrocytes lost volume and were positive for annexin­V without any effect on hemolysis. Cells also showed an abundance of ceramide and an increase of intracellular calcium. All these finding suggest that BI2536 provokes eryptosis in red blood cells, ostensibly in part due to Ca2+ entry and ceramide accumulation.


Subject(s)
Erythrocytes , Protein Serine-Threonine Kinases , Pteridines , Protein Serine-Threonine Kinases/antagonists & inhibitors , Humans , Erythrocytes/chemistry , Erythrocytes/cytology , Erythrocytes/drug effects , Eryptosis/drug effects , Pteridines/pharmacology , Ceramides/analysis , Calcium/analysis , Hemolysis/drug effects
8.
Proc Natl Acad Sci U S A ; 119(34): e2207592119, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35969769

ABSTRACT

Vaso-occlusive episode (VOE) is a common and critical complication of sickle cell disease (SCD). Its pathogenesis is incompletely understood. von Willebrand factor (VWF), a multimeric plasma hemostatic protein synthesized and secreted by endothelial cells and platelets, is increased during a VOE. However, whether and how VWF contributes to the pathogenesis of VOE is not fully understood. In this study, we found increased VWF levels during tumor necrosis factor (TNF)-induced VOE in a humanized mouse model of SCD. Deletion of endothelial VWF decreased hemolysis, vascular occlusion, and organ damage caused by TNF-induced VOE in SCD mice. Moreover, administering ADAMTS13, the VWF-cleaving plasma protease, reduced plasma VWF levels, decreased inflammation and vaso-occlusion, and alleviated organ damage during VOE. These data suggest that promoting VWF cleavage via ADAMTS13 may be an effective treatment for reducing hemolysis, inflammation, and vaso-occlusion during VOE.


Subject(s)
Anemia, Sickle Cell , Vascular Diseases , von Willebrand Factor , ADAMTS13 Protein/metabolism , ADAMTS13 Protein/pharmacology , ADAMTS13 Protein/therapeutic use , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Gene Deletion , Hemolysis/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Mice , Vascular Diseases/drug therapy , Vascular Diseases/etiology , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
10.
N Engl J Med ; 386(15): 1432-1442, 2022 04 14.
Article in English | MEDLINE | ID: mdl-35417638

ABSTRACT

BACKGROUND: Pyruvate kinase deficiency is a rare, hereditary, chronic condition that is associated with hemolytic anemia. In a phase 2 study, mitapivat, an oral, first-in-class activator of erythrocyte pyruvate kinase, increased the hemoglobin level in patients with pyruvate kinase deficiency. METHODS: In this global, phase 3, randomized, placebo-controlled trial, we evaluated the efficacy and safety of mitapivat in adults with pyruvate kinase deficiency who were not receiving regular red-cell transfusions. The patients were assigned to receive either mitapivat (5 mg twice daily, with potential escalation to 20 or 50 mg twice daily) or placebo for 24 weeks. The primary end point was a hemoglobin response (an increase from baseline of ≥1.5 g per deciliter in the hemoglobin level) that was sustained at two or more scheduled assessments at weeks 16, 20, and 24. Secondary efficacy end points were the average change from baseline in the hemoglobin level, markers of hemolysis and hematopoiesis, and the change from baseline at week 24 in two pyruvate kinase deficiency-specific patient-reported outcome measures. RESULTS: Sixteen of the 40 patients (40%) in the mitapivat group had a hemoglobin response, as compared with none of the 40 patients in the placebo group (adjusted difference, 39.3 percentage points; 95% confidence interval, 24.1 to 54.6; two-sided P<0.001). Patients who received mitapivat had a greater response than those who received placebo with respect to each secondary end point, including the average change from baseline in the hemoglobin level. The most common adverse events were nausea (in 7 patients [18%] in the mitapivat group and 9 patients [23%] in the placebo group) and headache (in 6 patients [15%] and 13 patients [33%], respectively). Adverse events of grade 3 or higher occurred in 10 patients (25%) who received mitapivat and 5 patients (13%) who received placebo. CONCLUSIONS: In patients with pyruvate kinase deficiency, mitapivat significantly increased the hemoglobin level, decreased hemolysis, and improved patient-reported outcomes. No new safety signals were identified in the patients who received mitapivat. (Funded by Agios Pharmaceuticals; ACTIVATE ClinicalTrials.gov number, NCT03548220.).


Subject(s)
Piperazines , Pyruvate Kinase , Quinolines , Adult , Anemia, Hemolytic, Congenital Nonspherocytic/drug therapy , Double-Blind Method , Hemoglobins/analysis , Hemoglobins/drug effects , Hemolysis/drug effects , Humans , Piperazines/pharmacology , Piperazines/therapeutic use , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/drug therapy , Quinolines/pharmacology , Quinolines/therapeutic use
11.
Mar Drugs ; 20(3)2022 Mar 20.
Article in English | MEDLINE | ID: mdl-35323516

ABSTRACT

Sea stars or starfish (class Asteroidea) and holothurians or sea cucumbers (class Holothuroidea), belonging to the phylum Echinodermata (echinoderms), are characterized by different sets of glycosidic metabolites: the steroid type in starfish and the triterpene type in holothurians. However, herein we report the isolation of eight new triterpene glycosides, pacificusosides D−K (1−3, 5−9) along with the known cucumarioside D (4), from the alcoholic extract of the Far Eastern starfish Solaster pacificus. The isolated new compounds are closely related to the metabolites of sea cucumbers, and their structures of 1−3 and 5−9 were determined by extensive NMR and ESIMS techniques. Compounds 2, 5, and 8 have a new type of tetrasaccharide chain with a terminal non-methylated monosaccharide unit. Compounds 3, 6, and 9 contain another new type of tetrasaccharide chain, having 6-O-SO3-Glc as one of the sugar units. The cytotoxic activity of 1−9 against non-cancerous mouse epidermal cells JB6 Cl41 and human melanoma cell lines SK-MEL-2, SK-MEL-28, and RPMI-7951 was determined by MTS assay. Compounds 1, 3, 4, 6, and 9 showed potent cytotoxicity against these cell lines, but the cancer selectivity (SI > 9) was observed only against the SK-MEL-2 cell line. Compounds 1, 3, 4, 6, and 9 at the non-toxic concentration of 0.1 µM significantly inhibited neoplastic cell transformation of JB6 Cl41 cells induced by chemical carcinogens (EGF, TPA) or ionizing radiation (X-rays and UVB). Moreover, compounds 1 and 4 at the non-toxic concentration of 0.1 µM possessed the highest inhibiting activity on colony formation among the investigated compounds and decreased the colonies number of SK-MEL-2 cells by 64% and 70%, respectively. Thus, triterpene glycosides 1 and 4 can be considered as prospective cancer-preventive and anticancer-compound leaders.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Glycosides/pharmacology , Starfish/chemistry , Triterpenes/pharmacology , Animals , Anticarcinogenic Agents/isolation & purification , Antineoplastic Agents/isolation & purification , Cell Line , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Erythrocytes/drug effects , Glycosides/isolation & purification , Hemolysis/drug effects , Humans , Mice , Triterpenes/isolation & purification
12.
Mar Drugs ; 20(3)2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35323465

ABSTRACT

Among the most potent and proteolytically resistant antimicrobial peptides (AMPs) of animal origin are molecules forming a ß-hairpin structure stabilized by disulfide bonds. In this study, we investigated the mechanism of action and therapeutic potential of the ß-hairpin AMP from the marine polychaeta Capitella teleta, named capitellacin. The peptide exhibits a low cytotoxicity toward mammalian cells and a pronounced activity against a wide range of bacterial pathogens including multi-resistant bacteria, but the mechanism of its antibacterial action is still obscure. In view of this, we obtained analogs of capitellacin and tachyplesin-inspired chimeric variants to identify amino acid residues important for biological activities. A low hydrophobicity of the ß-turn region in capitellacin determines its modest membranotropic activity and slow membrane permeabilization. Electrochemical measurements in planar lipid bilayers mimicking the E. coli membrane were consistent with the detergent-like mechanism of action rather than with binding to a specific molecular target in the cell. The peptide did not induce bacterial resistance after a 21-day selection experiment, which also pointed at a membranotropic mechanism of action. We also found that capitellacin can both prevent E. coli biofilm formation and destroy preformed mature biofilms. The marked antibacterial and antibiofilm activity of capitellacin along with its moderate adverse effects on mammalian cells make this peptide a promising scaffold for the development of drugs for the treatment of chronic E. coli infections, in particular those caused by the formation of biofilms.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Peptides , Polychaeta/chemistry , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Anti-Bacterial Agents/pharmacology , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/isolation & purification , Antimicrobial Peptides/pharmacology , Aquatic Organisms/chemistry , Biofilms/drug effects , Cell Line , Cell Membrane Permeability/drug effects , Erythrocytes/drug effects , Escherichia coli/drug effects , Escherichia coli/physiology , Fibroblasts/drug effects , Hemolysis/drug effects , Humans , Protein Conformation
13.
Bioengineered ; 13(2): 4328-4339, 2022 02.
Article in English | MEDLINE | ID: mdl-35137655

ABSTRACT

Healing of various skin wounds is a lengthy process and often combined with bacterial infection and scar formation. Biomimetic electrospun nanofibrous wound dressing loaded with materials that possess properties of dual antibacterial and tissue repair would be developed to address this problem. In this study, a composite chitosan electrospun nanofibrous material containing Cur@ß-CD/AgNPs nanoparticles composed of silver and curcumin possessed synergic effects on antibacterial activity and wound healing. The developed functionalized silver nanoparticles showed effective activity against both Gram-negative and Gram-positive bacteria. In vivo, Cur@ß-CD/AgNPs chitosan dressing displayed enhanced wound closure rates compared to commercial AquacelAg. Moreover, Cur@ß-CD/AgNPs chitosan dressing contributed to the most uniform collagen distribution by Masson's trichrome staining. In brief, Cur@ß-CD/AgNPs chitosan nanofibers work as a potential wound dressing with antibacterial and antiscarring properties.


Subject(s)
Bandages , Curcumin , Metal Nanoparticles/chemistry , Nanofibers/chemistry , Silver/chemistry , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cells, Cultured , Chitosan/chemistry , Curcumin/chemistry , Curcumin/pharmacokinetics , Curcumin/pharmacology , Electrochemical Techniques , Erythrocytes/drug effects , Hemolysis/drug effects , Male , Mice , Skin/drug effects , Wound Healing/drug effects
14.
Carbohydr Polym ; 282: 119108, 2022 Apr 15.
Article in English | MEDLINE | ID: mdl-35123744

ABSTRACT

A bioinspired chitosan/vitamin E conjugate (Ch/VES, 1:4) was synthesized, optimized based on chitosan's molecular weight (15, 300 kDa), and was assembled to entrap oxaliplatin (OXPt). 1H NMR, infrared spectroscopy, chromatography, X-ray photoelectron spectroscopy, X-ray diffraction, drug release, hemolysis, and stability studies were performed to characterize OXPt@Ch/VES micelles. The therapeutic efficacy of the micelles was tested in vitro in ER+/PR+/HER2- and triple-negative sensitive/resistant breast cancer cells, MCF-7 and MDA-MB-231 via cellular uptake, cytotoxicity, nuclear staining, DNA fragmentation, mitochondrial membrane potential, ROS generation, apoptosis, and cell cycle assays and in vivo using 4T1(Luc)-tumor-bearing mice. OXPt@Ch/VES Ms exhibited decreased IC50 towards MCF-7, MDA-MB-231 (sensitive/resistant) than OXPt. OXPt@Ch/VES Ms caused extensive DNA damage, mitochondrial depolarization, apoptosis, and cell-growth arrest (G2/M). OXPt@Ch/VES Ms treatment retarded tumor growth significantly, prolonged survival, and decreased nephrotoxicity than OXPt. The OXPt@Ch/VES Ms could serve as a potential nanomedicine to overcome conventional OXPt-mediated drug resistance/nephrotoxicity in breast cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Chitosan/administration & dosage , Drug Carriers/administration & dosage , Oxaliplatin/administration & dosage , alpha-Tocopherol/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Chitosan/pharmacokinetics , Drug Carriers/pharmacokinetics , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Erythrocytes/drug effects , Female , Hemolysis/drug effects , Humans , Male , Mice, Inbred BALB C , Micelles , Oxaliplatin/pharmacokinetics , Rats, Wistar , Reactive Oxygen Species/metabolism , alpha-Tocopherol/pharmacokinetics
15.
ACS Appl Mater Interfaces ; 14(9): 11177-11191, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35192338

ABSTRACT

Silk sutures with antibacterial and anti-inflammatory functions were developed for sustained dual-drug delivery to prevent surgical site infections (SSIs). The silk sutures were prepared with core-shell structures braided from degummed silk filaments and then coated with a silk fibroin (SF) layer loaded with berberine (BB) and artemisinin (ART). Both the rapid release of drugs to prevent initial biofilm formation and the following sustained release to maintain effective concentrations for more than 42 days were demonstrated. In vitro assays using human fibroblasts (Hs 865.Sk) demonstrated cell proliferation on the materials, and hemolysis was 2.4 ± 0.8%, lower than that required by ISO 10993-4 standard. The sutures inhibited platelet adhesion and promoted collagen deposition and blood vessel formation. In vivo assessments using Sprague-Dawley (SD) rats indicated that the coating reduced the expression of pro-inflammatory cytokines interleukin-10 (IL-10) and tumor necrosis factor-α (TNF-α), shortening the inflammatory period and promoting angiogenesis. The results demonstrated that these new sutures exhibited stable structures, favorable biocompatibility, and sustainable antibacterial and anti-inflammatory functions with potential for surgical applications.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Silk/chemistry , Silk/pharmacology , Surgical Wound Infection/prevention & control , Sutures , Animals , Anti-Bacterial Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Artemisinins/chemistry , Artemisinins/pharmacology , Artemisinins/therapeutic use , Berberine/chemistry , Berberine/pharmacology , Berberine/therapeutic use , Cell Line , Cell Survival/drug effects , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/therapeutic use , Disease Models, Animal , Drug Liberation , Drug Therapy, Combination/methods , Escherichia coli/drug effects , Hemolysis/drug effects , Humans , Male , Physical Phenomena , Rats, Sprague-Dawley , Silk/therapeutic use , Staphylococcus aureus/drug effects , Surgical Wound Infection/metabolism , Surgical Wound Infection/pathology
16.
Molecules ; 27(4)2022 Feb 11.
Article in English | MEDLINE | ID: mdl-35209001

ABSTRACT

New isopropylated fused azaisocytosine-containing congeners (I-VI) have previously been reported as promising anticancer drug candidates, so further research on these molecules in the preclinical development phase is fully justified and necessary. For this reason, in the present paper, we assess the toxicity/safety profiles of all the compounds using Danio rerio and red blood cell models, and examine the effect of the most selective congeners on the activation of apoptotic caspases in cancer and normal cells. In order to evaluate the effect of each molecule on the development of zebrafish embryos/larvae and to select the safest compounds for further study, various phenotypic parameters (i.e., mortality, hatchability, heart rate, heart oedema, yolk sac utilization, swim bladder development and body shape) were observed, and the half maximal lethal concentration, the maximal non-lethal concentration and no observed adverse effect concentration for each compound were established. The effect of all the isopropylated molecules was compared to that of an anticancer agent pemetrexed. The lipophilicity-dependent structure-toxicity correlations were also determined. To establish the possible interaction of the compounds with red blood cells, an ex vivo hemolysis test was performed. It was shown that almost all of the investigated isopropylated congeners have no adverse phenotypic effect on zebrafish development during five-day exposure at concentrations up to 50 µM (I-III) or up to 20 µM (IV-V), and that they are less toxic for embryos/larvae than pemetrexed, demonstrating their safety. At the same time, all the molecules did not adversely affect the red blood cells, which confirms their very good hemocompatibility. Moreover, they proved to be activators of apoptotic caspases, as they increased caspase-3, -7 and -9 levels in human breast carcinoma cells. The conducted research allows us to select-from among the anticancer active drug candidates-compounds that are safe for developing zebrafish and red blood cells, suitable for further in vivo pharmacological tests.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cytosine/chemistry , Embryo, Nonmammalian/drug effects , Erythrocytes/drug effects , Animals , Cell Line, Tumor , Cytosine/analogs & derivatives , Dose-Response Relationship, Drug , Hemolysis/drug effects , Humans , Molecular Structure , Toxicity Tests , Zebrafish
17.
Molecules ; 27(4)2022 Feb 16.
Article in English | MEDLINE | ID: mdl-35209120

ABSTRACT

(1) Background: Mangiferin (MGN) is a natural compound, showing anti-inflammatory and antioxidant activities for the potential treatment of eye diseases. The poor physicochemical features of MGN (low solubility and high instability) justify its nanoencapsulation into nanostructured lipid carriers (NLC) to improve its ocular bioavailability. (2) Methods: Firstly, MGN-NLC were prepared by the high shear homogenization coupled with the ultrasound (HSH-US) method. Finally, unloaded and MGN-loaded NLC were analyzed in terms of ocular tolerance. (3) Results: MGN-NLC showed good technological parameters suitable for ocular administration (particle size below 200 nm). The ORAC assay was performed to quantify the antioxidant activity of MGN, showing that the antioxidant activity of MGN-NLC (6494 ± 186 µM TE/g) was higher than that of the free compound (3521 ± 271 µM TE/g). This confirmed that the encapsulation of the drug was able to preserve and increase its activity. In ovo studies (HET-CAM) revealed that the formulation can be considered nonirritant. (4) Conclusions: Therefore, NLC systems are a promising approach for the ocular delivery of MGN.


Subject(s)
Drug Carriers/chemistry , Drug Delivery Systems , Nanostructures/chemistry , Nanotechnology , Xanthones/administration & dosage , Administration, Ophthalmic , Antioxidants/administration & dosage , Calorimetry , Eye/drug effects , Hemolysis/drug effects , Lipids/chemistry , Molecular Structure , Nanostructures/ultrastructure , Particle Size , Solubility , Spectrum Analysis
18.
ACS Appl Mater Interfaces ; 14(9): 11116-11123, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35225600

ABSTRACT

Blood-contacting medical devices (BCMDs) are inevitably challenged by thrombi formation, leading to occlusion of flow and device failure. Ideal BCMDs seek to mimic the intrinsic antithrombotic properties of the human vasculature to locally prevent thrombotic complications, negating the need for systemic anticoagulation. An emerging category of BCMD technology utilizes nitric oxide (NO) as a hemocompatible agent, as the vasculature's endothelial layer naturally releases NO to inhibit platelet activation and consumption. In this paper, we report for the first time the novel impregnation of S-nitrosoglutathione (GSNO) into polymeric poly(vinyl chloride) (PVC) tubing via an optimized solvent-swelling method. Material testing revealed an optimized GSNO-PVC material that had adequate GSNO loading to achieve NO flux values within the physiological endothelial NO flux range for a 4 h period. Through in vitro hemocompatibility testing, the optimized material was deemed nonhemolytic (hemolytic index <2%) and capable of reducing platelet activation, suggesting that the material is suitable for contact with whole blood. Furthermore, an in vivo 4 h extracorporeal circulation (ECC) rabbit thrombogenicity model confirmed the blood biocompatibility of the optimized GSNO-PVC. Platelet count remained near 100% for the novel GSNO-impregnated PVC loops (1 h, 91.08 ± 6.27%; 2 h, 95.68 ± 0.61%; 3 h, 97.56 ± 8.59%; 4 h, 95.11 ± 8.30%). In contrast, unmodified PVC ECC loops occluded shortly after the 2 h time point and viable platelet counts quickly diminished (1 h, 85.67 ± 12.62%; 2 h, 54.46 ± 10.53%; 3 h, n/a; 4 h, n/a). The blood clots for GSNO-PVC loops (190.73 ± 72.46 mg) compared to those of unmodified PVC loops (866.50 ± 197.98 mg) were significantly smaller (p < 0.01). The results presented in this paper recommend further investigation in long-term animal models and suggest that GSNO-PVC has the potential to serve as an alternative to systemic anticoagulation in BCMD applications.


Subject(s)
Polymers/pharmacology , S-Nitrosoglutathione/pharmacology , Animals , Blood Coagulation/drug effects , Extracorporeal Circulation/methods , Hemolysis/drug effects , Male , Materials Testing , Models, Animal , Nitric Oxide/chemistry , Nitric Oxide/metabolism , Nitric Oxide/pharmacology , Platelet Activation/drug effects , Polymers/therapeutic use , Polyvinyl Chloride/chemistry , Rabbits , S-Nitrosoglutathione/chemistry , S-Nitrosoglutathione/therapeutic use , Surface Properties , Swine , Thrombosis/prevention & control
19.
Molecules ; 27(3)2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35164286

ABSTRACT

Ultrasound- and microwave-assisted green synthetic strategies were applied to furnish benzofuran-oxadiazole 5a-g and benzofuran-triazole 7a-h derivatives in good to excellent yields (60-96%), in comparison with conventional methods (36-80% yield). These synthesized derivatives were screened for hemolysis, thrombolysis and anticancer therapeutic potential against an A549 lung cancer cell line using an MTT assay. Derivatives 7b (0.1%) and 5e (0.5%) showed the least toxicity against RBCs. Hybrid 7f showed excellent thrombolysis activity (61.4%) when compared against reference ABTS. The highest anticancer activity was displayed by the 5d structural hybridwith cell viability 27.49 ± 1.90 and IC50 6.3 ± 0.7 µM values, which were considerably lower than the reference drug crizotinib (IC50 8.54 ± 0.84 µM). Conformational analysis revealed the spatial arrangement of compound 5d, which demonstrated its significant potency in comparison with crizotinib; therefore, scaffold 5d would be a promising anticancer agent on the basis of cytotoxicity studies, as well as in silico modeling studies.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Oxadiazoles/pharmacology , Triazoles/pharmacology , A549 Cells , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzofurans/chemical synthesis , Benzofurans/chemistry , Cell Survival/drug effects , Chemistry Techniques, Synthetic , Hemolysis/drug effects , Humans , Microwaves , Molecular Docking Simulation , Neoplasms/drug therapy , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Triazoles/chemical synthesis , Triazoles/chemistry
20.
ACS Appl Mater Interfaces ; 14(9): 11144-11155, 2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35195389

ABSTRACT

Bacterial infections are a common problem associated with wound treatment that imposes a significant burden on healthcare systems and patients. As a result, healthcare providers urgently need new treatment strategies to protect people. Hydrogel biomaterials with inherent antimicrobial properties offer an attractive and viable solution to this issue. Here, for the first time, we have developed a new efficient synthetic strategy to prepare cationic hydrogels (PHCI) with intrinsically efficient antimicrobial properties by chemically cross-linking trans-1,4-cyclohexanediamine with 1,3-dibromo-2-propanol using a condensation reaction without the use of toxic cross-linking agents. As expected, the prepared PHCI hydrogel possessed an inherent antibacterial ability that can adsorb and kill Staphylococcus aureus and Escherichia coli electrostatically. Notably, in vivo experiments on normal and diabetic rat models confirmed that the PHCI hydrogel can quickly stop bleeding, efficiently kill bacteria, promote the conversion of macrophages from the proinflammatory M1 phenotype to the repaired M2 phenotype, and accelerate collagen deposition and blood vessel formation, thereby achieving rapid wound healing. Overall, this work presents an effective antibacterial dressing that might provide a facile but effective approach for clinical wound management.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Infections/drug therapy , Hydrogels/chemistry , Hydrogels/pharmacology , Wound Healing/drug effects , Wound Infection/drug therapy , Animals , Anti-Bacterial Agents/therapeutic use , Cell Line , Cell Survival/drug effects , Diabetes Mellitus, Experimental/complications , Escherichia coli/drug effects , Hemolysis/drug effects , Hemostatics/chemistry , Hemostatics/pharmacology , Hemostatics/therapeutic use , Hydrogels/therapeutic use , Male , Mice , Rats, Sprague-Dawley , Staphylococcus aureus/drug effects , Wound Infection/complications , Wound Infection/metabolism , Wound Infection/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...