Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Int J Clin Pharm ; 46(2): 390-400, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38147281

ABSTRACT

BACKGROUND: Nutritional deficit and oral iron gastrointestinal intolerance may be a common cause of iron deficiency, which can be managed by pharmacists. AIM: To understand the prevalence of iron deficiency in women of childbearing age with a self-reported history of intolerance to oral iron and the tolerability of three doses of an iron-whey-protein formulation in the care of these women. METHOD: Ferritin and haemoglobin levels were documented in women of childbearing age with oral iron gastrointestinal intolerance. In those with iron deficiency (ferritin < 30 µg/L), adherence, gastrointestinal tolerability, ferritin, transferrin saturation and haemoglobin levels were compared between their prior oral iron product and iron-whey-protein microspheres randomised to three doses (14 mg daily, 25 mg daily and 50 mg daily) for 12 weeks. RESULTS: Most screened women had low iron stores (128 (62.7%); ferritin < 30 µg/L), 65 (31.9%) had moderate to severe iron deficiency (ferritin < 12 µg/L) and 33 (16.2%) had iron deficiency anaemia (ferritin < 30 µg/L, haemoglobin < 12 g/dL). Amongst the 59 women who participated in the prospective clinical study of iron-whey-protein microspheres over 12 weeks, 48 (81.4%) were classified as adherent/persistent and fewer instances of gastrointestinal intolerance were reported (0.59 ± 0.91) when compared to 12 (20.3%) and (4.0 ± 2.2) respectively while taking the prior oral iron (Fisher's Exact and T-test respectively, both p < 0.001). There was no difference in adherence or tolerability of different iron-whey-protein formulation doses. Ferritin, haemoglobin and energy levels increased significantly over 12 weeks. CONCLUSION: Undiagnosed iron deficiency is common in women of childbearing age with a history of intolerance to oral iron and iron-whey-protein microspheres can improve adherence, GI tolerability, iron stores, haemoglobin and energy levels in these women. CLINICAL TRIAL REGISTRATION: Clinicaltrials.gov identifier (registration includes full trial protocol): NCT04778072.


Subject(s)
Anemia, Iron-Deficiency , Iron Deficiencies , Female , Humans , Iron/adverse effects , Prospective Studies , Whey/metabolism , Anemia, Iron-Deficiency/diagnosis , Anemia, Iron-Deficiency/drug therapy , Anemia, Iron-Deficiency/epidemiology , Ferritins , Hemoglobins/metabolism
2.
Int J Mol Sci ; 24(21)2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37958838

ABSTRACT

Multiple myeloma (MM) is a hematological malignancy originated in the bone marrow and characterized by unhindered plasma cell proliferation that results in several clinical manifestations. Although the main role of blood platelets lies in hemostasis and thrombosis, platelets also play a pivotal role in a number of other pathological conditions. Platelets are the less-explored components from the tumor microenvironment in MM. Although some studies have recently revealed that MM cells have the ability to activate platelets even in the premalignant stage, this phenomenon has not been widely investigated in MM. Moreover, thrombocytopenia, along with bleeding, is commonly observed in those patients. In this review, we discuss the hemostatic disturbances observed in MM patients and the dynamic interaction between platelets and myeloma cells, along with present and future potential avenues for the use of platelets for diagnostic and therapeutic purposes.


Subject(s)
Multiple Myeloma , Thrombosis , Humans , Blood Platelets/physiology , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Hemorrhage , Hemostasis , Thrombosis/etiology , Cell Communication , Drug Delivery Systems , Tumor Microenvironment
3.
BMC Cancer ; 22(1): 1023, 2022 Sep 29.
Article in English | MEDLINE | ID: mdl-36171564

ABSTRACT

BACKGROUND: Cancer patients have an increased risk of developing venous thromboembolism, with up to 30% dying within a month of their development. Some cancer cells are known to induce platelet aggregation, and this interaction is understood to contribute to thrombosis and haematogenous metastasis. Many researchers have reported on extracellular vesicles (EVs) released from platelets. However, less is known about how cancer cells' EVs may affect platelet function. Here EVs released by triple-negative breast cancer (TNBC) cell line variants were extensively investigated in this regard. METHODS: EVs were separated from conditioned media of TNBC Hs578T and Hs578Ts(i)8 cells using filtration and ultracentrifugation and were characterised by nanoparticle tracking analysis, immunoblots, and transmission electron microscopy. Blood samples from consenting donors were procured, and their platelets collected by differential centrifugation. Light transmission aggregometry and optical microscopy evaluated the potential interaction of TNBC cells and their EVs with platelets. Global proteomic analysis was performed on the EVs, by in-solution digestion and mass spectrometry. Data analysis included the use of Perseus, FunRich, and Vesiclepedia. Immunoblotting was used as a secondary method to investigate some key EV cargo proteins identified by the global proteomics approach. RESULTS: Both TNBC cell variants induced platelet aggregation. Increasing cell numbers significantly reduced the time taken for platelet aggregation to occur. EVs released by the cells also resulted in platelet aggregation. The time to induce platelet aggregation was EV dose-dependent. Proteomics profiling and immunoblotting of the EVs' cargo identified candidate proteins (including uPAR and PDGFRß) that may be involved during this process. CONCLUSIONS: TNBC cells induce platelet aggregation. Furthermore, the cell-free EVs induced this undesirable effect. A number of EV cargo proteins were identified that may be relevant as therapeutic targets.


Subject(s)
Extracellular Vesicles , Triple Negative Breast Neoplasms , Culture Media, Conditioned/metabolism , Extracellular Vesicles/metabolism , Humans , Platelet Aggregation , Proteomics/methods , Triple Negative Breast Neoplasms/metabolism
4.
Int J Nanomedicine ; 17: 4383-4400, 2022.
Article in English | MEDLINE | ID: mdl-36164554

ABSTRACT

Purpose: In the search for new drug delivery platforms for cardiovascular diseases and coating of medical devices, we synthesized eptifibatide-functionalized silver nanoparticles (AgNPs-EPI) and examined the pharmacological activity of AgNPs-EPI on platelets and endothelial cells in vitro and ex vivo. Methods: Spherical AgNPs linked to eptifibatide were synthesized and characterized. Cytotoxicity was measured in microvascular endothelial cells (HMEC-1), platelets and red blood cells. Platelet mitochondrial respiration was measured using the Oxygraph-2k, a high-resolution modular respirometry system. The effect of AgNPs-EPI on the aggregation of washed platelets was measured by light aggregometry and the ex vivo occlusion time was determined using a reference laboratory method. The surface amount of platelet receptors such as P-selectin and GPIIb/IIIa was measured. The influence of AgNPS-EPI on blood coagulation science was assessed. Finally, the effect of AgNPs-EPI on endothelial cells was measured by the levels of 6-keto-PGF1alpha, tPa, cGMP and vWF. Results: We describe the synthesis of AgNPs using eptifibatide as the stabilizing ligand. The molecules of this drug are directly bonded to the surface of the nanoparticles. The synthesized AgNPs-EPI did not affect the viability of platelets, endothelial cells and erythrocytes. Preincubation of platelets with AgNPs-EPI protected by mitochondrial oxidative phosphorylation capacity. AgNPs-EPI inhibited aggregation-induced P-selectin expression and GPIIb/IIIa conformational changes in platelets. AgNPs-EPI caused prolongation of the occlusion time in the presence of collagen/ADP and collagen/adrenaline. AgNPs-EPI regulated levels of 6-keto-PGF1alpha, tPa, vWf and cGMP produced in thrombin stimulated HMEC-1 cells. Conclusion: AgNPs-EPI show anti-aggregatory activity at concentrations lower than those required by the free drug acting via regulation of platelet aggregation, blood coagulation, and endothelial cell activity. Our results provide proof-of-principle evidence that AgNPs may be used as an effective delivery platform for antiplatelet drugs.


Subject(s)
Metal Nanoparticles , P-Selectin , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Blood Platelets , Collagen/metabolism , Endothelial Cells/metabolism , Epinephrine/metabolism , Epinephrine/pharmacology , Eptifibatide/pharmacology , Ligands , P-Selectin/metabolism , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Silver/metabolism , Silver/pharmacology , Thrombin/metabolism , von Willebrand Factor/metabolism
5.
Eur J Pharm Sci ; 175: 106236, 2022 Aug 01.
Article in English | MEDLINE | ID: mdl-35710078

ABSTRACT

Current pathophysiological findings indicate that damage to the alveolar epithelium plays a decisive role in the development of idiopathic pulmonary fibrosis (IPF). The available pharmacological interventions (i.e., oral pirfenidone and nintedanib) only slow down progression of the disease, but do not offer a cure. In order to develop new drug candidates, the pathophysiology of IPF needs to be better understood on a molecular level. It has previously been reported that a loss of caveolin-1 (Cav-1) contributes to profibrotic processes by causing reduced alveolar barrier function and fibrosis-like alterations of the lung-parenchyma. Conversely, overexpression of caveolin-1 appears to counteract the development of fibrosis by inhibiting the inflammasome NLRP3 and the associated expression of interleukin-1ß. In this study, the interaction between Fyn-kinase and caveolin-1 in the alveolar epithelium of various bleomycin (BLM)/TGF-ß damage models using precision-cut lung slices (PCLS), wildtype (WT) and caveolin-1 knockout (KO) mice as well as the human NCI-H441 cell line, were investigated. In WT mouse lung tissues, strong signals for Fyn-kinase were detected in alveolar epithelial type I cells, whereas in caveolin-1 KO animals, expression shifted to alveolar epithelial type II cells. Caveolin-1 and Fyn-kinase were found to be co-localized in isolated lipid rafts of NCI-H441 cell membrane fractions. These findings were corroborated by co-immunoprecipitation studies in which a co-localization of Cav-1 and Fyn-kinase was detected in the cell membrane of the alveolar epithelium. After TGF-ß and BLM-induced damage to the alveolar epithelium both in PCLS and cell culture experiments, a decrease in caveolin-1 and Fyn-kinase was found. Furthermore, TEER (transepithelial electrical resistance) measurements indicated that TGF-ß and BLM have a damaging effect on cell-cell contacts and thus impair the barrier function in NCI-H441 cell monolayers. This effect was attenuated after co-incubation with the Fyn-kinase inhibitor, PP-2. Our data suggest an involvement of Fyn-kinase and caveolin-1 in TGF-ß/bleomycin-induced impairment of alveolar barrier function and thus a possible role in the early stages of pulmonary fibrosis. Fyn-kinase and/or its complex with caveolin-1 might, therefore, be novel therapeutic targets in IPF.


Subject(s)
Alveolar Epithelial Cells , Caveolin 1 , Idiopathic Pulmonary Fibrosis , Proto-Oncogene Proteins c-fyn , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Bleomycin/pharmacology , Caveolin 1/metabolism , Fibrosis , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-fyn/metabolism , Transforming Growth Factor beta/metabolism
6.
Life Sci ; 290: 120236, 2022 Feb 01.
Article in English | MEDLINE | ID: mdl-34953891

ABSTRACT

AIMS: We have recently described a novel guanidinium-based compound, VP79s, which induces cytotoxicity in various cancer cell lines. Here, we aim to investigate the activity of VP79s and associated mechanisms of action in multiple myeloma (MM) cells in vitro and ex vivo. MAIN METHODS: The effects of VP79s on cell viability and induction of apoptosis was examined in a panel of drug-sensitive and drug-resistant MM cell lines, as well as ex vivo patient samples and normal donor lymphocytes and platelets. Cell signaling pathways associated with the biological effects of VP79s were analysed by immunoblotting and flow cytometry. Gene expression changes were assessed by quantitative real-time PCR analysis. KEY FINDINGS: VP79s was found to rapidly inhibit both constitutively active and IL-6-induced STAT3 signaling with concurrent downregulation of the IL-6 receptors, CD130 and CD126. VP79s induced a rapid and dose-dependent downregulation of anti-apoptotic Bcl-2 family member, myeloid cell leukaemia-1 (MCL-1). VP79s enhanced bortezomib induced cell death and was also found to overcome bone marrow stromal cell induced drug resistance. VP79s exhibited activity in ex vivo patient samples at concentrations which had no effect on peripheral blood mononuclear cells, lymphocytes and platelets isolated from healthy donors. SIGNIFICANCE: As VP79s resulted in rapid inhibition of the key IL-6/STAT3 signaling pathway and downregulation of MCL-1 expression with subsequent selective anti-myeloma activity, VP79s may be a potential therapeutic agent with a novel mechanism of action in MM cells.


Subject(s)
Guanidine/pharmacology , Multiple Myeloma/drug therapy , STAT3 Transcription Factor/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression/genetics , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Guanidine/analogs & derivatives , Humans , Interleukin-6/metabolism , Janus Kinase 1/metabolism , Janus Kinases/metabolism , Leukemia/drug therapy , Leukocytes, Mononuclear/metabolism , Multiple Myeloma/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/drug effects , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Myeloid Cells , STAT3 Transcription Factor/drug effects , Signal Transduction/drug effects
7.
Front Pharmacol ; 12: 733743, 2021.
Article in English | MEDLINE | ID: mdl-35153735

ABSTRACT

Purpose: To study and compare the antibacterial properties and the potential cytotoxic effects of commercially available uncoated silver nanoparticles (AgNPs) with lipoic acid coated silver nanoparticles (AgNPsLA) developed by our group. The antibacterial, cytotoxic, and hemolytic properties of those NPs were assessed with the main objective of investigating if AgNPsLA could maintain their antibacterial properties while improving their biosafety profile over uncoated AgNPs within the blood vessel's microenvironment. Methods: Comercially available uncoated 2.6 nm AgNPs and 2.5 nm AgNPsLA synthesized and characterized as previously described by our group, were used in this study. Antimicrobial activity was assessed on a wide range of pathogens and expressed by minimal inhibitory concentrations (MIC). Assessment of cytotoxicity was carried out on human umbilical vein endothelial cells (HUVEC) using an MTT test. Detection of reactive oxygen species, cell apoptosis/necrosis in HUVEC, and measurement of mitochondrial destabilization in HUVEC and platelets were performed by flow cytometry. The potential harmful effect of nanoparticles on red blood cells (RBCs) was investigated measuring hemoglobin and LDH released after exposure to NPs. Transmission electron microscopy was also used to determine if AgNPs and AgNPsLA could induce any ultrastructural changes on HUVEC cells and Staphylococcus aureus bacteria. Results: AgNPs and AgNPsLA had antimicrobial properties against pathogens associated with catheter-related bloodstream infections. AgNPs, in contrast to AgNPsLA, induced ROS production and apoptosis in HUVEC, ultrastructural changes in HUVEC and S. aureus, depolarization of mitochondrial membrane in HUVEC and platelets, and also hemolysis. Conclusion: AgNPsLA synthesized by our group have antimicrobial activity and a better biosafety profile than uncoated AgNPs of similar size. Those observations are of critical importance for the future in vivo investigations and the potential application of AgNPsLA in medical devices for human use.

8.
Pharmaceutics ; 12(8)2020 Aug 06.
Article in English | MEDLINE | ID: mdl-32781726

ABSTRACT

The crystallization of poorly soluble drug molecules with an excipient into new solid phases called cocrystals has gained a considerable popularity in the pharmaceutical field. In this work, the cocrystal approach was explored for a very poorly water soluble antifungal active, itraconazole (ITR), which was, for the first time, successfully converted into this multicomponent solid using an aromatic coformer, terephthalic acid (TER). The new cocrystal was characterized in terms of its solid-state and structural properties, and a panel of pharmaceutical tests including wettability and dissolution were performed. Evidence of the cocrystal formation was obtained from liquid-assisted grinding, but not neat grinding. An efficient method of the ITR-TER cocrystal formation was ball milling. The stoichiometry of the ITR-TER phase was 2:1 and the structure was stabilized by H-bonds. When comparing ITR-TER with other cocrystals, the intrinsic dissolution rates and powder dissolution profiles correlated with the aqueous solubility of the coformers. The rank order of the dissolution rates of the active pharmaceutical ingredient (API) from the cocrystals was ITR-oxalic acid > ITR-succinic acid > ITR-TER. Additionally, the ITR-TER cocrystal was stable in aqueous conditions and did not transform to the parent drug. In summary, this work presents another cocrystal of ITR that might be of use in pharmaceutical formulations.

9.
Pharmaceutics ; 12(3)2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32178442

ABSTRACT

The use of nanocarriers is being researched to achieve oral peptide delivery. Insulin-associated anionic polyelectrolyte nanoparticle complexes (PECs) were formed that comprised hyaluronic acid and chitosan in an optimum mass mixing ratio of 5:1 (MR 5), followed by coating with a pH-dependent polymer. Free insulin was separated from PECs by size exclusion chromatography and then measured by HPLC. The association efficiency of insulin in PECs was >95% and the loading was ~83 µg/mg particles. Dynamic light scattering and nanoparticle tracking analysis of PECs revealed low polydispersity, a negative zeta potential range of -40 to -50 mV, and a diameter range of 95-200 nm. Dissolution studies in simulated small intestinal fluid (FaSSIF-V2) revealed that the PECs were colloidally stable. PECs that were coated with Eudragit® L-100 delayed insulin release in FaSSIF-V2 and protected insulin against pancreatin attack more than uncoated PECs. Uncoated anionic PECs interacted weakly with mucin in vitro and were non-cytotoxic to Caco-2 cells. The coated and uncoated PECs, both concentrated further by ultrafiltration, permitted dosing of 50 IU/kg in rat jejunal instillations, but they failed to reduce plasma glucose or deliver insulin to the blood. When ad-mixed with the permeation enhancer (PE), sucrose laurate (100 mM), the physicochemical parameters of coated PECs were relatively unchanged, however blood glucose was reduced by 70%. In conclusion, the use of a PE allowed for the PEC-released bioactive insulin to permeate the jejunum. This has implications for the design of orally delivered particles that can release the payload when formulated with enhancers.

10.
Front Cardiovasc Med ; 6: 139, 2019.
Article in English | MEDLINE | ID: mdl-31620449

ABSTRACT

Nanomaterials have been recently introduced as potential diagnostic and therapeutic tools in the medical field. One of the main concerns in relation to the use of nanomaterials in humans is their potential toxicity profile and blood compatibility. In fact, and due to their small size, NPs can translocate into the systemic circulation even after dermal contact, inhalation, or oral ingestion. Once in the blood stream, nanoparticles become in contact with the different components of the blood and can potentially interfere with normal platelet function leading to bleeding or thrombosis. Metallic NPs have been already used for diagnosis and treatment purposes due to their unique characteristics. However, the potential interactions between metallic NPs and platelets has not been widely studied and reported. This review focuses on the factors that can affect platelet activation and aggregation by metal NPs and the nature of such interactions, providing a summary of the effect of various metal NPs on platelet function available in the literature.

11.
Int J Nanomedicine ; 14: 7399-7417, 2019.
Article in English | MEDLINE | ID: mdl-31571858

ABSTRACT

PURPOSE: We studied the effects of silver nanoparticles (AgNPs) on human blood platelet function. We hypothesized that AgNPs, a known antimicrobial agent, can be used as blood-compatible, "ideal material'' in medical devices or as a drug delivery system. Therefore, the aim of the current study was to investigate if functionalized AgNPs affect platelet function and platelets as well as endothelial cell viability in vitro. METHODS: AgNPs, functionalized with reduced glutathione (GSH), polyethylene glycol (PEG) and lipoic acid (LA) were synthesized. Quartz crystal microbalance with dissipation was used to measure the effect of AgNPs on platelet aggregation. Platelet aggregation was measured by changes in frequency and dissipation, and the presence of platelets on the sensor surface was confirmed and imaged by phase contrast microscopy. Flow cytometry was used to detect surface abundance of platelet receptors. Lactate dehydrogenase test was used to assess the potential cytotoxicity of AgNPs on human blood platelets, endothelial cells, and fibroblasts. Commercially available ELISA tests were used to measure the levels of thromboxane B2 and metalloproteinases (MMP-1, MMP-2) released by platelets as markers of platelet activation. RESULTS: 2 nm AgNPs-GSH, 3.7 nm AgNPs-PEG both at 50 and 100 µg/mL, and 2.5 nm AgNPs-LA at 100 µg/mL reduced platelet aggregation, inhibited collagen-mediated increase in total P-selectin and GPIIb/IIIa, TXB2 formation, MMP-1, and MMP-2 release. The tested AgNPs concentrations were not cytotoxic as they did not affect, platelet, endothelial cell, or fibroblast viability. CONCLUSION: All tested functionalized AgNPs inhibited platelet aggregation at nontoxic concentrations. Therefore, functionalized AgNPs can be used as an antiplatelet agent or in design and manufacturing of blood-facing medical devices, such as vascular grafts, stents, heart valves, and catheters.


Subject(s)
Blood Platelets/drug effects , Metal Nanoparticles/chemistry , Platelet Aggregation/drug effects , Silver/pharmacology , Collagen/metabolism , Fibroblasts/drug effects , Flow Cytometry , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Ligands , Matrix Metalloproteinases/metabolism , Metal Nanoparticles/ultrastructure , P-Selectin/metabolism , Particle Size , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Polyethylene Glycols/chemistry , Quartz Crystal Microbalance Techniques , Spectroscopy, Fourier Transform Infrared , Thromboxane B2/metabolism
12.
Eur J Pharm Biopharm ; 144: 57-67, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31493509

ABSTRACT

Itraconazole (ITR) is a broad-spectrum antifungal drug with a very low solubility. In this work, the application of a heat induced evaporative antisolvent nanoprecipitation process yielded disordered nanoparticles (NPs) of ITR. The inclusion of different types of poly(ethylene glycol) (PEG) allowed PEGylation of NPs by adsorption to be achieved. The NP dispersions were composed of monodispersed particles in a nanometric size range (<290 nm) and although PEGylation had no impact on the average particle size, the surface potential was partially neutralised in the modified NPs. The solid state analysis using powder X-ray diffraction and thermal analysis revealed a disordered, liquid crystalline smectic organisation of the non-PEGylated NPs, while some of the PEGylated NPs were more crystalline. The PEGylated NPs exhibited mucoadhesive potential in stationary conditions (dynamic light scattering analysis) but when flow conditions were applied (nanoparticle tracking analysis and quartz crystal microbalance with dissipation) the particles had mucopenetrative properties. The non-PEGylated ITR NPs did not interact with mucin and therefore, this system was considered as having a mucopenetrative character. This study demonstrates that the properties of NPs made of organic drug molecules can be modified by the addition of polymers, which may impact on their interaction with mucin and therefore on their potential systemic absorption.


Subject(s)
Adhesives/chemistry , Itraconazole/chemistry , Nanoparticles/chemistry , Polyethylene Glycols/chemistry , Polymers/chemistry , Adsorption/drug effects , Antifungal Agents/chemistry , Drug Carriers/chemistry , Drug Compounding/methods , Particle Size , Solubility/drug effects , Surface Properties/drug effects
13.
Int J Pharm ; 552(1-2): 27-38, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30236648

ABSTRACT

This work investigates the impact of nanoparticle (NP) composition and effectiveness of cryo-/lyo-protectants in a freeze drying process, which was employed to convert liquid dispersions of polyelectrolyte complex (PEC) NPs into completely redispersible powders. PEC NPs, with and without peptide, were produced by complex coacervation. The cryo-/lyo-protectants investigated were mannitol, trehalose (TRE) and poly(ethylene glycol) (PEG). The solid state of lyophilised powders was studied by thermal analysis and X-ray diffraction. Cytotoxicity studies were done by MTS assay and flow cytometry. The presence of a cryoprotectant was essential to achieve a successful powder reconstitution. The concentration of TRE was optimised for each type of PEC NPs. Protamine- and hyaluronate-based NPs reconstituted better than chitosan- and chondroitin sulphate-based NPs, respectively. PEG polymers were found to be more effective cryoprotectants than TRE and best results were achieved using co-freeze drying of NPs with TRE and PEG. These ternary NPs/TRE/PEG samples were crystalline, with expected better storage stability. PEG polymers were well tolerated by Caco-2 cells, with the exception of linear PEG 10 kDa. This work shows that, as regards the formulation design and maximising NP loading in the dried product, optimisation of the cryoprotectant type and content is needed as it is highly dependent not only on the type of polyelectrolyte pair in the PEC, but also the polyions ratio.


Subject(s)
Chitosan/chemistry , Chondroitin Sulfates/chemistry , Cryoprotective Agents/chemistry , Hyaluronic Acid/chemistry , Nanoparticles/chemistry , Protamines/chemistry , Caco-2 Cells , Cell Survival/drug effects , Chitosan/administration & dosage , Chondroitin Sulfates/administration & dosage , Cryoprotective Agents/administration & dosage , Freeze Drying , Humans , Hyaluronic Acid/administration & dosage , Nanoparticles/administration & dosage , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Protamines/administration & dosage , Trehalose/administration & dosage , Trehalose/chemistry
14.
PLoS One ; 13(5): e0196478, 2018.
Article in English | MEDLINE | ID: mdl-29734352

ABSTRACT

BACKGROUND: The development of thrombocytopenia in sepsis is a poor prognostic indicator associated with a significantly increased mortality risk. Mechanisms underlying this phenomenon remain to be clearly elucidated. Matrix metalloproteinases (MMPs) are enzymes that regulate the turnover of the extra-cellular matrix. MMP-2 is recognised as a platelet agonist with MMP-9 proposed as an inhibitor of platelet activation. The existence of MMP-9 in platelets is a subject of debate. There is limited evidence thus far to suggest that toll-like receptor 4 (TLR-4) and platelet-leukocyte aggregate (PLA) formation may be implicated in the development of sepsis-associated thrombocytopenia. OBJECTIVES: To investigate whether MMP -2/-9, toll-like receptor 4 (TLR-4) or platelet-leukocyte aggregate (PLA) formation are implicated in a decline in platelet numbers during septic shock. METHODS: This was an observational study which recruited healthy controls, non-thrombocytopenic septic donors and thrombocytopenic septic donors. MMP-2, MMP-9 and TLR-4 platelet surface expression as well as PLA formation was examined using flow cytometry. In addition MMP-2 and MMP-9 were examined by gelatin zymography and enzyme-linked immunosorbent assay (ELISA) using a 3 compartment model (plasma, intraplatelet and platelet membrane). RESULTS: There was no difference found in MMP-2, MMP-9 or TLR-4 levels between non-thrombocytopenic and thrombocytopenic septic donors. PLA formation was increased in thrombocytopenic patients. MMP-9 was detected in platelets using flow cytometry, gelatin zymography and ELISA techniques. CONCLUSIONS: Platelet consumption into PLAs may account for the development of thrombocytopenia in septic shock. MMP-9 is found in platelets and it is upregulated during septic shock.


Subject(s)
Blood Platelets/pathology , Leukocytes/pathology , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 9/blood , Shock, Septic/blood , Thrombocytopenia/blood , Toll-Like Receptor 4/blood , Blood Platelets/enzymology , Blood Platelets/metabolism , Case-Control Studies , Female , Humans , Leukocytes/enzymology , Leukocytes/metabolism , Male , Middle Aged , Platelet Activation/physiology , Platelet Count , Shock, Septic/enzymology , Shock, Septic/pathology , Thrombocytopenia/enzymology , Thrombocytopenia/pathology
15.
Platelets ; 29(3): 301-304, 2018 May.
Article in English | MEDLINE | ID: mdl-29120698

ABSTRACT

Antiplatelet therapies remain an area of potential interest for the treatment of sepsis; however, studies of platelet aggregation in sepsis have yielded conflicting results. We examined platelet aggregation patterns in patients with septic shock using quartz crystal microbalance with dissipation technology, a microfluidic device capable of measuring platelet microaggregate formation under flow conditions. Platelet aggregation was increased in the washed platelet samples of septic patients. Conversely, these same platelets aggregated less than healthy controls when examined in their plasma.


Subject(s)
Blood Platelets/metabolism , Platelet Aggregation , Platelet Function Tests , Sepsis/blood , Sepsis/diagnosis , Aged , Analysis of Variance , Blood Platelets/chemistry , Case-Control Studies , Female , Humans , Male , Middle Aged , Platelet Function Tests/instrumentation , Platelet Function Tests/methods , Time Factors
16.
Cardiovasc Res ; 113(14): 1719-1731, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29016749

ABSTRACT

AIMS: In addition to maintaining haemostasis, circulating blood platelets are the cellular culprits that form occlusive thrombi in arteries and veins. Compared to blood leucocytes, which exist as functionally distinct subtypes, platelets are considered to be relatively simple cell fragments that form vascular system plugs without a differentially regulated cellular response. Hence, investigation into platelet subpopulations with distinct functional roles in haemostasis/thrombosis has been limited. In our present study, we investigated whether functionally distinct platelet subpopulations exist based on their ability to generate and respond to nitric oxide (NO), an endogenous platelet inhibitor. METHODS AND RESULTS: Utilizing highly sensitive and selective flow cytometry protocols, we demonstrate that human platelet subpopulations exist based on the presence and absence of endothelial nitric oxide synthase (eNOS). Platelets lacking eNOS (approximately 20% of total platelets) fail to produce NO and have a down-regulated soluble guanylate cyclase-protein kinase G (sGC-PKG)-signalling pathway. In flow chamber and aggregation experiments eNOS-negative platelets primarily initiate adhesion to collagen, more readily activate integrin αIIbß3 and secrete matrix metalloproteinase-2, and form larger aggregates than their eNOS-positive counterparts. Conversely, platelets having an intact eNOS-sGC-PKG-signalling pathway (approximately 80% of total platelets) form the bulk of an aggregate via increased thromboxane synthesis and ultimately limit its size via NO generation. CONCLUSION: These findings reveal previously unrecognized characteristics and complexity of platelets and their regulation of adhesion/aggregation. The identification of platelet subpopulations also has potentially important consequences to human health and disease as impaired platelet NO-signalling has been identified in patients with coronary artery disease.


Subject(s)
Blood Platelets/metabolism , Nitric Oxide Synthase Type III/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Animals , Blood Platelets/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Nitric Oxide/metabolism
18.
Nanomedicine (Lond) ; 10(9): 1451-75, 2015 May.
Article in English | MEDLINE | ID: mdl-25996119

ABSTRACT

In physiological conditions, the interactions between blood platelets and endothelial cells play a major role in vascular reactivity and hemostasis. By contrast, increased platelet activation contributes to the pathogenesis of vascular pathology such as atherosclerosis, thrombosis, diabetes mellitus, hypertension and carcinogenesis. Nanomedicine, including nanodiagnostics and nanotherapeutics is poised to be used in the management of vascular diseases. However, the inherent risk and potential toxicity resultant from the use of nanosized (<100 nm) materials need to be carefully considered. This review, basing on a systematic search of literature provides state-of-the-art and focuses on new discoveries, as well as the potential benefits and threats in the field of nanodiagnostics, nanopharmacology and nanotoxicology of platelet-vessel wall interactions.


Subject(s)
Blood Platelets/cytology , Cell Adhesion , Nanotechnology , Tunica Intima/cytology , Blood Platelets/ultrastructure , Humans , Nanoparticles , Vascular Diseases/therapy
19.
Eur J Pharm Biopharm ; 93: 242-53, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25907005

ABSTRACT

A new type of self-assembled polyelectrolyte complex nanocarrier composed of chondroitin (CHON) and protamine (PROT) was designed and the ability of the carriers to bind salmon calcitonin (sCT) was examined. The response of sCT-loaded CHON/PROT NPs to a change in the properties of the liquid medium, e.g. its pH, composition or ionic strength was studied and in vitro peptide release was assessed. The biocompatibility of the NPs was evaluated in Caco-2 cells. CHON/PROT NPs were successfully obtained with properties that were dependent on the concentration of the polyelectrolytes and their mixing ratio. X-ray diffraction determined the amorphous nature of the negatively charged NPs, while those with the positive surface potential were semi-crystalline. sCT was efficiently associated with the nanocarriers (98-100%) and a notably high drug loading (13-38%) was achieved. The particles had negative zeta potential values and were homogenously dispersed with sizes between 60 and 250 nm. CHON/PROT NPs released less than 10% of the total loaded peptide in the first hour of the in vitro release studies. The enthalpy of the decomposition exotherm correlated with the amount of sCT remaining in NPs after the release experiments. The composition of medium and its ionic strength was found to have a considerable influence on the release of sCT from CHON/PROT NPs. Complexation to CHON markedly reduced the toxic effects exerted by PROT and the NPs were compatible and well tolerated by Caco-2 cells.


Subject(s)
Calcitonin/chemistry , Chondroitin/chemistry , Drug Carriers , Nanoparticles , Protamines/chemistry , Caco-2 Cells , Calcitonin/administration & dosage , Calcitonin/metabolism , Cell Survival/drug effects , Chemistry, Pharmaceutical , Chondroitin/analogs & derivatives , Chondroitin/metabolism , Chondroitin/toxicity , Crystallography, X-Ray , Delayed-Action Preparations , Humans , Hydrogen-Ion Concentration , Kinetics , Nanomedicine , Osmolar Concentration , Protamines/metabolism , Protamines/toxicity , Protein Binding , Solubility , Surface Properties , Technology, Pharmaceutical/methods
20.
Toxicol In Vitro ; 29(4): 663-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25701151

ABSTRACT

Several studies have reported that CuO nanoparticles (CuONPs) have the capacity to cross the blood brain barrier and exert a toxic effect. The aims of our study were to investigate mechanisms underlying CuONPs-induced neurotoxicity in vitro and neuroprotective effects of crocetin. We investigated the toxicological effects of exposure of HT22 hippocampal cells to CuONPs (31 nm) in the presence or absence of crocetin. Crocetin is a carotenoid with wide spectrum of pharmacological effects and the ability to cross blood-brain barrier. Exposure of HT22 cells to CuONPs resulted in: (1) increased cell death in a time- and concentration-dependent manner, with a LC50 of 25.9 µg/ml after 24 h; (2) decreased antioxidant/detoxification enzymes activities: glutathione peroxidase (GPx), superoxide dismutase (SOD), glutathione S-tranferase (GST), and reduced glutathione (GSH) levels; (3) decreased gene expression of GPx and SOD; (4) reactive oxygen species (ROS) generation; (5) enhanced apoptosis; and (6) up-regulation of the pro-apoptotic genes Bax, and down-regulation of anti-apoptotic genes Bcl-2. Importantly, all these effects were significantly attenuated by co-incubation of hippocampal cells with 5 µM crocetin.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Carotenoids/pharmacology , Copper/toxicity , Hippocampus/drug effects , Metal Nanoparticles/toxicity , Animals , Cell Line , Cell Survival/drug effects , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Hippocampus/cytology , Light , Mice , Reactive Oxygen Species/metabolism , Scattering, Radiation , Superoxide Dismutase/metabolism , Vitamin A/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...