Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nanomedicine ; 17: 266-275, 2019 04.
Article in English | MEDLINE | ID: mdl-30794962

ABSTRACT

The complement system plays an essential role in both innate and adaptive immunity. The traditional understanding of this system comes from studies investigating complement proteins produced by the liver and present in plasma to "complement" the immune cell-mediated response to invading pathogens. Recently, it has been reported that immune cells including, but not limited to, T-cells and monocytes, express complement proteins. This complement is referred to as intracellular (IC) and implicated in the regulation of T-cell activation. The mechanisms and the structure-activity relationship between nanomaterials and IC, however, are currently unknown. Herein, we describe a structure-activity relationship study demonstrating that under in vitro conditions, only polymeric materials with cationic surfaces activate IC in T-cells. The effect also depends on particle size and occurs through a mechanism involving membrane damage, thereby IC on the cell surface serves as a self-opsonization marker in response to the nanoparticle-triggered danger affecting the cell integrity.


Subject(s)
Complement Activation , Lymphocyte Activation , Nanoparticles/adverse effects , Polymers/adverse effects , T-Lymphocytes/immunology , Cations/adverse effects , Cations/chemistry , Cells, Cultured , Complement Activation/drug effects , Humans , Jurkat Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Nanoparticles/chemistry , Polymers/chemistry , T-Lymphocytes/drug effects
2.
Methods Mol Biol ; 1682: 19-22, 2018.
Article in English | MEDLINE | ID: mdl-29039089

ABSTRACT

Bacterial contamination can confound the results of in vitro and in vivo preclinical tests. This protocol describes a procedure for detection of microbial contamination in nanotechnology-based formulations. Nanoparticle samples and controls are spread on the surface of agar and growth of bacterial colonies is monitored after 72 h of incubation. The intended purpose of this assay is to avoid introduction of microbial contamination into in vitro cell cultures and in vivo animal studies utilizing the test nanomaterial. This assay is not intended to certify the material as sterile.


Subject(s)
Bacteria/growth & development , Colony Count, Microbial/methods , Nanoparticles/microbiology , Agar/chemistry , Animals , Bacteria/isolation & purification , Equipment Contamination , Humans
3.
Methods Mol Biol ; 1682: 91-102, 2018.
Article in English | MEDLINE | ID: mdl-29039096

ABSTRACT

Hemolysis is damage to red blood cells (RBCs), which results in the release of the iron-containing protein hemoglobin into plasma. An in vitro assay was developed and described earlier for the analysis of nanoparticle hemolytic properties. Herein, we present a revised version of the original protocol. In this protocol, analyte nanoparticles and controls are incubated in blood. Undamaged RBCs are removed by centrifugation and hemoglobin, released by the damaged erythrocytes, is converted to cyanmethemoglobin by incubation with Drabkin's reagent. The amount of cyanmethemoglobin in the supernatant is measured by spectrophotometry. This measured absorbance is compared to a standard curve to determine the concentration of hemoglobin in the supernatant. The measured hemoglobin concentration is then compared to the total hemoglobin concentration to obtain the percentage of nanoparticle-induced hemolysis. The revision includes updated details about nanoparticle sample preparation, selection of nanoparticle concentration for the in vitro study, updated details about assay controls and case studies about nanoparticle interference with the in vitro hemolysis assay.


Subject(s)
Erythrocytes/pathology , Hemolysis , Nanoparticles/adverse effects , Blood Specimen Collection/methods , Centrifugation/methods , Hemoglobins/analysis , Humans , Indicators and Reagents , Methemoglobin/analogs & derivatives , Methemoglobin/analysis , Spectrophotometry/methods
4.
Methods Mol Biol ; 1682: 125-133, 2018.
Article in English | MEDLINE | ID: mdl-29039098

ABSTRACT

This chapter provides a protocol for analysis of nanoparticle effects on the function of phagocytic cells. The protocol relies on luminol chemiluminescence to detect zymosan uptake. Zymosan is an yeast particle which is typically eliminated by phagocytic cells via the complement receptor pathway. The luminol, co-internalized with zymosan, is processed inside the phagosome to generate a chemiluminescent signal. If a test nanoparticle affects the phagocytic function of the cell, the amount of phagocytosed zymosan and, proportionally, the level of generated chemiluminescent signal change. Comparing the zymosan uptake of untreated cells with that of cells exposed to a nanoparticle provides information about the nanoparticle's effects on the normal phagocytic function. This method has been described previously and is presented herein with several changes. The revised method includes details about nanoparticle concentration selection, updated experimental procedure, and examples of the method performance.


Subject(s)
Luminescent Measurements/methods , Phagocytes/cytology , Phagocytosis , Zymosan/analysis , HL-60 Cells , Humans , Luminol/analysis , Phagocytes/immunology , Zymosan/immunology
5.
Methods Mol Biol ; 1682: 103-124, 2018.
Article in English | MEDLINE | ID: mdl-29039097

ABSTRACT

Blood clotting is a complex process which involves both cellular and biochemical components. The key cellular players in the blood clotting process are thrombocytes or platelets. Other cells, including leukocytes and endothelial cells, contribute to clotting by expressing the so-called pro-coagulant activity (PCA) complex on their surface. The biochemical component of blood clotting is represented by the plasma coagulation cascade, which includes plasma proteins also known as coagulation factors. The coordinated interaction between platelets, leukocytes, endothelial cells, and plasma coagulation factors is necessary for maintaining hemostasis and for preventing excessive bleeding. Undesirable activation of all or some of these components may lead to pathological blood coagulation and life-threatening conditions such as consumptive coagulopathy or disseminated intravascular coagulation (DIC). In contrast, unintended inhibition of the coagulation pathways may lead to hemorrhage. Thrombogenicity is the property of a test material to induce blood coagulation by affecting one or more elements of the clotting process. Anticoagulant activity refers to the property of a test material to inhibit coagulation. The tendency to cause platelet aggregation, perturb plasma coagulation, and induce leukocyte PCA can serve as an in vitro measure of a nanomaterial's likelihood to be pro- or anticoagulant in vivo. This chapter describes three procedures for in vitro analyses of platelet aggregation, plasma coagulation time, and activation of leukocyte PCA. Platelet aggregation and plasma coagulation procedures have been described earlier. The revision here includes updated details about nanoparticle sample preparation, selection of nanoparticle concentration for the in vitro study, and updated details about assay controls. The chapter is expanded to describe a method for the leukocyte PCA analysis and case studies demonstrating the performance of these in vitro assays.


Subject(s)
Blood Coagulation Tests/methods , Blood Coagulation , Materials Testing/methods , Nanoparticles/adverse effects , Platelet Function Tests/methods , Blood Specimen Collection/methods , Humans , Leukocytes/cytology , Leukocytes/metabolism , Nanoparticles/metabolism , Platelet Aggregation
6.
Methods Mol Biol ; 1682: 149-160, 2018.
Article in English | MEDLINE | ID: mdl-29039100

ABSTRACT

The complement system is a group of proteins, which function in plasma to assist the innate immunity in rapid clearance of pathogens. The complement system also contributes to coordination of the adaptive immune response. Complement Activation Related Pseudo Allergy or CARPA is a life-threatening condition commonly reported with certain types of drugs and nanotechnology-based combination products. While CARPA symptoms are similar to that of anaphylaxis, the mechanism behind this pathology does not involve IgE and is mediated by the complement system. In vitro assays using serum or plasma derived from healthy donor volunteers correlate with the in vivo complement-mediated reactions, and therefore are helpful in understanding the propensity of a given drug formulation to cause CARPA in patients. In the first edition of this book, we have described an in vitro method for qualitative assessment of the complement activation by nanomaterials using western blotting. Herein, we present a similar method utilizing enzyme-linked immunoassay for quantitative analysis of the complement activation, and we compare the performance of this approach to that of the qualitative western blotting technique. The revised chapter also includes new details about nanoparticle sample preparation.


Subject(s)
Complement Activation , Immunoenzyme Techniques/methods , Nanoparticles/adverse effects , Blood Specimen Collection/methods , Blotting, Western/methods , Complement C3/analysis , Complement C3/immunology , Humans
7.
Methods Mol Biol ; 1682: 173-187, 2018.
Article in English | MEDLINE | ID: mdl-29039102

ABSTRACT

Cytokines, chemokines, and interferons are released by the immune cells in response to cellular stress, damage and/or pathogens, and are widely used as biomarkers of inflammation. Certain levels of cytokines are needed to stimulate an immune response in applications such as vaccines or immunotherapy where immune stimulation is desired. However, undesirable elevation of cytokine levels, as may occur in response to a drug or a device, may lead to severe side effects such as systemic inflammatory response syndrome or cytokine storm. Therefore, preclinical evaluation of a test material's propensity to cause cytokine secretion by healthy immune cells is an important parameter for establishing its safety profile. Herein, we describe in vitro methods for analysis of cytokines, chemokines, and type II interferon in whole blood cultures derived from healthy donor volunteers. First, whole blood is incubated with controls and tested nanomaterials for 24 h. Then, culture supernatants are analyzed by ELISA to detect IL-1ß, TNFα, IL-8, and IFNγ. The culture supernatants can also be analyzed for the presence of other biomarkers secreted by the immune cells. Such testing would require additional assays not covered in this chapter and/or optimization of the test procedure to include relevant positive controls and/or cell types.


Subject(s)
Cytokines/blood , Enzyme-Linked Immunosorbent Assay/methods , Inflammation/blood , Inflammation/etiology , Nanoparticles/adverse effects , Cytokines/immunology , Humans , Inflammation/immunology , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-1beta/blood , Interleukin-1beta/immunology , Interleukin-8/blood , Interleukin-8/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
8.
Toxicol Appl Pharmacol ; 299: 70-7, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26773813

ABSTRACT

Nanoparticle immunogenicity and antigenicity have been under investigation for many years. During the past decade, significant progress has been made in understanding what makes a nanoparticle immunogenic, how immune cells respond to nanoparticles, what consequences of nanoparticle-specific antibody formation exist and how they challenge the application of nanoparticles for drug delivery. Moreover, it has been recognized that accidental contamination of therapeutic protein formulations with nanosized particulate materials may contribute to the immunogenicity of this type of biotechnology products. While the immunological properties of engineered nanomaterials and their application as vaccine carriers and adjuvants have been given substantial consideration in the current literature, little attention has been paid to nanoparticle immuno- and antigenicity. To fill in this gap, we herein provide an overview of this subject to highlight the current state of the field, review past and present research, and discuss future research directions.


Subject(s)
Antigenic Modulation/immunology , Comprehension , Immunity, Cellular/immunology , Immunogenetic Phenomena/immunology , Nanostructures/toxicity , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/toxicity , Animals , Antigenic Modulation/drug effects , Drug Carriers/administration & dosage , Drug Carriers/toxicity , Drug Delivery Systems/adverse effects , Drug Delivery Systems/methods , Forecasting , Humans , Immunity, Cellular/drug effects , Immunogenetic Phenomena/drug effects , Nanostructures/administration & dosage
9.
Nanomedicine ; 11(8): 1925-38, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26282378

ABSTRACT

Understanding the ability of cytotoxic oncology drugs, and their carriers and formulation excipients, to induce pro-inflammatory responses is important for establishing safe and efficacious formulations. Literature data about cytokine response induction by the traditional formulation of paclitaxel, Taxol®, are controversial, and no data are available about the pro-inflammatory profile of the nano-albumin formulation of this drug, Abraxane®. Herein, we demonstrate and explain the difference in the cytokine induction profile between Taxol® and Abraxane®, and describe a novel mechanism of cytokine induction by a nanosized excipient, Cremophor EL, which is not unique to Taxol® and is commonly used in the pharmaceutical industry for delivery of a wide variety of small molecular drugs. FROM THE CLINICAL EDITOR: Advances in nanotechnology have enabled the production of many nano-formulation drugs. The cellular response to drugs has been reported to be different between traditional and nano-formulations. In this article, the authors investigated and compared cytokine response induction profiles between Taxol® and Abraxane®. The findings here provided further understanding to create drugs with better safety profiles.


Subject(s)
Albumin-Bound Paclitaxel/adverse effects , Antineoplastic Agents/adverse effects , Glycerol/analogs & derivatives , Oxidative Stress/drug effects , Paclitaxel/adverse effects , Pharmaceutical Vehicles/adverse effects , Polyethylene Glycols/adverse effects , Animals , Cell Line , Glycerol/adverse effects , Humans , Interleukin-8/blood , Interleukin-8/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Mice
10.
Nanomedicine (Lond) ; 9(9): 1311-26, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24279459

ABSTRACT

AIM: Disseminated intravascular coagulation is an increasing concern for certain types of engineered nanomaterials. Recent studies have shed some light on the nanoparticle physicochemical properties contributing to this toxicity; however, the mechanisms are poorly understood. Leukocyte procoagulant activity (PCA) is a key factor contributing to the initiation of this toxicity. We have previously reported on the exaggeration of endotoxin-induced PCA by cationic dendrimers. Herein, we report an effort to discern the mechanism. MATERIALS & METHODS: Poly(amidoamine) dendrimers with various sizes and surface functionalities were studied in vitro by the recalcification test, flow cytometry and other relevant assays. RESULTS & CONCLUSION: Cationic dendrimers exaggerated endotoxin-induced PCA, but their anionic or neutral counterparts did not; the cationic charge prompts this phenomenon, but different cationic surface chemistries do not influence it. Cationic dendrimers and endotoxin differentially affect the PCA complex. The inhibition of phosphoinositol 3 kinase by dendrimers contributes to the exaggeration of the endotoxin-induced PCA.


Subject(s)
Blood Coagulation Factors/biosynthesis , Endotoxins/toxicity , Nanoparticles/chemistry , Nanoparticles/toxicity , Phosphoinositide-3 Kinase Inhibitors , Cations/chemistry , Cations/toxicity , Dendrimers/chemistry , Dendrimers/toxicity , Disseminated Intravascular Coagulation/etiology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Humans , In Vitro Techniques , Leukocytes/drug effects , Leukocytes/metabolism , Lipopolysaccharides/toxicity , Polyamines/chemistry , Polyamines/toxicity
11.
Nanomedicine (Lond) ; 8(6): 969-81, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23730696

ABSTRACT

Nanoparticle interactions with the blood coagulation system can be beneficial or adverse depending on the intended use of a nanomaterial. Nanoparticles can be engineered to be procoagulant or to carry coagulation-initiating factors to treat certain disorders. Likewise, they can be designed to be anticoagulant or to carry anticoagulant drugs to intervene in other pathological conditions in which coagulation is a concern. An overview of the coagulation system was given and a discussion of a desirable interface between this system and engineered nanomaterials was assessed in part I, which was published in the May 2013 issue of Nanomedicine. Unwanted pro- and anti-coagulant properties of nanoparticles represent significant concerns in the field of nanomedicine, and often hamper the development and transition into the clinic of many promising engineered nanocarriers. This part will focus on the undesirable effects of engineered nanomaterials on the blood coagulation system. We will discuss the relationship between the physicochemical properties of nanoparticles (e.g., size, charge and hydrophobicity) that determine their negative effects on the blood coagulation system in order to understand how manipulation of these properties can help to overcome unwanted side effects.


Subject(s)
Blood Coagulation/drug effects , Nanoparticles/adverse effects , Nanoparticles/chemistry , Animals , Humans , Nanomedicine , Nanoparticles/metabolism
12.
Nanomedicine (Lond) ; 8(5): 773-84, 2013 May.
Article in English | MEDLINE | ID: mdl-23656264

ABSTRACT

Nanotechnology is proven to provide certain benefits in drug delivery by improving solubility, increasing uptake to target sites and changing pharmacokinetics profiles of traditional drugs. Since properties of many materials change tremendously at the nanoscale levels, nanotechnology is also being explored in various industrial applications. As such, nanoparticles are rapidly entering various areas of industry, biology and medicine. The benefits of using nanotechnology for industrial and biomedical applications are often tempered by concerns about the safety of these new materials. One such area of concern includes their effect on the immune system. While nanoparticle interactions with various constituents of the immune system have been reviewed before, little attention was given to nanoparticle effects on the blood coagulation system. Nanoparticle interface with the blood coagulation system may lead to either benefits to the host or adverse reactions. This article reviews recent advances in our understanding of nanoparticle interactions with plasma coagulation factors, platelets, endothelial cells and leukocytes. Part I is focused on desirable interactions between nanoparticles and the coagulation system, and discusses benefits of using nanotechnology to intervene in coagulation disorders. Undesirable interactions posing safety concerns are covered in part II, which will be published in the June issue of Nanomedicine.


Subject(s)
Blood Coagulation , Nanomedicine/methods , Nanoparticles/metabolism , Animals , Blood Coagulation/drug effects , Blood Coagulation Factors/metabolism , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Drug Delivery Systems/methods , Humans , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Nanotechnology/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...