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










Publication year range
1.
Platelets ; 31(3): 399-402, 2020.
Article in English | MEDLINE | ID: mdl-31146647

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder in which autoantibodies and/or autoreactive T cells destroy platelets and megakaryocytes in the spleen and bone marrow, respectively. Thrombopoietin receptor agonists (TPO-RA e.g. Romiplostim and Eltrombopag) have made a substantial contribution to the treatment of patients with ITP, which are refractory to first-line treatments and approximately 30% demonstrate sustained elevated platelet counts after drug tapering. How TPO-RA induce these sustained responses is not known. We analyzed the efficacy of a murine TPO-RA in a well-established murine model of active ITP. Treatment with TPO-RA (10 ug/kg, based on pilot dose escalation experiments) significantly raised the platelet counts in ITP-mice. Immunomodulation was assessed by measuring serum IgG anti-platelet antibody levels; TPO-RA-treated mice had significantly reduced IgG anti-platelet antibodies despite the increasing platelet counts. These results suggest that TPO-RA is not only an efficacious therapy but also reduces anti-platelet humoral immunity in ITP.


Subject(s)
Autoantibodies/immunology , Blood Platelets/immunology , Blood Platelets/metabolism , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/etiology , Purpura, Thrombocytopenic, Idiopathic/metabolism , Receptors, Thrombopoietin/agonists , Animals , Autoimmunity , Biopsy , Blood Platelets/drug effects , Disease Models, Animal , Disease Susceptibility , Humans , Immunomodulation , Mice , Mice, Knockout , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/pathology
2.
Blood Adv ; 2(13): 1651-1663, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29991496

ABSTRACT

Transfusion-related acute lung injury (TRALI) is a syndrome of respiratory distress upon blood transfusion and is the leading cause of transfusion-related fatalities. Whether the gut microbiota plays any role in the development of TRALI is currently unknown. We observed that untreated barrier-free (BF) mice suffered from severe antibody-mediated acute lung injury, whereas the more sterile housed specific pathogen-free (SPF) mice and gut flora-depleted BF mice were both protected from lung injury. The prevention of TRALI in the SPF mice and gut flora-depleted BF mice was associated with decreased plasma macrophage inflammatory protein-2 levels as well as decreased pulmonary neutrophil accumulation. DNA sequencing of amplicons of the 16S ribosomal RNA gene revealed a varying gastrointestinal bacterial composition between BF and SPF mice. BF fecal matter transferred into SPF mice significantly restored TRALI susceptibility in SPF mice. These data reveal a link between the gut flora composition and the development of antibody-mediated TRALI in mice. Assessment of gut microbial composition may help in TRALI risk assessment before transfusion.


Subject(s)
Chemokine CXCL2/blood , Gastrointestinal Microbiome , Lung/metabolism , Neutrophils/metabolism , Transfusion-Related Acute Lung Injury/microbiology , Animals , Lung/pathology , Mice , Neutrophils/pathology , Transfusion-Related Acute Lung Injury/pathology
4.
Blood ; 129(18): 2557-2569, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28202460

ABSTRACT

Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-related fatalities and is characterized by acute respiratory distress following blood transfusion. Donor antibodies are frequently involved; however, the pathogenesis and protective mechanisms in the recipient are poorly understood, and specific therapies are lacking. Using newly developed murine TRALI models based on injection of anti-major histocompatibility complex class I antibodies, we found CD4+CD25+FoxP3+ T regulatory cells (Tregs) and CD11c+ dendritic cells (DCs) to be critical effectors that protect against TRALI. Treg or DC depletion in vivo resulted in aggravated antibody-mediated acute lung injury within 90 minutes with 60% mortality upon DC depletion. In addition, resistance to antibody-mediated TRALI was associated with increased interleukin-10 (IL-10) levels, and IL-10 levels were found to be decreased in mice suffering from TRALI. Importantly, IL-10 injection completely prevented and rescued the development of TRALI in mice and may prove to be a promising new therapeutic approach for alleviating lung injury in this serious complication of transfusion.


Subject(s)
Acute Lung Injury , Dendritic Cells/immunology , Interleukin-10 , T-Lymphocytes, Regulatory/immunology , Transfusion Reaction , Acute Lung Injury/etiology , Acute Lung Injury/immunology , Acute Lung Injury/prevention & control , Animals , Antibodies/immunology , Dendritic Cells/pathology , Interleukin-10/immunology , Interleukin-10/pharmacology , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes, Regulatory/pathology
5.
Blood Adv ; 1(20): 1773-1785, 2017 Sep 12.
Article in English | MEDLINE | ID: mdl-29296823

ABSTRACT

Megakaryocytes (MKs) are bone marrow-derived cells that are primarily responsible for generating platelets for the maintenance of hemostasis. Although MK can variably express major histocompatibility complex (MHC) class I and II molecules during their differentiation, little is known whether they can elicit nonhemostatic immune functions such as T-cell activation. Here, we demonstrate that mature CD34- MHC class II- CD41+ MKs can endocytose exogenous ovalbumin (OVA) and proteolytically generate its immunogenic peptide ligand, which is crosspresented on their surface in association with MHC class I molecules. This crosspresentation triggered in vitro and in vivo OVA-specific CD8+ T-cell activation and proliferation. In addition, the OVA-MHC class I complexes were transferred from MK to pro-platelets upon thrombopoiesis in vitro. MK could also present endogenous MK-associated (CD61) peptides to activate CD61-specific CD8+ T cells and mediate immune thrombocytopenia in vivo. These results suggest that, in addition to their hemostatic role, mature MKs can significantly affect antigen-specific CD8+ T-cell responses via antigen presentation and are able to spread this immunogenic information through platelets.

6.
Am J Respir Crit Care Med ; 195(7): 889-905, 2017 04 01.
Article in English | MEDLINE | ID: mdl-27735193

ABSTRACT

RATIONALE: Effective and rapid bacterial clearance is a fundamental determinant of outcomes in sepsis. DJ-1 is a well-established reactive oxygen species (ROS) scavenger. OBJECTIVES: Because cellular ROS status is pivotal to inflammation and bacterial killing, we determined the role of DJ-1 in bacterial sepsis. METHODS: We used cell and murine models with gain- and loss-of-function experiments, plasma, and cells from patients with sepsis. MEASUREMENTS AND MAIN RESULTS: Stimulation of bone marrow-derived macrophages (BMMs) with endotoxin resulted in increased DJ-1 mRNA and protein expression. Cellular and mitochondrial ROS was increased in DJ-1-deficient (-/-) BMMs compared with wild-type. In a clinically relevant model of polymicrobial sepsis (cecal ligation and puncture), DJ-1-/- mice had improved survival and bacterial clearance. DJ-1-/- macrophages exhibited enhanced phagocytosis and bactericidal activity in vitro, and adoptive transfer of DJ-1-/- bone marrow-derived mononuclear cells rescued wild-type mice from cecal ligation and puncture-induced mortality. In stimulated BMMs, DJ-1 inhibited ROS production by binding to p47phox, a critical component of the NADPH oxidase complex, disrupting the complex and facilitating Nox2 (gp91phox) ubiquitination and degradation. Knocking down DJ-1 (siRNA) in THP-1 (human monocytic cell line) and polymorphonuclear cells from patients with sepsis enhanced bacterial killing and respiratory burst. DJ-1 protein levels were elevated in plasma from patients with sepsis. Higher levels of circulating DJ-1 were associated with increased organ failure and death. CONCLUSIONS: These novel findings reveal DJ-1 impairs optimal ROS production for bacterial killing with important implications for host survival in sepsis.


Subject(s)
Protein Deglycase DJ-1/blood , Sepsis/blood , Animals , Disease Models, Animal , Humans , Male , Mice , Reactive Oxygen Species/blood
7.
Blood ; 127(6): 735-8, 2016 Feb 11.
Article in English | MEDLINE | ID: mdl-26556550

ABSTRACT

Immune thrombocytopenia (ITP) is an autoimmune bleeding disorder with a complex pathogenesis, which includes both antibody- and T-cell-mediated effector mechanisms. Rituximab (an anti-human CD20 monoclonal antibody [mAb]) is one of the treatments for ITP and is known to deplete B cells but may also work by affecting the T-cell compartments. Here, we investigated the outcome of B-cell depletion (Bdep) therapy on CD8(+) T-cell-mediated ITP using a murine model. CD61 knockout (KO) mice were immunized with CD61(+) platelets, and T-cell-mediated ITP was initiated by transfer of their splenocytes into severe combined immunodeficiency (SCID) mice. The CD61 KO mice were administrated an anti-mouse CD20 mAb either before or after CD61(+) platelet immunization. This resulted in efficient Bdep in vivo, accompanied by significant increases in splenic and lymph node CD4(+) and CD8(+) T cells and proportional increases of FOXP3(+) in CD4(+)and CD8(+) T cells. Moreover, Bdep therapy resulted in significantly decreased splenic CD8(+) T-cell proliferation in vitro that could be rescued by interleukin-2. This correlated with normalization of in vivo platelet counts in the transferred SCID mice suggesting that anti-CD20 therapy significantly reduces the ability of CD8(+) T cells to activate and mediate ITP.


Subject(s)
Antigens, CD20/metabolism , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Depletion/methods , Purpura, Thrombocytopenic, Idiopathic/therapy , Animals , B-Lymphocytes/pathology , Integrin beta3/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/pathology
8.
Blood ; 123(3): 422-7, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24258817

ABSTRACT

Platelet transfusions are life-saving treatments for many patients with thrombocytopenia; however, their use is generally discouraged in the autoimmune disorder known as immune thrombocytopenia (ITP). We examined whether allogeneic platelet major histocompatibility complex (MHC) class I transfusions affected antiplatelet CD61-induced ITP. BALB/c CD61 knockout mice (CD61(-)/H-2(d)) were immunized against platelets from wild-type syngeneic BALB/c (CD61(+)/H-2(d)), allogeneic C57BL/6 (CD61(+)/H-2(b)), or C57BL/6 CD61 KO (CD61(-)/H-2(b)) mice, and their splenocytes were transferred into severe combined immunodeficient (SCID) mice to induce ITP. When nondepleted splenocytes were transferred to induce antibody-mediated ITP, both CD61(+) platelet immunizations generated immunity that caused thrombocytopenia independently of allogeneic MHC molecules. In contrast, when B-cell-depleted splenocytes were transferred to induce T-cell-mediated ITP, transfer of allogeneic MHC-immunized splenocytes completely prevented CD61-induced ITP development. In addition, allogeneic platelet transfusions into SCID mice with established CD61-induced ITP rescued the thrombocytopenia. Compared with thrombocytopenic mice, bone marrow histology in the rescued mice showed normalized megakaryocyte morphology, and in vitro CD61-specific T-cell cytotoxicity was significantly suppressed. These results indicate that antibody-mediated ITP is resistant to allogeneic platelet transfusions, while the T-cell-mediated form of the disease is susceptible, suggesting that transfusion therapy may be beneficial in antibody-negative ITP.


Subject(s)
Platelet Transfusion/methods , T-Lymphocytes/immunology , Thrombocytopenia/genetics , Thrombocytopenia/prevention & control , Animals , Bone Marrow Cells/cytology , Disease Models, Animal , Female , Histocompatibility Antigens Class I/metabolism , Immunoglobulin G/immunology , Integrin beta3/metabolism , Megakaryocytes/cytology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Spleen/cytology , Thrombocytopenia/immunology , Time Factors
9.
Blood ; 120(10): 2127-32, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22760780

ABSTRACT

Immune thrombocytopenia (ITP) is a bleeding disorder in which antibodies and/or T cells lead to enhanced peripheral platelet destruction and reduced bone marrow platelet production. Several reports have observed that ITP is associated with a peripheral deficiency of tolerance-inducing CD4+CD25+FoxP3+ T regulatory cells (Tregs). Using a murine model of ITP, we analyzed Tregs in the spleen and thymus. CD61 knockout mice were immunized against wild-type (CD61+) platelets, and their splenocytes were transferred into severe combined immunodeficient (SCID) mice. Compared with SCID mice receiving naive splenocytes, within 2 weeks after transfer, the ITP SCID mice became thrombocytopenic (< 200 × 10(9) platelets/L) and had increased serum anti-CD61 antibodies. The quantity of thymic Tregs by 2 weeks after transfer was significantly elevated, whereas Tregs in the spleens were significantly reduced. Treatment of the ITP mice with 2 g/kg intravenous immunoglobulin raised the platelet counts, reduced antibody production, and normalized the thymic and splenic Treg populations. Compared with thymocytes from ITP mice treated with intravenous immunoglobulin, thymocytes from untreated ITP mice delayed the onset of ITP when administered before engraftment with immune splenocytes. These results suggest that ITP in mice is associated with a peripheral Treg deficiency because of thymic retention and therapy normalizes the Tregs.


Subject(s)
Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Thrombocytopenia/immunology , Thymus Gland/immunology , Animals , Antibodies/administration & dosage , Antibodies/blood , Antibodies/immunology , Blood Platelets/immunology , CD4 Lymphocyte Count , Disease Models, Animal , Female , Forkhead Transcription Factors/immunology , Immune System Diseases , Immunoglobulins, Intravenous/therapeutic use , Integrin beta3/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Transfusion , Mice , Mice, Knockout , Mice, SCID , Organ Specificity , Spleen/drug effects , Spleen/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , Thrombocytopenia/drug therapy , Thrombocytopenia/pathology , Thymus Gland/drug effects , Thymus Gland/pathology
10.
Blood ; 116(16): 3073-9, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20616220

ABSTRACT

Transfusion-related acute lung injury (TRALI) is a serious complication of transfusion and has been ranked as one of the leading causes of transfusion-related fatalities. Nonetheless, many details of the immunopathogenesis of TRALI, particularly with respect to recipient factors are unknown. We used a murine model of antibody-mediated TRALI in an attempt to understand the role that recipient lymphocytes might play in TRALI reactions. Intravenous injection of an IgG2a antimurine major histocompatibility complex class I antibody (34-1-2s) into BALB/c mice induced moderate hypothermia and pulmonary granulocyte accumulation but no pulmonary edema nor mortality. In contrast, 34-1-2s injections into mice with severe combined immunodeficiency caused severe hypothermia, severe pulmonary edema, and approximately 40% mortality indicating a critical role for T and B lymphocytes in suppressing TRALI reactions. Adoptive transfer of purified CD8(+) T lymphocytes or CD4(+) T cells but not CD19(+) B cells into the severe combined immunodeficiency mice alleviated the antibody-induced hypothermia, lung damage, and mortality, suggesting that T lymphocytes were responsible for the protective effect. Taken together, these results suggest that recipient T lymphocytes play a significant role in suppressing antibody-mediated TRALI reactions. They identify a potentially new recipient mechanism that controls the severity of TRALI reactions.


Subject(s)
Acute Lung Injury/etiology , Acute Lung Injury/immunology , Antibodies/adverse effects , T-Lymphocytes/immunology , Transfusion Reaction , Animals , Antibodies/immunology , Chemokine CXCL2/blood , Chemokine CXCL2/immunology , Histocompatibility Antigens Class I/immunology , Hypothermia/etiology , Hypothermia/immunology , Immunoglobulin G/adverse effects , Immunoglobulin G/immunology , Lung/cytology , Lung/pathology , Lymphocyte Transfusion , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Neutrophils/immunology
11.
Blood ; 115(6): 1247-53, 2010 Feb 11.
Article in English | MEDLINE | ID: mdl-20007808

ABSTRACT

Immune thrombocytopenia (ITP) is a bleeding disorder characterized by antibody-opsonized platelets being prematurely destroyed in the spleen, although some patients with ITP may have a cell-mediated form of thrombocytopenia. Although several animal models of ITP have been developed, few mimic primary chronic ITP nor have any shown cell-mediated platelet destruction. To create this type of model, splenocytes from CD61 knockout mice immunized against CD61(+) platelets were transferred into severe combined immunodeficient (SCID) (CD61(+)) mouse recipients, and their platelet counts and phenotypes were observed. As few as 5 x 10(4) splenocytes induced a significant thrombocytopenia and bleeding mortality (80%) in recipients within 3 weeks after transfer. Depletion of lymphocyte subsets before transfer showed that the splenocyte's ability to induce thrombocytopenia and bleeding completely depended on CD4(+) T helper cells and that both CD19(+) B cell (antibody)- and CD8(+) T cell (cell)-mediated effector mechanisms were responsible. Treatment of the SCID mouse recipients with intravenous gamma-globulins raised platelet counts and completely prevented bleeding mortality induced by antibody-mediated effector mechanisms but did not affect cell-mediated disease. This novel model not only shows both antibody- and cell-mediated ITP and bleeding but also suggests that these 2 effector mechanisms have a differential response to therapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Immunoglobulins, Intravenous/administration & dosage , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/therapy , Animals , Antigens, CD19/immunology , Blood Platelets/immunology , Female , Flow Cytometry , Integrin beta3/immunology , Lymphocyte Depletion , Megakaryocytes/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Platelet Count , Purpura, Thrombocytopenic, Idiopathic/pathology , Spleen/physiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology
12.
Transfusion ; 48(9): 1917-24, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18564400

ABSTRACT

BACKGROUND: Studies have demonstrated that immunity against platelet (PLT) transfusions is dependent on recipient antigen-presenting cells (APCs) and their ability to produce nitric oxide (NO). To further analyze this, we focused on NO's major metabolite peroxynitrite (ONOO(-)) and its ability to affect PLT immunity. STUDY DESIGN AND METHODS: To address how NO and its major metabolite may mediate PLT immunity, GP91(PHOX) knockout (KO) mice that lack the ability to produce the ONOO(-) were transfused weekly with allogeneic BALB/c PLTs, and donor antibody development was analyzed. RESULTS: Compared with controls, GP91(PHOX) KO mice developed significantly (p < 0.0001) higher-titered immunoglobulin G (IgG) donor antibodies by two transfusions, and this immune response could be inhibited by treating the recipient mice with aminoguanidine, a relatively selective inhibitor of inducible nitric oxide synthase. In vitro nitration of PLTs did not alter PLT antibody binding but significantly inhibited the transfused PLT's ability to stimulate IgG immunity in either wild-type or KO mice. The lack of nitrated PLT immunity correlated with an inability of APCs to mediate phagocytosis of nitrated PLTs. The lack of nitrated PLT immunity could only be restored when normal PLTs were mixed with the nitrated PLTs and transfused. CONCLUSION: The results identify a dual role for NO metabolism within APCs that significantly modulates PLT immunity; nitration of PLT antigens leads to lack of immunity due to an inability of APCs to move PLT antigens intracellularly whereas there exists an NO-dependent pathway that stimulates anti-PLT immunity.


Subject(s)
Antigen-Presenting Cells/metabolism , Antigens, Human Platelet/metabolism , Nitrates/metabolism , Signal Transduction/immunology , Animals , Antibodies, Anti-Idiotypic/immunology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, Human Platelet/immunology , Guanidines/pharmacology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidase 2 , NADPH Oxidases/genetics , Nitric Oxide Synthase Type II/antagonists & inhibitors , Platelet Transfusion/adverse effects , Signal Transduction/drug effects
13.
Transfusion ; 48(9): 1778-86, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18522705

ABSTRACT

BACKGROUND: Transfusion-related immunomodulation (TRIM) has been correlated with the presence of white cells (WBCs) in blood transfusions, but the role of components such as platelets (PLTs) in mediating TRIM has not been extensively examined. We designed a murine PLT transfusion model to study whether leukoreduced PLTs mediate TRIM effects. STUDY DESIGN AND METHODS: CBA recipient mice were administered four weekly transfusions of either fresh (4 hr) or aged (24 and 72 hr) donor leukoreduced PLTs from allogeneic BALB/c mice and then transplanted with skin grafts from donor-matched mice. TRIM was measured by comparing the times to graft rejection and these were correlated with immunoglobulin G (IgG) antibody development measured by flow cytometry. RESULTS: Compared with nontransfused control recipients, four transfusions of fresh, extremely leukoreduced (<0.05 WBCs/mL), allogeneic PLTs significantly (p < 0.002) reduced the recipient's ability to reject donor-matched skin grafts (survival >49 days compared with <14 days in nontransfused controls) despite the presence of high-titered serum IgG donor antibodies. In contrast, however, aged PLTs or fresh PLTs devoid of MHC Class I molecules were unable to affect skin graft survival nor stimulate antibody production. The PLT age-related inability to induce TRIM was shown to be due to loss of PLT-associated MHC Class I molecules; soluble supernatant MHC molecules that were transfused were unable to induce TRIM. CONCLUSION: These results suggest that fresh PLTs can induce TRIM independently of WBCs due to their MHC antigen expression whereas aging results in loss of MHC and ability to mediate TRIM. The findings support the concept that either active MHC removal from fresh PLTs or passive removal by, for example, storage, may reduce any deleterious effects of TRIM in transfusion recipients.


Subject(s)
Blood Platelets/immunology , Histocompatibility Antigens Class I/immunology , Leukocytes/immunology , Animals , Leukocytes/cytology , Mice , Models, Animal , Platelet Transfusion/adverse effects
14.
J Oral Maxillofac Surg ; 66(4): 632-8, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18355586

ABSTRACT

PURPOSE: Autologous platelet gels can be prepared using the patient's own platelet-rich plasma and thrombin produced by the Thrombin Processing Device (Thermogenesis Corp, Rancho Cordova, CA). As the Thrombin Processing Device thrombin contains a residual amount of ethanol, the purpose of this study was to investigate the effect of the Thrombin Processing Device thrombin on growth factor release from platelet gels, and the effect on cell viability and cell proliferation. MATERIALS AND METHODS: Platelet gels were prepared using Thrombin Processing Device-produced human thrombin at platelet-rich plasma to thrombin ratios of 3.3 to 1 and 7 to 1. Commercially available bovine thrombin was used as control. The content of the growth factors, platelet-derived growth factor beta polypeptide, and transforming growth factor beta, were assessed in both the clot and supernatant. The influence of different concentrations of ethanol on cell viability was assessed by flow cytometry and cell proliferation was assessed by (3)H-thymidine incorporation. RESULTS: Using a ratio of 3.3 to 1, the supernatant of the platelet gel produced with Thrombin Processing Device thrombin had a lower growth factor content compared with bovine thrombin but was similar when prepared using a ratio of platelet-rich plasma to thrombin of 7 to 1. Supernatants from the platelet gels did not affect the viability of human macrophage line cells or a fibroblast cell line. When the different platelet gels or their supernatants were tested for their ability to stimulate cell proliferation, similar rates of proliferation were observed. CONCLUSIONS: These data suggest that residual ethanol in the Thrombin Processing Device-produced thrombin does not affect any of the tested parameters and has similar characteristics as commercially available bovine thrombin.


Subject(s)
Platelet Activation/drug effects , Platelet-Rich Plasma/drug effects , Thrombin/chemical synthesis , Thrombin/toxicity , Animals , Cattle , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Ethanol/toxicity , Fibroblasts/drug effects , Gels , Humans , Macrophages/drug effects , Platelet-Rich Plasma/metabolism , Proto-Oncogene Proteins c-sis/biosynthesis , Transforming Growth Factor beta/biosynthesis
15.
Transfusion ; 47(11): 2161-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17958546

ABSTRACT

BACKGROUND: Anti-D treatment is effective in increasing platelet (PLT) counts in patients with autoimmune thrombocytopenic purpura (AITP); however, the exact mechanism of action is unknown. Previous results have suggested that anti-D-coated red blood cells (RBCs) affect reticuloendothelial system phagocytosis by stimulating agents (e.g., reactive oxygen species) that alter signaling pathways within the phagocyte. To address this, a flow cytometric assay was used to compare the kinetics and signaling pathways responsible for opsonized PLT and RBC phagocytosis. STUDY DESIGN AND METHODS: Human RBCs or PLTs were labeled with the fluorescent dye CM-Green, opsonized with Rh immune globulin or anti-MHC, respectively, and incubated with THP-1 monocytes with or without signal transduction inhibitors and intracellular fluorescence was analyzed. RESULTS: Compared with opsonized PLTs, phagocytosis of opsonized RBCs was significantly slower (p<0.0001) and, within 2 hours, induced a state of phagocytic refractoriness; resting the mononuclear cells (MNCs) for up to 24 hours did not rescue their ability to further mediate PLT phagocytosis. Inhibitors of phosphatidylinositol 3-kinase (wortmannin, LY294002, myricetin, and quercetin), protein kinase C (staurosporine), and Syk kinase (piceatannol) inhibited both opsonized RBC and opsonized PLT phagocytosis. In contrast, opsonized RBC phagocytosis was significantly (p<0.0001) enhanced by the tyrosine phosphatase inhibitor phenyl arsine oxide, whereas PLT phagocytosis was significantly reduced (p<0.0001). Of interest, phosphatase inhibition during opsonized RBC phagocytosis induced a longer (48 hr) phagocytic refractoriness period in the MNCs. CONCLUSION: These results suggest that the early kinetics and signaling events related to phosphatase activity regulate how mononuclear phagocytes engulf opsonized RBCs and induce phagocytic refractoriness for further PLT phagocytosis.


Subject(s)
Blood Platelets/immunology , Erythrocytes/immunology , Leukocytes, Mononuclear/enzymology , Phagocytosis , Phosphoric Monoester Hydrolases/immunology , Flow Cytometry , Humans , Kinetics , Leukocytes, Mononuclear/immunology , Opsonin Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Signal Transduction
16.
Blood ; 110(4): 1359-61, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17456719

ABSTRACT

Rh immune globulin (WinRho SDF; Cangene, Mississauga, ON, Canada) is an effective treatment for autoimmune thrombocytopenic purpura; however, maintaining a sustained supply for its use in autoimmune thrombocytopenic purpura and its primary indication, hemolytic disease of the newborn, makes the development of alternative reagents desirable. We compared Rh immune globulin and 6 human monoclonal anti-D antibodies (MoAnti-D) with differing isotypes and specificities for their ability to opsonize erythrocytes and inhibit platelet phagocytosis in an in vitro assay. Results demonstrated that opsonization of erythrocytes with Rh immune globulin significantly (P < .001) reduced phagocytosis of fluorescently labeled opsonized platelets in an Fc-dependent manner. Of the MoAnti-D that shared specificity but differed in isotype, only IgG3 antibodies could significantly (P < .001) inhibit platelet phagocytosis. In contrast, 2 MoAnti-D shared isotypes and differed in specificity; however, only one could significantly (P < .001) inhibit platelet phagocytosis. The results suggest that MoAnti-D epitope specificity and isotypes are critical requirements for optimal inhibition of opsonized platelet phagocytosis.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Human Platelet/immunology , Blood Platelets/metabolism , Epitopes/immunology , Opsonin Proteins , Phagocytosis , Purpura, Thrombocytopenic, Idiopathic/immunology , Rho(D) Immune Globulin/immunology , Antibodies, Monoclonal/metabolism , Antigens, Human Platelet/metabolism , Autoantibodies/immunology , Autoantibodies/metabolism , Blood Platelets/immunology , Humans , Immunoglobulin Isotypes , Purpura, Thrombocytopenic, Idiopathic/metabolism , Rh Isoimmunization
17.
Blood ; 109(11): 4803-5, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17299089

ABSTRACT

Platelets express Toll-like receptor 4 (TLR4), and this has been shown to be responsible for the thrombocytopenia induced by lipopolysaccharide (LPS) administration in vivo. We studied the role of LPS in mediating platelet phagocytosis by THP-1 cells in vitro by flow cytometry. Opsonization of platelets with an IgG monoclonal (W6/32) antibody or with IgG autoantibody-positive sera from patients with autoimmune thrombocytopenia (AITP) significantly enhanced platelet phagocytosis (P < .001). In contrast, platelet phagocytosis did not occur if platelets were bound with only LPS. If, however, the LPS-bound platelets were also opsonized with either W6/32 or autoantibody-positive sera with titers greater than 4, there was a significant and synergistic increase in Fc-dependent platelet phagocytosis (P < .001, P = .003, P = .048, and P = .047). These results suggest that, in the presence of antiplatelet antibodies, bacterial products can significantly alter platelet phagocytosis, and this may have relevance to how Gram-negative infections enhance platelet destruction in some patients with AITP.


Subject(s)
Blood Platelets/metabolism , Immunoglobulin G/chemistry , Lipopolysaccharides/metabolism , Opsonin Proteins/chemistry , Phagocytosis , Receptors, Fc/metabolism , Antibodies, Monoclonal/chemistry , Antigens, Human Platelet/chemistry , Autoantibodies/chemistry , Cell Line , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Platelet Aggregation , Toll-Like Receptor 4/metabolism
18.
Blood ; 107(2): 637-41, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16179373

ABSTRACT

Toll-like receptors (TLRs) play a critical role in stimulating innate immunity by recognizing pathogen-associated molecular patterns (PAMPs) on invading microorganisms. Platelets also play a role in innate immunity, and we studied whether they express TLR. Results show that human and murine platelets variably expressed TLR2, TLR4, and TLR9 by flow cytometry and Western blotting. TLR4 expression was confirmed by demonstrating murine platelet binding to lipopolysaccharide (LPS). Thrombin activation of the platelets significantly enhanced the expression of TLR9, suggesting that at least some TLRs may derive from intracellular compartments. When LPS was administered to LPS-sensitive C3H/HeN and LPS-resistant C3H/HeJ mice, functional TLR4 expression in vivo was shown to be responsible for LPS-induced thrombocytopenia. However, when the C3H/HeN mice were first rendered thrombocytopenic by an antiplatelet antibody and then administered LPS, a significant reduction occurred in their ability to produce TNF-alpha. The decreased cytokine production in the thrombocytopenic mice was restored with platelet transfusion. These results suggest that platelets express various TLRs and that the functional significance of one of these, TLR4, appears to be a role in the modulation of LPS-induced thrombocytopenia and TNF-alpha production. This work implicates platelets as important mediators of innate immune responses against invading microorganisms.


Subject(s)
Blood Platelets/metabolism , Lipopolysaccharides/adverse effects , Thrombocytopenia/chemically induced , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Binding Sites , Blood Platelets/cytology , Blood Platelets/immunology , Blotting, Western , Female , Flow Cytometry , Humans , Integrin beta3/genetics , Integrin beta3/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Knockout , Thrombin/metabolism , Thrombocytopenia/metabolism
19.
Transfusion ; 44(11): 1572-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15504162

ABSTRACT

BACKGROUND: Immunoglobulin G (IgG) anti-platelet (PLT) immunity has been shown to be initiated by indirect allorecognition where recipient T cells recognize donor PLT antigens presented by class II molecules encoded by the major histocompatibility complex (MHC) on recipient antigen-presenting cells. To understand how the recipient's MHC class II molecules may influence PLT alloimmunity, immune responsiveness against transfused PLTs was tested in different mouse strains. STUDY DESIGN AND METHODS: Various inbred and mutant mouse strains were transfused with allogeneic PLTs and IgG donor antibodies were measured by flow cytometry. RESULTS: When recipient mice, expressing both MHC class II I-A and MHC class II I-E molecules, were transfused weekly with allogeneic PLTs, high titers of IgG donor antibodies were generated. In comparison, however, recipient mice expressing only MHC class II I-A molecules had significantly (p < 0.001) reduced IgG antibody responsiveness against PLT transfusions. The low IgG responder status against allogeneic PLT transfusions was rescued in transgenic mice expressing I-E molecules and in mice genetically deficient in either beta2-microglobulin or CD8+ T cells. CONCLUSION: IgG immune responsiveness against allogeneic PLT transfusions is dependent on recipient expression of I-E MHC class II molecules, whereas I-A expression is linked with CD8-mediated suppression of PLT immunity. The data suggest that strategies to modify recipient MHC class II presentation of donor PLT antigens would be effective in eliminating PLT alloimmunity.


Subject(s)
Histocompatibility Antigens Class II/analysis , Platelet Transfusion , Animals , Blood Platelets/immunology , CD8 Antigens/genetics , Female , Histocompatibility Antigens Class II/immunology , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Phenotype , Transplantation, Homologous/immunology , beta 2-Microglobulin/deficiency
20.
Blood ; 103(7): 2705-9, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-14656873

ABSTRACT

The mechanisms responsible for immunoglobulin G (IgG) immunity against allogeneic platelets are poorly understood. We studied the role that murine recipient CD8+ T and natural killer (NK) cells play in immunity against allogeneic platelets. BALB/c mice were depleted of the cells by cell-specific antibodies, transfused weekly with platelets from C57BL/6 mice, and serum IgG antidonor antibodies were measured by flow cytometry. While allogeneic platelet transfusions into wild-type recipients stimulated IgG antidonor antibodies in all mice by the fifth transfusion, CD8-depleted mice had significantly (P<.001) enhanced antibody production. Isotype analysis revealed that CD8+ T cells suppressed T-helper 2 (Th2)-associated IgG1 but enhanced Th1-associated IgG2a. Compared with wild-type mice, platelet transfusions into CD8-depleted mice stimulated enhanced intracellular interferon (IFN)-gamma production by CD4- lymphocytes within 24 hours after the first transfusion. The early IFN-gamma response correlated with nitric oxide-dependent splenic cytotoxicity (P<.001). In asialo ganglioside monosialic acid 1 (GM1)-depleted mice transfused with allogeneic platelets, the IFN-gamma production, splenic cytotoxicity, and IgG antidonor antibody response were significantly suppressed. These results demonstrate that IgG antiplatelet immunity is dependent on an early NK cell-derived IFN-gamma response that is negatively regulated by CD8+ T cells and suggest that targeting innate NK cell responses may significantly reduce platelet alloimmunization.


Subject(s)
Blood Platelets/immunology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Immunoglobulin G/immunology , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Platelet Transfusion , Animals , CD4-Positive T-Lymphocytes/immunology , Female , Flow Cytometry , Homeostasis/immunology , Isoantibodies/immunology , Lymphocyte Depletion , Mastocytoma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Thymoma/immunology , Thymus Neoplasms/immunology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...