Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 124(2): 543-52, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24463452

ABSTRACT

Th cells are the major effector cells in transplant rejection and can be divided into Th1, Th2, Th17, and Treg subsets. Th differentiation is controlled by transcription factor expression, which is driven by positive and negative cytokine and chemokine stimuli at the time of T cell activation. Here we discovered that chemokine platelet factor 4 (PF4) is a negative regulator of Th17 differentiation. PF4-deficient and platelet-deficient mice had exaggerated immune responses to cardiac transplantation, including increased numbers of infiltrating Th17 cells and increased plasma IL-17. Although PF4 has been described as a platelet-specific molecule, we found that activated T cells also express PF4. Furthermore, bone marrow transplantation experiments revealed that T cell-derived PF4 contributes to a restriction in Th17 differentiation. Taken together, the results of this study demonstrate that PF4 is a key regulator of Th cell development that is necessary to limit Th17 differentiation. These data likely will impact our understanding of platelet-dependent regulation of T cell development, which is important in many diseases, in addition to transplantation.


Subject(s)
Graft Rejection/metabolism , Heart Transplantation , Platelet Factor 4/metabolism , Th17 Cells/cytology , Animals , Blood Platelets/cytology , Bone Marrow Cells/cytology , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Enzyme-Linked Immunosorbent Assay , Homeostasis , Humans , Interleukin-17/blood , Jurkat Cells , Mice , Mice, Transgenic , Recombinant Proteins/metabolism , Time Factors , Transgenes
2.
J Immunol ; 190(9): 4685-91, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23536632

ABSTRACT

Platelets are most recognized as the cellular mediator of thrombosis, but they are increasingly appreciated for their immunomodulatory roles, including responses to Plasmodium infection. Platelet interactions with endothelial cells and leukocytes contribute significantly to the pathogenesis of experimental cerebral malaria (ECM). Recently, it has been suggested that platelets not only have an adverse role in cerebral malaria, but platelets may also be protective in animal models of uncomplicated malaria. We now demonstrate that these diverse and seemingly contradictory roles for platelets extend to cerebral malaria models and are dependent on the timing of platelet activation during infection. Our data show that platelets are activated very early in ECM and have a central role in initiation of the acute-phase response to blood-stage infection. Unlike platelet depletion or inhibition postinfection, preinfection platelet depletion or treatment with a platelet inhibitor is not protective. Additionally, we show that platelet-driven acute-phase responses have a major role in protecting mice from ECM by limiting parasite growth. Our data now suggest that platelets have a complex role in ECM pathogenesis: platelets help limit parasite growth early postinfection, but with continued platelet activation as the disease progresses, platelets contribute to ECM-associated inflammation.


Subject(s)
Acute-Phase Reaction/immunology , Blood Platelets/immunology , Malaria, Cerebral/blood , Malaria/blood , Platelet Activation/immunology , Animals , Disease Models, Animal , Malaria/immunology , Malaria, Cerebral/immunology , Mice , Mice, Inbred C57BL , Plasmodium berghei
3.
J Immunol ; 189(2): 916-23, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22706078

ABSTRACT

Platelets are most recognized for their vital role as the cellular mediator of thrombosis, but platelets also have important immune functions. Platelets initiate and sustain vascular inflammation in many disease conditions, including arthritis, atherosclerosis, transplant rejection, and severe malaria. We now demonstrate that platelets express T cell costimulatory molecules, process and present Ag in MHC class I, and directly activate naive T cells in a platelet MHC class I-dependent manner. Using an experimental cerebral malaria mouse model, we also demonstrate that platelets present pathogen-derived Ag to promote T cell responses in vivo, and that platelets can be used in a cell-based vaccine model to induce protective immune responses. Our study demonstrates a novel Ag presentation role for platelets.


Subject(s)
Antigen Presentation/immunology , Blood Platelets/immunology , Histocompatibility Antigens Class I/immunology , Animals , Blood Platelets/metabolism , Blood Platelets/parasitology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/parasitology , CD8-Positive T-Lymphocytes/pathology , Cell Movement/immunology , Histocompatibility Antigens Class I/blood , Histocompatibility Antigens Class I/genetics , Humans , Lymphocyte Activation/immunology , Malaria/blood , Malaria/immunology , Mice , Mice, Inbred C57BL , Plasmodium berghei/immunology
4.
Infect Immun ; 79(4): 1750-8, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21245265

ABSTRACT

Cerebral malaria (CM) is a major complication of Plasmodium falciparum infection, particularly in children. The pathogenesis of cerebral malaria involves parasitized red blood cell (RBC)-mediated vascular inflammation, immune stimulation, loss of blood-brain barrier integrity, and obstruction of cerebral capillaries. Therefore, blunting vascular inflammation and immune cell recruitment is crucial in limiting the disease course. Beta interferon (IFN-ß) has been used in the treatment of diseases, such as multiple sclerosis (MS) but has not yet been explored in the treatment of CM. Therefore, we sought to determine whether IFN-ß also limits disease progression in experimental cerebral malaria (ECM). Plasmodium berghei-infected mice treated with IFN-ß died later and showed increased survival, with improved blood-brain barrier function, compared to infected mice. IFN-ß did not alter systemic parasitemia. However, we identified multiple action sites that were modified by IFN-ß administration. P. berghei infection resulted in increased expression of chemokine (C-X-C motif) ligand 9 (CXCL9) in brain vascular endothelial cells that attract T cells to the brain, as well as increased T-cell chemokine (C-X-C motif) receptor 3 (CXCR3) expression. The infection also increased the cellular content of intercellular adhesion molecule 1 (ICAM-1), a molecule important for attachment of parasitized RBCs to the endothelial cell. In this article, we report that IFN-ß treatment leads to reduction of CXCL9 and ICAM-1 in the brain, reduction of T-cell CXCR3 expression, and downregulation of serum tumor necrosis factor alpha (TNF-α). In addition, IFN-ß-treated P. berghei-infected mice also had fewer brain T-cell infiltrates, further demonstrating its protective effects. Hence, IFN-ß has important anti-inflammatory properties that ameliorate the severity of ECM and prolong mouse survival.


Subject(s)
Brain/drug effects , Immunologic Factors/pharmacology , Interferon-beta/therapeutic use , Malaria, Cerebral/drug therapy , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/immunology , Brain/metabolism , Chemokine CXCL9/biosynthesis , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Intercellular Adhesion Molecule-1/biosynthesis , Malaria, Cerebral/immunology , Mice , Mice, Inbred C57BL , Plasmodium berghei , Receptors, CXCR3/biosynthesis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/blood
5.
PLoS One ; 5(5): e10413, 2010 May 03.
Article in English | MEDLINE | ID: mdl-20454664

ABSTRACT

Cerebral malaria continues to be a difficult to treat complication of Plasmodium falciparum infection in children. We have shown that platelets can have major deleterious immune functions in experimental cerebral malaria (ECM). One of the platelet derived mediators we have identified as particularly important is platelet factor 4/CXCL4. Our prior work demonstrated that PF4(-/-) mice are protected from ECM, have reduced plasma cytokines, and have reduced T-cell trafficking to the brain. We now show that PF4 drives monocyte cytokine production in a Kruppel like factor 4 (KLF4) dependent manner. Monocyte depleted Plasmodium berghei infected mice have improved survival, and KLF4 is greatly increased in control, but not monocyte depleted mice. PF4(-/-) mice have less cerebral monocyte trafficking and no change in KLF4 expression. These data indicate that PF4 induction of monocyte KLF4 expression may be an important step in the pathogenesis of ECM.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Monocytes/metabolism , Platelet Factor 4/metabolism , Animals , Cytokines/biosynthesis , Humans , Kruppel-Like Factor 4 , Mice , Mice, Inbred C57BL
6.
Circ Res ; 105(6): 595-603, 2009 Sep 11.
Article in English | MEDLINE | ID: mdl-19679838

ABSTRACT

RATIONALE: Glutamate is a major signaling molecule that binds to glutamate receptors including the ionotropic glutamate receptors; kainate (KA) receptor (KAR), the N-methyl-d-aspartate receptor, and the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor. Each is well characterized in the central nervous system, but glutamate has important signaling roles in peripheral tissues as well, including a role in regulating platelet function. OBJECTIVE: Our previous work has demonstrated that glutamate is released by platelets in high concentrations within a developing thrombus and increases platelet activation and thrombosis. We now show that platelets express a functional KAR that drives increased agonist induced platelet activation. METHODS AND RESULTS: KAR induced increase in platelet activation is in part the result of activation of platelet cyclooxygenase in a mitogen-activated protein kinase-dependent manner. Platelets derived from KAR subunit knockout mice (GluR6(-/-)) are resistant to KA effects and have a prolonged time to thrombosis in vivo. Importantly, we have also identified polymorphisms in KAR subunits that are associated with phenotypic changes in platelet function in a large group of whites and blacks. CONCLUSIONS: Our data demonstrate that glutamate regulation of platelet activation is in part cyclooxygenase-dependent and suggest that the KAR is a novel antithrombotic target.


Subject(s)
Blood Platelets/metabolism , Platelet Activation , Prostaglandin-Endoperoxide Synthases/metabolism , Receptors, Kainic Acid/metabolism , Thrombosis/metabolism , Animals , Excitatory Amino Acid Agonists/pharmacology , Humans , Kainic Acid/pharmacology , Mice , Mice, Knockout , Prostaglandin-Endoperoxide Synthases/genetics , Receptors, Kainic Acid/genetics , Thrombosis/genetics , GluK2 Kainate Receptor
7.
Cell Host Microbe ; 4(2): 179-87, 2008 Aug 14.
Article in English | MEDLINE | ID: mdl-18692777

ABSTRACT

Cerebral malaria (CM) is a major complication of Plasmodium falciparum infection in children. The pathogenesis of CM involves vascular inflammation, immune stimulation, and obstruction of cerebral capillaries. Platelets have a prominent role in both immune responses and vascular obstruction. We now demonstrate that the platelet-derived chemokine, platelet factor 4 (PF4)/CXCL4, promotes the development of experimental cerebral malaria (ECM). Plasmodium-infected red blood cells (RBCs) activated platelets independently of vascular effects, resulting in increased plasma PF4. PF4 or chemokine receptor CXCR3 null mice had less severe ECM, including decreased T cell recruitment to the brain, and platelet depletion or aspirin treatment reduced the development of ECM. We conclude that Plasmodium-infected RBCs can directly activate platelets, and platelet-derived PF4 then contributes to immune activation and T cell trafficking as part of the pathogenesis of ECM.


Subject(s)
Host-Pathogen Interactions , Malaria, Cerebral/immunology , Plasmodium falciparum/physiology , Platelet Factor 4/immunology , Animals , Brain/immunology , Brain/parasitology , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasmodium falciparum/immunology , Platelet Activation , Platelet Factor 4/genetics , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism
8.
Cancer Res ; 67(9): 4328-36, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483346

ABSTRACT

15(S)-Hydroxyeicosatetraenoic acid [15(S)-HETE] activated signal transducer and activator of transcription 3 (STAT3) as measured by its tyrosine phosphorylation, translocation from the cytoplasm to the nucleus, DNA binding, and reporter gene activity in human dermal microvascular endothelial cells (HDMVEC). Inhibition of STAT3 activation via adenovirus-mediated expression of its dominant-negative mutant suppressed 15(S)-HETE-induced HDMVEC migration and tube formation in vitro and aortic ring and Matrigel plug angiogenesis in vivo. 15(S)-HETE induced the expression of vascular endothelial growth factor (VEGF) in a time- and STAT3-dependent manner in HDMVEC. In addition, neutralizing anti-VEGF antibodies blocked 15(S)-HETE-induced HDMVEC migration and tube formation in vitro and aortic ring and Matrigel plug angiogenesis in vivo. Together, these results show for the first time that 15(S)-HETE-induced angiogenesis requires STAT3-dependent expression of VEGF. In view of these findings, it is suggested that eicosanoids, particularly 15(S)-HETE, via its capacity to stimulate angiogenesis, may influence the progression of cancer and vascular disease.


Subject(s)
Hydroxyeicosatetraenoic Acids/pharmacology , Neovascularization, Physiologic/drug effects , STAT3 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Aorta/drug effects , Cell Movement/physiology , Collagen , Drug Combinations , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , In Vitro Techniques , Laminin , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Proteoglycans , Rats , Rats, Inbred F344
9.
Circ Res ; 98(7): 913-22, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16527988

ABSTRACT

Previously, we have demonstrated that STAT-3 plays a role in thrombin-induced VSMC motility. To learn more about the role of STATs in the mitogenic and chemotactic signaling events of thrombin, here we have studied the role of STAT-5. Thrombin activated STAT-5 as measured by its tyrosine phosphorylation, DNA binding, and reporter gene activity. Inhibition of STAT-5B, but not STAT-5A, by adenovirus-mediated expression of its respective dominant-negative mutants suppressed thrombin-induced VSMC growth and motility. Thrombin induced the expression of Hsp27 and FGF-2 in a time- and STAT-5B-dependent manner in VSMC. In addition, small interfering RNA-directed depletion of Hsp27 levels or adenovirus-mediated expression of its dominant-negative mutant attenuated thrombin-induced FGF-2 expression, growth, and motility of VSMC. An increased association of STAT-5B with STAT-3 occurred in response to thrombin and adenovirus-mediated expression of dnSTAT-3 suppressed thrombin-induced Hsp27 and FGF-2 induction, DNA synthesis and motility in VSMC. Together, these results indicate that thrombin-induced VSMC growth and motility require STAT-5B/STAT-3-dependent expression of Hsp27 and FGF-2. These observations also suggest that STAT-5B/STAT-3/Hsp27/FGF-2 signaling via its involvement in the regulation of VSMC growth and motility may play an important role in the pathogenesis of vascular diseases such as restenosis after angioplasty.


Subject(s)
Fibroblast Growth Factor 2/physiology , Heat-Shock Proteins/metabolism , Muscle, Smooth, Vascular/physiology , Neoplasm Proteins/metabolism , STAT3 Transcription Factor/physiology , STAT5 Transcription Factor/metabolism , Thrombin/physiology , Animals , Cell Movement , DNA Primers , Fibroblast Growth Factor 2/genetics , HSP27 Heat-Shock Proteins , Heat-Shock Proteins/genetics , Muscle, Smooth, Vascular/growth & development , Neoplasm Proteins/genetics , Polymerase Chain Reaction , Rats , STAT5 Transcription Factor/genetics , Transcription, Genetic
10.
Clin Chim Acta ; 317(1-2): 213-20, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11814478

ABSTRACT

BACKGROUND: Non-insulin-dependent diabetes mellitus (NIDDM) is characterized by the presence of abnormally active platelets in the circulation, leading to increased incidence of thrombotic complications. In this study, we have attempted to understand the pathophysiology of the platelets in NIDDM. METHODS: Platelet aggregation was induced by thrombin receptor-activating peptide or epinephrine. Membrane fluidity was derived from the steady-state fluorescence anisotropy of diphenylhexatriene incorporated in the membrane. The phosphotyrosine content of the platelet proteins was probed using specific monoclonal antibodies. The extent of calpain activity was assessed from the proteolysis of calpain substrates. RESULTS: Aggregation was significantly enhanced (p<0.001) in the platelets obtained from the cases of NIDDM. Anisotropy measurements reflected a significant increase in the microviscosity of platelet membranes from 3.315 (+/-0.103) in the control to 4.153 (+/-0.119) in NIDDM. Proteins of relative mobilities of 131, 100, 47 and 38 kDa were found to remain phosphorylated on tyrosine in the resting platelets obtained from NIDDM patients, while they were not phosphorylated in the control counterparts. This was associated with heightened activity of the calcium-dependent thiol protease, calpain, in NIDDM. CONCLUSIONS: Taken together, these data indicated significant changes in the signaling mechanism in the platelets obtained from NIDDM, which could lead to platelet hyperactivity in this disease.


Subject(s)
Blood Platelets/metabolism , Cell Membrane/physiology , Diabetes Mellitus, Type 2/blood , Adult , Blood Platelets/cytology , Calcimycin/pharmacology , Calpain/drug effects , Calpain/metabolism , Case-Control Studies , Cell Membrane/drug effects , Diabetes Mellitus, Type 2/pathology , Epinephrine/pharmacology , Humans , Middle Aged , Phosphorylation , Pilot Projects , Platelet Activation , Platelet Aggregation/drug effects , Reference Values , Signal Transduction , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...