Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Sci ; 22(4)2021 Feb 20.
Article in English | MEDLINE | ID: mdl-33672724

ABSTRACT

The resolution of arterial thrombi is critically dependent on the endogenous fibrinolytic system. Using well-established and complementary whole blood models, we investigated the endogenous fibrinolytic potential of the tissue-type plasminogen activator (tPA) and the intra-thrombus distribution of fibrinolytic proteins, formed ex vivo under shear. tPA was present at physiologically relevant concentrations and fibrinolysis was monitored using an FITC-labelled fibrinogen tracer. Thrombi were formed from anticoagulated blood using a Chandler Loop and from non-anticoagulated blood perfused over specially-prepared porcine aorta strips under low (212 s-1) and high shear (1690 s-1) conditions in a Badimon Chamber. Plasminogen, tPA and plasminogen activator inhibitor-1 (PAI-1) concentrations were measured by ELISA. The tPA-PAI-1 complex was abundant in Chandler model thrombi serum. In contrast, free tPA was evident in the head of thrombi and correlated with fibrinolytic activity. Badimon thrombi formed under high shear conditions were more resistant to fibrinolysis than those formed at low shear. Plasminogen and tPA concentrations were elevated in thrombi formed at low shear, while PAI-1 concentrations were augmented at high shear rates. In conclusion, tPA primarily localises to the thrombus head in a free and active form. Thrombi formed at high shear incorporate less tPA and plasminogen and increased PAI-1, thereby enhancing resistance to degradation.


Subject(s)
Fibrinolysis , Shear Strength , Stress, Mechanical , Thrombosis/metabolism , Tissue Plasminogen Activator/metabolism , Animals , Fibrin/metabolism , Humans , Plasminogen/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Swine
3.
Nat Immunol ; 21(5): 513-524, 2020 05.
Article in English | MEDLINE | ID: mdl-32284594

ABSTRACT

Oxidative stress is a central part of innate immune-induced neurodegeneration. However, the transcriptomic landscape of central nervous system (CNS) innate immune cells contributing to oxidative stress is unknown, and therapies to target their neurotoxic functions are not widely available. Here, we provide the oxidative stress innate immune cell atlas in neuroinflammatory disease and report the discovery of new druggable pathways. Transcriptional profiling of oxidative stress-producing CNS innate immune cells identified a core oxidative stress gene signature coupled to coagulation and glutathione-pathway genes shared between a microglia cluster and infiltrating macrophages. Tox-seq followed by a microglia high-throughput screen and oxidative stress gene network analysis identified the glutathione-regulating compound acivicin, with potent therapeutic effects that decrease oxidative stress and axonal damage in chronic and relapsing multiple sclerosis models. Thus, oxidative stress transcriptomics identified neurotoxic CNS innate immune populations and may enable discovery of selective neuroprotective strategies.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/genetics , Gene Expression Profiling/methods , Microglia/physiology , Multiple Sclerosis/genetics , Neurogenic Inflammation/genetics , Animals , Antioxidants/therapeutic use , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Female , Gene Regulatory Networks , High-Throughput Screening Assays , Humans , Immunity, Innate , Isoxazoles/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Multiple Sclerosis/drug therapy , Neurogenic Inflammation/drug therapy , Oxidative Stress , Sequence Analysis, RNA , Single-Cell Analysis
4.
Nat Immunol ; 19(11): 1212-1223, 2018 11.
Article in English | MEDLINE | ID: mdl-30323343

ABSTRACT

Activation of innate immunity and deposition of blood-derived fibrin in the central nervous system (CNS) occur in autoimmune and neurodegenerative diseases, including multiple sclerosis (MS) and Alzheimer's disease (AD). However, the mechanisms that link disruption of the blood-brain barrier (BBB) to neurodegeneration are poorly understood, and exploration of fibrin as a therapeutic target has been limited by its beneficial clotting functions. Here we report the generation of monoclonal antibody 5B8, targeted against the cryptic fibrin epitope γ377-395, to selectively inhibit fibrin-induced inflammation and oxidative stress without interfering with clotting. 5B8 suppressed fibrin-induced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation and the expression of proinflammatory genes. In animal models of MS and AD, 5B8 entered the CNS and bound to parenchymal fibrin, and its therapeutic administration reduced the activation of innate immunity and neurodegeneration. Thus, fibrin-targeting immunotherapy inhibited autoimmunity- and amyloid-driven neurotoxicity and might have clinical benefit without globally suppressing innate immunity or interfering with coagulation in diverse neurological diseases.


Subject(s)
Antibodies, Monoclonal/immunology , Fibrinogen/antagonists & inhibitors , Neurodegenerative Diseases/immunology , Animals , Epitopes , Humans , Inflammation/immunology , Mice , Rats
5.
Nat Commun ; 6: 8164, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26353940

ABSTRACT

Autoimmunity and macrophage recruitment into the central nervous system (CNS) are critical determinants of neuroinflammatory diseases. However, the mechanisms that drive immunological responses targeted to the CNS remain largely unknown. Here we show that fibrinogen, a central blood coagulation protein deposited in the CNS after blood-brain barrier disruption, induces encephalitogenic adaptive immune responses and peripheral macrophage recruitment into the CNS leading to demyelination. Fibrinogen stimulates a unique transcriptional signature in CD11b(+) antigen-presenting cells inducing the recruitment and local CNS activation of myelin antigen-specific Th1 cells. Fibrinogen depletion reduces Th1 cells in the multiple sclerosis model, experimental autoimmune encephalomyelitis. Major histocompatibility complex (MHC) II-dependent antigen presentation, CXCL10- and CCL2-mediated recruitment of T cells and macrophages, respectively, are required for fibrinogen-induced encephalomyelitis. Inhibition of the fibrinogen receptor CD11b/CD18 protects from all immune and neuropathologic effects. Our results show that the final product of the coagulation cascade is a key determinant of CNS autoimmunity.


Subject(s)
Autoimmunity/immunology , Brain/immunology , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Fibrinogen/immunology , Genes, MHC Class II/immunology , Macrophages/immunology , Spinal Cord/immunology , Th1 Cells/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/genetics , Adaptive Immunity/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/genetics , Antigen Presentation/immunology , Autoimmunity/drug effects , Autoimmunity/genetics , Blood-Brain Barrier , Brain/drug effects , Brain/metabolism , Brain/pathology , CD11b Antigen/genetics , CD11b Antigen/immunology , CX3C Chemokine Receptor 1 , Cell Proliferation , Chemokine CCL2/immunology , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokines , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Demyelinating Diseases/genetics , Fibrin , Fibrinogen/pharmacology , Flow Cytometry , Gene Expression Profiling , Genes, MHC Class II/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Immunohistochemistry , Mice , Mice, Knockout , Microglia , Myelin-Oligodendrocyte Glycoprotein/immunology , Rats , Receptors, Antigen, T-Cell/immunology , Receptors, Chemokine/genetics , Receptors, Chemokine/immunology , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/pathology
6.
eNeuro ; 2(2)2015.
Article in English | MEDLINE | ID: mdl-26213713

ABSTRACT

Hypoxia-like tissue alterations, characterized by the upregulation of hypoxia-inducible factor-1α (HIF-1α), have been described in the normal appearing white matter and pre-demyelinating lesions of multiple sclerosis (MS) patients. As HIF-1α regulates the transcription of a wide set of genes involved in neuroprotection and neuroinflammation, HIF-1α expression may contribute to the pathogenesis of inflammatory demyelination. To test this hypothesis, we analyzed the effect of cell-specific genetic ablation or overexpression of HIF-1α on the onset and progression of experimental autoimmune encephalomyelitis (EAE), a mouse model for MS. HIF-1α was mainly expressed in astrocytes and microglia/macrophages in the mouse spinal cord at the peak of EAE. However, genetic ablation of HIF-1α in astrocytes and/or myeloid cells did not ameliorate clinical symptoms. Furthermore, conditional knock-out of Von Hippel Lindau, a negative regulator of HIF-1α stabilization, failed to exacerbate the clinical course of EAE. In accordance with clinical symptoms, genetic ablation or overexpression of HIF-1α did not change the extent of spinal cord inflammation and demyelination. Overall, our data indicate that despite dramatic upregulation of HIF-1α in astrocytes and myeloid cells in EAE, HIF-1α expression in these two cell types is not required for the development of inflammatory demyelination. Despite numerous reports indicating HIF-1α expression in glia, neurons, and inflammatory cells in the CNS of MS patients, the cell-specific contribution of HIF-1α to disease pathogenesis remains unclear. Here we show that although HIF-1α is dramatically upregulated in astrocytes and myeloid cells in EAE, cell-specific depletion of HIF-1α in these two cell types surprisingly does not affect the development of neuroinflammatory disease. Together with two recently published studies showing a role for oligodendrocyte-specific HIF-1α in myelination and T-cell-specific HIF-1α in EAE, our results demonstrate a tightly regulated cellular specificity for HIF-1α contribution in nervous system pathogenesis.

7.
Ann Neurol ; 75(2): 303-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24740641

ABSTRACT

Although multiple sclerosis (MS) has been associated with the coagulation system, the temporal and spatial regulation of coagulation activity in neuroinflammatory lesions is unknown. Using a novel molecular probe, we characterized the activity pattern of thrombin, the central protease of the coagulation cascade, in experimental autoimmune encephalomyelitis. Thrombin activity preceded onset of neurological signs, increased at disease peak, and correlated with fibrin deposition, microglial activation, demyelination, axonal damage, and clinical severity. Mice with a genetic deficit in prothrombin confirmed the specificity of the thrombin probe. Thrombin activity might be exploited for developing sensitive probes for preclinical detection and monitoring of neuroinflammation and MS progression.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Thrombin/metabolism , Animals , Axons/pathology , Blood Coagulation Factors/chemistry , Connexin 30 , Connexins/genetics , Demyelinating Diseases/etiology , Demyelinating Diseases/pathology , Disease Models, Animal , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Fibrin/metabolism , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Basic Protein/metabolism , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , Poly I-C/toxicity , Thrombin/chemistry
8.
Nat Commun ; 3: 1227, 2012.
Article in English | MEDLINE | ID: mdl-23187627

ABSTRACT

Blood-brain barrier disruption, microglial activation and neurodegeneration are hallmarks of multiple sclerosis. However, the initial triggers that activate innate immune responses and their role in axonal damage remain unknown. Here we show that the blood protein fibrinogen induces rapid microglial responses toward the vasculature and is required for axonal damage in neuroinflammation. Using in vivo two-photon microscopy, we demonstrate that microglia form perivascular clusters before myelin loss or paralysis onset and that, of the plasma proteins, fibrinogen specifically induces rapid and sustained microglial responses in vivo. Fibrinogen leakage correlates with areas of axonal damage and induces reactive oxygen species release in microglia. Blocking fibrin formation with anticoagulant treatment or genetically eliminating the fibrinogen binding motif recognized by the microglial integrin receptor CD11b/CD18 inhibits perivascular microglial clustering and axonal damage. Thus, early and progressive perivascular microglial clustering triggered by fibrinogen leakage upon blood-brain barrier disruption contributes to axonal damage in neuroinflammatory disease.


Subject(s)
Axons/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Fibrinogen/physiology , Microglia/pathology , Animals , Axons/physiology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Fibrin/physiology , Mice , Mice, Inbred C57BL , Microglia/physiology , Microscopy, Fluorescence, Multiphoton , Reactive Oxygen Species/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology
9.
Dev Neurobiol ; 71(11): 1018-39, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21780303

ABSTRACT

The blood-brain barrier (BBB) is formed primarily to protect the brain microenvironment from the influx of plasma components, which may disturb neuronal functions. The BBB is a functional unit that consists mainly of specialized endothelial cells (ECs) lining the cerebral blood vessels, astrocytes, and pericytes. The BBB is a dynamic structure that is altered in neurologic diseases, such as stroke. ECs and astrocytes secrete extracellular matrix (ECM) proteins to generate and maintain the basement membranes (BMs). ECM receptors, such as integrins and dystroglycan, are also expressed at the brain microvasculature and mediate the connections between cellular and matrix components in physiology and disease. ECM proteins and receptors elicit diverse molecular signals that allow cell adaptation to environmental changes and regulate growth and cell motility. The composition of the ECM is altered upon BBB disruption and directly affects the progression of neurologic disease. The purpose of this review is to discuss the dynamic changes of ECM composition and integrin receptor expression that control BBB functions in physiology and pathology.


Subject(s)
Blood-Brain Barrier/physiology , Brain/metabolism , Brain/physiology , Extracellular Matrix Proteins/physiology , Receptors, Cell Surface/physiology , Stroke/physiopathology , Animals , Blood-Brain Barrier/metabolism , Brain/blood supply , Humans , Microcirculation/physiology , Stroke/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...