Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Arthritis Rheum ; 56(8): 2512-22, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17665439

ABSTRACT

OBJECTIVE: Fibroblast-like synoviocytes (FLS) are a major constituent of the hyperplastic synovial pannus that aggressively invades cartilage and bone during the course of rheumatoid arthritis (RA). Fractalkine (FKN/CX(3)CL1) expression is up-regulated in RA synovium and RA synovial fluid. While RA FLS express the FKN receptor, CX(3)CR1, the pathophysiologic relevance of FKN stimulation of RA FLS is not understood. This study was undertaken to better characterize the relationship between FKN and the RA FLS that both produce it and express its receptor. METHODS: RA FLS were subjected to chemotaxis and proliferation assays, Western blotting, enzyme-linked immunosorbent assays, and filamentous actin staining to characterize the relationship between FKN and RA FLS. RESULTS: FKN secretion by RA FLS was regulated mainly by tumor necrosis factor alpha. Stimulation of RA FLS with FKN led to significant cytoskeletal rearrangement but no proliferation. Chemotaxis assays revealed that FKN was a novel chemoattractant for RA FLS. Stimulation of RA FLS with FKN resulted in activation of MAP kinases and Akt. JNK, ERK-1/2, and Akt (at both Ser-473 and Thr-308) were each up-regulated in a time-dependent manner. Inhibition of ERK-1/2-mediated signaling, but not JNK or Akt, significantly repressed FKN-induced RA FLS migration. CONCLUSION: These findings indicate a novel role of FKN in regulating RA FLS cytoskeletal structure and migration. FKN specifically induces RA FLS phosphorylation of the MAP kinases JNK and ERK-1/2, as well as full activation of Akt.


Subject(s)
Arthritis, Rheumatoid/metabolism , Chemokines, CX3C/metabolism , Chemotactic Factors/metabolism , MAP Kinase Signaling System/physiology , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Synovial Membrane/metabolism , Actin Cytoskeleton/metabolism , Adult , Aged , Arthritis, Rheumatoid/pathology , Blotting, Western , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/pharmacology , Chemotactic Factors/pharmacology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , MAP Kinase Signaling System/drug effects , Male , Membrane Proteins/pharmacology , Middle Aged , Recombinant Proteins , Synovial Membrane/drug effects , Synovial Membrane/pathology
2.
Circulation ; 116(7): 764-73, 2007 Aug 14.
Article in English | MEDLINE | ID: mdl-17679613

ABSTRACT

BACKGROUND: The membrane-bound chemokine fractalkine (CX3CL1) is expressed on various cell types such as activated endothelial cells and has been implicated in the inflammatory process of atherosclerosis. The aim of the present study was to dissect the role of fractalkine in leukocyte recruitment to inflamed endothelium under arterial shear forces. METHODS AND RESULTS: With the use of immunofluorescence and laminar flow assays, the present study shows that human umbilical vein endothelial cells stimulated with tumor necrosis factor-alpha and interferon-gamma abundantly express CX3CL1 and promote substantial leukocyte accumulation under arterial flow conditions. In the presence of high shear, firm adhesion of leukocytes to inflamed endothelial cells is reduced by approximately 40% by a function-blocking anti-fractalkine antibody or by an antibody directed against the fractalkine receptor (CX3CR1). With the use of intravital video-fluorescence microscopy we demonstrate that inhibition of fractalkine signaling attenuates leukocyte adhesion to the atherosclerotic carotid artery of apolipoprotein E-deficient mice, which suggests that the CX3CL1-CX3CR1 axis is critically involved in leukocyte adhesion to inflamed endothelial cells under high shear forces both in vitro and in vivo. Surprisingly, platelets were strictly required for fractalkine-induced leukocyte adhesion at high shear rates. Correspondingly, specific inhibition of platelet adhesion to inflamed endothelial cells also significantly reduced leukocyte accumulation. We show that both soluble and membrane-bound fractalkine induces platelet degranulation and subsequent surface expression of P-selectin, which thereby promotes direct platelet-leukocyte interaction. CONCLUSIONS: Fractalkine expressed by inflamed endothelial cells triggers P-selectin exposure on adherent platelets, which thereby initiates the local accumulation of leukocytes under arterial shear, an essential step in the development of atherosclerotic lesions.


Subject(s)
Atherosclerosis/immunology , Blood Platelets/metabolism , Chemokines, CX3C/metabolism , Chemotaxis, Leukocyte , Endothelium, Vascular/immunology , Membrane Proteins/metabolism , P-Selectin/metabolism , Platelet Activation , Animals , Blood Circulation , Blood Platelets/physiology , CHO Cells , Cell Adhesion , Cell Degranulation , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/pharmacology , Cricetinae , Cricetulus , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelium, Vascular/cytology , Humans , Inflammation/immunology , Inflammation Mediators/pharmacology , Leukocytes/immunology , Membrane Proteins/pharmacology , Mice
3.
Acta Med Okayama ; 61(2): 89-98, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17471309

ABSTRACT

CD16+ monocytes, identified as a minor population of monocytes in human peripheral blood, have been implicated in several inflammatory diseases, including rheumatoid arthritis (RA). Fractalkine (FKN, CX3CL1), a member of the CX3 C subfamily, is induced by pro-inflammatory cytokines, while a receptor for FKN, CX3CR1, is capable of mediating both leukocyte migration and firm adhesion. Here, we investigated the role of FKN and CX3CR1 in activation of CD16+ monocytes and their recruitment into synovial tissues in RA patients. High levels of soluble FKN were detected in the synovial fluid and sera of RA patients. Circulating CD16+ monocytes showed a higher level of CX3CR1 expression than CD16- monocytes in both RA patients and healthy subjects. High level expression of CX3CR1 was also seen in CD16+ monocytes localized to the lining layer in RA synovial tissue. In the in vitro culture experiments, IL-10 induced CX3CR1 expression on the surface of monocytes, and TNFalpha induced membrane-bound FKN as well as soluble FKN expression in synovial fibroblasts. Moreover, soluble FKN was capable of inducing IL-1beta and IL-6 by activated monocytes. These results suggest that FKN might preferentially mediate migration and recruitment of CD16+ monocytes, and might contribute to synovial tissue inflammation.


Subject(s)
Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Chemokines, CX3C/metabolism , Membrane Proteins/metabolism , Monocytes/metabolism , Monocytes/pathology , Receptors, Chemokine/metabolism , Receptors, IgG/metabolism , Aged , Arthritis, Rheumatoid/blood , CX3C Chemokine Receptor 1 , Cell Membrane/metabolism , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/blood , Chemokines, CX3C/pharmacology , Endothelial Cells/metabolism , Female , Humans , Interleukin-10/pharmacology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Membrane Proteins/blood , Membrane Proteins/pharmacology , Monocytes/drug effects , Osteoarthritis/blood , Osteoarthritis/metabolism , Osteoarthritis/pathology , Recombinant Proteins/pharmacology , Synovial Fluid/metabolism , Synovial Membrane/metabolism , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/pharmacology
4.
J Immunol ; 178(9): 5779-88, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17442962

ABSTRACT

NK cells are cytotoxic lymphocytes that also secrete regulatory cytokines and can therefore influence adaptive immune responses. NK cell function is largely controlled by genes present in a genomic region named the NK complex. It has been shown that the NK complex is a genetic determinant of murine cerebral malaria pathogenesis mediated by Plasmodium berghei ANKA. In this study, we show that NK cells are required for cerebral malaria disease induction and the control of parasitemia. NK cells were found infiltrating brains of cerebral malaria-affected mice. NK cell depletion resulted in inhibition of T cell recruitment to the brain of P. berghei-infected animals. NK cell-depleted mice displayed down-regulation of CXCR3 expression and a significant reduction of T cells migrating in response to IFN-gamma-inducible protein 10, indicating that this chemokine pathway plays an essential role in leukocyte trafficking leading to cerebral disease and fatalities.


Subject(s)
Brain/immunology , Killer Cells, Natural/immunology , Malaria, Cerebral/immunology , Plasmodium berghei , Receptors, Chemokine/metabolism , T-Lymphocyte Subsets/immunology , Animals , Brain/pathology , Chemokine CXCL10 , Chemokines, CX3C/metabolism , Chemokines, CX3C/pharmacology , Chemokines, CXC/metabolism , Chemokines, CXC/pharmacology , Disease Models, Animal , Down-Regulation , Lymphocyte Depletion , Malaria, Cerebral/pathology , Mice , Mice, Inbred C57BL , Receptors, CXCR3 , Receptors, Chemokine/analysis , Spleen/immunology , T-Lymphocyte Subsets/chemistry , T-Lymphocyte Subsets/drug effects
5.
J Neurosci ; 26(41): 10488-98, 2006 Oct 11.
Article in English | MEDLINE | ID: mdl-17035533

ABSTRACT

We examined the effects of the chemokine fractalkine (CX3CL1) on EPSCs evoked by electrical stimulation of Schaffer collaterals in patch-clamped CA1 pyramidal neurons from rat hippocampal slices. Acute application of CX3CL1 caused a sustained reduction of EPSC amplitude, with partial recovery after washout. CX3CL1-induced EPSC depression is postsynaptic in nature, because paired-pulse ratio was maintained, amplitude distribution of spontaneous excitatory postsynaptic currents shifted to lower values, and whole-cell current responses to AMPA were reversibly inhibited. EPSC depression by CX3CL1 is mediated by CX3CL1 receptor (CX3CR1), because CX3CL1 was unable to influence EPSC amplitude in CA1 pyramidal neurons from CX3CR1 knock-out mice. CX3CL1-induced depression of both EPSC and AMPA current was not observed in the absence of afferent fiber stimulation or AMPA receptor activation, respectively, indicating the requirement of sustained receptor activity for its development. Findings obtained from hippocampal slices, cultured hippocampal neurons, and transfected human embryonic kidney cells indicate that a Ca2+-, cAMP-, and phosphatase-dependent process is likely to modulate CX3CL1 effects because of the following: (1) CX3CL1-induced depression was antagonized by intracellular BAPTA, 8Br-cAMP, phosphatase inhibitors, and pertussis toxin (PTX); (2) CX3CL1 inhibited forskolin-induced cAMP formation sensitive to PTX; and (3) CX3CL1 inhibited forskolin-induced Ser845 GluR1 phosphorylation, which was sensitive to PTX and dependent on Ca2+ and phosphatase activity. Together, these findings indicate that CX3CL1 negatively modulates AMPA receptor function at active glutamatergic synapses through cell-signaling pathways by influencing the balance between kinase and phosphatase activity.


Subject(s)
Chemokines, CX3C/metabolism , Glutamic Acid/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , CX3C Chemokine Receptor 1 , Cell Line , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/genetics , Chemokines, CX3C/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Synapses/drug effects
6.
FEBS Lett ; 580(18): 4306-10, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16842787

ABSTRACT

Fractalkine (FKN) evokes nociceptive behavior in nai ve rats, whereas minocycline attenuates pain acutely after neuronal injury. We show that, in nai ve rats, FKN causes hyperresponsiveness of lumbar wide dynamic range neurons to brush, pressure and pinch applied to the hindpaw. One day after spinal nerve ligation (SNL), minocycline attenuates after-discharge and responses to brush and pressure. In contrast, minocycline does not alter evoked neuronal responses 10 days after SNL or sciatic constriction, but increases spontaneous discharge. We speculate that microglia rapidly alter sensory neuronal activity in nai ve and neuropathic rats acutely, but not chronically, after injury.


Subject(s)
Chemokines, CX3C/pharmacology , Membrane Proteins/pharmacology , Minocycline/pharmacology , Posterior Horn Cells/drug effects , Action Potentials/drug effects , Animals , Chemokine CX3CL1 , Chemokines, CX3C/antagonists & inhibitors , Ligation , Male , Membrane Proteins/antagonists & inhibitors , Pain/chemically induced , Posterior Horn Cells/physiology , Rats , Rats, Sprague-Dawley , Spinal Nerves/surgery
7.
Neuropharmacology ; 51(4): 816-21, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16815480

ABSTRACT

This work reports the effect of chemokine fractalkine/CX3CL1, an endogenous small peptide highly expressed in the central nervous system, on evoked synaptic responses investigated in mouse CA1 stratum radiatum using an electrophysiological approach. We report that in acute mouse hippocampal slices, superfusion of CX3CL1 resulted in a reversible depression of the field excitatory postsynaptic potential (fEPSP) which developed within few seconds, increased for up to 10 min of application and disappeared within 30 min after the end of CX3CL1 treatment. We also show that CX3CL1-induced synaptic depression is (i) dose-dependent with IC50 and nH values of 0.7 nM and 1, respectively, (ii) not associated with a change in paired-pulse facilitation, (iii) mediated through CX3CL1 receptor (CX3CR1), being absent in CX3CR1-/- mice and inhibited in wild-type mice by a specific blocking antibody, and (iv) occluded by the induction of homosynaptic long-term depression (LTD). We conclude that CX3CL1 is a potent neuromodulator of the evoked excitatory synaptic transmission, sharing common mechanisms with LTD.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiology , Receptors, Chemokine/physiology , Synaptic Transmission/physiology , Animals , Animals, Newborn , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Chemokines, CX3C/immunology , Chemokines, CX3C/pharmacology , Dose-Response Relationship, Drug , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/radiation effects , Hippocampus/cytology , Hippocampus/drug effects , Humans , In Vitro Techniques , Membrane Proteins/immunology , Membrane Proteins/pharmacology , Mice , Mice, Knockout , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Inhibition/radiation effects , Patch-Clamp Techniques/methods , Receptors, Chemokine/deficiency , Synaptic Transmission/drug effects , Synaptic Transmission/radiation effects
8.
Eur J Neurosci ; 22(11): 2775-82, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16324111

ABSTRACT

Fractalkine is a chemokine that is tethered to the extracellular surface of neurons. Fractalkine can be released, forming a diffusible signal. Spinal fractalkine (CX3CL1) is expressed by sensory afferents and intrinsic neurons, whereas its receptor (CX3CR1) is predominantly expressed by microglia. Pain enhancement occurs in response both to intrathecally administered fractalkine and to spinal fractalkine endogenously released by peripheral neuropathy. The present experiments examine whether fractalkine-induced pain enhancement is altered by a microglial inhibitor (minocycline) and/or by antagonists/inhibitors of three putative glial products implicated in pain enhancement: interleukin-1 (IL1), interleukin-6 (IL6) and nitric oxide (NO). In addition, it extends a prior study that demonstrated that intrathecal fractalkine-induced mechanical allodynia is blocked by a neutralizing antibody to the rat fractalkine receptor, CX3CR1. Here, intrathecal anti-CX3CR1 also blocked fractalkine-induced thermal hyperalgesia. Furthermore, blockade of microglial activation with minocycline prevented both fractalkine-induced mechanical allodynia (von Frey test) and thermal hyperalgesia (Hargreaves test). Microglial activation appears to lead to the release of IL1, given that pretreatment with IL1 receptor antagonist blocked both fractalkine-induced mechanical allodynia and thermal hyperalgesia. IL1 is not the only proinflammatory cytokine implicated, as a neutralizing antibody to rat IL6 also blocked fractalkine-induced pain facilitation. Lastly, NO appears to be importantly involved, as l-NAME, a broad-spectrum NO synthase inhibitor, also blocked fractalkine-induced effects. Taken together, these data support that neuronally released fractalkine enhances pain via activation of spinal cord glia. Thus, fractalkine may be a neuron-to-glia signal triggering pain facilitation.


Subject(s)
Chemokines, CX3C/pharmacology , Membrane Proteins/pharmacology , Pain/chemically induced , Pain/physiopathology , Spinal Cord/physiopathology , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Blocking/pharmacology , Chemokine CX3CL1 , Chemokines, CX3C/administration & dosage , Chemokines, CX3C/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hot Temperature , Hyperalgesia/prevention & control , Injections, Spinal , Interleukin-6/pharmacology , Male , Membrane Proteins/administration & dosage , Membrane Proteins/antagonists & inhibitors , Microglia/drug effects , Microinjections , Minocycline/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Pain Measurement/drug effects , Pain Threshold/drug effects , Physical Stimulation , Rats , Rats, Sprague-Dawley
9.
Arterioscler Thromb Vasc Biol ; 25(12): 2567-72, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16224053

ABSTRACT

OBJECTIVE: Recent data derived primarily from studies in animal models suggest that fractalkine (CX3CL1) and its cognate receptor, CX3CR1, play a role in atherogenesis. We, therefore, hypothesized that enhanced CX3CL1/CX3CR1 expression may promote atherogenesis in patients with coronary artery disease (CAD). METHODS AND RESULTS: We examined the plasma levels of CX3CL1 and CX3CR1 expression in peripheral blood mononuclear cells (PBMC) in various CAD populations (30 patients with previous myocardial infarction, 40 patients with stable angina, 40 patients with unstable angina, and a total of 35 controls) and used various experimental approaches to characterize CX3CL1-mediated leukocyte responses. We found that the plasma levels of CX3CL1 are greatly increased in CAD, particularly in unstable disease. The parallel increase of CX3CR1 expression in PBMC was predominantly attributable to an expansion of the (CX3CR1+)(CD3+)(CD8+) T cell subset and was associated with enhanced chemotactic, adhesive, and inflammatory responses to CX3CL1. Statin therapy for 6 months reduced the expression of CX3CL1 and CX3CR1, reaching statistical significance for both parameters only during aggressive (atorvastatin, 80 mg qd) but not conventional (simvastatin, 20 mg qd) therapy. Consequently, the functional responses of the PBMC to CX3CL1 including migration, adhesion, and secretion of interleukin-8 were attenuated by the treatments. CONCLUSIONS: Our results suggest that the CX3CL1/CX3CR1 dyad may contribute to atherogenesis and plaque destabilization in human CAD.


Subject(s)
Chemokines, CX3C/blood , Coronary Artery Disease/drug therapy , Coronary Artery Disease/physiopathology , Heptanoic Acids/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Membrane Proteins/blood , Membrane Proteins/genetics , Pyrroles/administration & dosage , Receptors, Chemokine/genetics , Angina, Unstable/drug therapy , Angina, Unstable/metabolism , Angina, Unstable/physiopathology , Atorvastatin , CX3C Chemokine Receptor 1 , Cell Adhesion/drug effects , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/genetics , Chemokines, CX3C/pharmacology , Chemotaxis/drug effects , Cholesterol, LDL/blood , Coronary Artery Disease/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Gene Expression/drug effects , Gene Expression/physiology , Humans , Interleukin-8/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Membrane Proteins/pharmacology , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Simvastatin/administration & dosage , Umbilical Veins/cytology
10.
J Immunol ; 175(6): 3737-45, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16148119

ABSTRACT

CX3CL1 (fractalkine), the only member of the delta subclass of chemokines, is a known chemotactic factor for monocytes/macrophages as well as NK cells and T lymphocytes. In several pathologies, excessive production of CX3CL1 at specific sites leads primarily to monocyte/macrophage recruitment, which causes tissue and vascular damage. Despite their clinical relevance, the mechanisms underlying monocyte/macrophage chemotaxis to CX3CL1 remain poorly documented. The present report addresses this issue and identifies cell signaling crucial for this process. Using the murine monocyte/macrophage RAW cell line, we show that CX3CL1 treatment elicits a rapid and transient increase in F-actin and the formation of F-actin-enriched cell protrusions. CX3CL1 also triggers tyrosine phosphorylation of proteins localized in those protrusions. The protein tyrosine kinase Syk is activated upon CX3CL1 treatment, and reduction of Syk expression using RNA-mediated interference results in a specific and massive impairment of RAW cell migration to CX3CL1. Similar results are obtained using the Syk inhibitor, piceatannol. Cells with reduced Syk expression also exhibit a major defect in CX3CL1-induced cytoskeletal remodeling. These data suggest that in monocytes/macrophages, Syk is essential for proper reorganization of the actin cytoskeleton in response to CX3CL1 and is therefore required for cell chemotaxis to CX3CL1.


Subject(s)
Chemokines, CX3C/pharmacology , Chemotaxis , Macrophages/physiology , Membrane Proteins/pharmacology , Protein-Tyrosine Kinases/physiology , Actins/drug effects , Animals , Cell Line , Cell Surface Extensions/drug effects , Chemokine CX3CL1 , Cytoskeleton/drug effects , Intracellular Signaling Peptides and Proteins , Mice , Monocytes/physiology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Syk Kinase
11.
J Neuroimmunol ; 166(1-2): 19-28, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16019082

ABSTRACT

Excitotoxicity is a cell death caused by excessive exposure to glutamate (Glu), contributing to neuronal degeneration in many acute and chronic CNS diseases. We explored the role of fractalkine/CX3CL1 on survival of hippocampal neurons exposed to excitotoxic doses of Glu. We found that: CX3CL1 reduces excitotoxicity when co-applied with Glu, through the activation of the ERK1/2 and PI3K/Akt pathways, or administered up to 8 h after Glu insult; CX3CL1 reduces the Glu-activated whole-cell current through mechanisms dependent on intracellular Ca2+; CX3CL1 is released from hippocampal cells after excitotoxic insult, likely providing an endogenous protective mechanism against excitotoxic cell death.


Subject(s)
Chemokines, CX3C/physiology , Glutamic Acid/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Membrane Proteins/physiology , Neuroprotective Agents , Neurotoxins/pharmacology , Animals , Cell Survival/physiology , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/administration & dosage , Chemokines, CX3C/metabolism , Chemokines, CX3C/pharmacology , Drug Administration Schedule , Drug Combinations , Electric Conductivity , Excitatory Amino Acid Agonists/pharmacology , Hippocampus/physiology , Membrane Proteins/administration & dosage , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
12.
Eur J Neurosci ; 20(12): 3222-32, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15610155

ABSTRACT

Our purpose was to investigate in human neurons the neuroprotective pathways induced by Fractalkine (FKN) against glutamate receptor-induced excitotoxicity. CX(3)CR1 and FKN are expressed constitutively in the tested human embryonic primary neurons and SK-N-SH, a human neuroblastoma cell line. Microfluorometry assay demonstrated that CX(3)CR1 was functional in 44% of primary neurons and in 70% of SK-N-SH. Fractalkine induced ERK1/2 phosphorylation within 1 min and Akt phosphorylation after 10 min, and both phosphorylation decreased after 20 min. No p38 and SAPK/JNK activation was observed after FKN treatment. Application of FKN triggered a 53% reduction of the NMDA-induced neuronal calcium influx, which was insensitive to pertussis toxin and LY294002 an inhibitor of Akt pathway, but abolished by PD98059, an ERK1/2 pathway inhibitor. Moreover, FKN significantly reduced neuronal NMDA-induced apoptosis, which was pertussis toxin insensitive and abolished in presence of PD98059 and LY294002. In conclusion, FKN protected human neurons from NMDA-mediated excitotoxicity in at least two ways with different kinetics: (i) an early ERK1/2 activation which reduced NMDA-mediated calcium flux; and (ii), a late Akt activation associated with the previously induced ERK1/2 activation.


Subject(s)
Apoptosis/physiology , Calcium/metabolism , Chemokines, CX3C/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Membrane Proteins/biosynthesis , N-Methylaspartate/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/genetics , Chemokines, CX3C/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Humans , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Neurons/drug effects , Neurons/enzymology
13.
J Neurosci ; 24(33): 7353-65, 2004 Aug 18.
Article in English | MEDLINE | ID: mdl-15317861

ABSTRACT

The present experiments examined the role of spinal proinflammatory cytokines [interleukin-1beta (IL-1)] and chemokines (fractalkine) in acute analgesia and in the development of analgesic tolerance, thermal hyperalgesia, and tactile allodynia in response to chronic intrathecal morphine. Chronic (5 d), but not acute (1 d), intrathecal morphine was associated with a rapid increase in proinflammatory cytokine protein and/or mRNA in dorsal spinal cord and lumbosacral CSF. To determine whether IL-1 release modulates the effects of morphine, intrathecal morphine was coadministered with intrathecal IL-1 receptor antagonist (IL-1ra). This regimen potentiated acute morphine analgesia and inhibited the development of hyperalgesia, allodynia, and analgesic tolerance. Similarly, intrathecal IL-1ra administered after the establishment of morphine tolerance reversed hyperalgesia and prevented the additional development of tolerance and allodynia. Fractalkine also appears to modulate the effects of intrathecal morphine because coadministration of morphine with intrathecal neutralizing antibody against the fractalkine receptor (CX3CR1) potentiated acute morphine analgesia and attenuated the development of tolerance, hyperalgesia, and allodynia. Fractalkine may be exerting these effects via IL-1 because fractalkine (CX3CL1) induced the release of IL-1 from acutely isolated dorsal spinal cord in vitro. Finally, gene therapy with an adenoviral vector encoding for the release of the anti-inflammatory cytokine IL-10 also potentiated acute morphine analgesia and attenuated the development of tolerance, hyperalgesia, and allodynia. Taken together, these results suggest that IL-1 and fractalkine are endogenous regulators of morphine analgesia and are involved in the increases in pain sensitivity that occur after chronic opiates.


Subject(s)
Analgesics, Opioid/pharmacology , Chemokines, CX3C/physiology , Hyperalgesia/immunology , Interleukin-1/physiology , Membrane Proteins/physiology , Morphine/pharmacology , Analgesics, Opioid/administration & dosage , Animals , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Chemokines, CX3C/pharmacology , Drug Tolerance , Genetic Therapy , Hot Temperature , Hyperalgesia/therapy , Inflammation/immunology , Injections, Spinal , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/biosynthesis , Interleukin-1/cerebrospinal fluid , Interleukin-10/genetics , Male , Membrane Proteins/pharmacology , Morphine/administration & dosage , Pain/immunology , Pain Management , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Cytokine/antagonists & inhibitors , Receptors, HIV/antagonists & inhibitors , Sialoglycoproteins/administration & dosage , Sialoglycoproteins/pharmacology , Spinal Cord/drug effects , Spinal Cord/immunology
14.
Stem Cells ; 22(3): 415-27, 2004.
Article in English | MEDLINE | ID: mdl-15153618

ABSTRACT

Mesenchymal stem cells (MSCs), cultured ex vivo, recently were shown to be able to migrate into sites of brain injuries when transplanted systemically or locally, suggesting that MSCs possess migratory capacity. However, the mechanisms underlying the migration of these cells remain unclear. In this study, we examined the role of some chemokines and their receptors in the trafficking of rat MSCs (rMSCs) in a rat model of left hypoglossal nerve injury. rMSCs transplanted into the lateral ventricles of the rat brain migrated to the avulsed hypoglossal nucleus, where the expression of chemokines, stromal-cell-derived factor 1 (SDF-1), and fractalkine was observed to be increased. This increase temporally paralleled the migration of rMSCs into the avulsed nucleus at 1 and 2 weeks after operation. It has been found that rMSCs express CXCR4 and CX3CR1, the respective receptors for SDF-1 and fractalkine, and other chemokine receptors, CCR2 and CCR5. Furthermore, in vitro analysis revealed that recombinant human SDF-1 alpha (rhSDF-1alpha) and recombinant rat fractalkine (rrfractalkine) induced the migration of rMSCs in a G-protein-dependent manner. Intracerebral injection of rhSDF-1alpha has also been shown to stimulate the homing of transplanted rMSCs to the site of injection in the brain. These data suggest that the interactions of fractalkine-CX3CR1 and SDF-1-CXCR4 could partially mediate the trafficking of transplanted rMSCs. This study provides an important insight into the understanding of the mechanisms governing the trafficking of transplanted rMSCs and also significantly expands the potential role of MSCs in cell therapy for brain injuries and diseases.


Subject(s)
Cell Movement/physiology , Chemokines, CXC/pharmacology , Hypoglossal Nerve Injuries , Mesenchymal Stem Cells/cytology , Receptors, Chemokine/metabolism , Animals , Brain/pathology , Cell Movement/drug effects , Cells, Cultured , Chemokine CX3CL1 , Chemokine CXCL12 , Chemokines, CX3C/pharmacology , GTP-Binding Proteins/metabolism , Hypoglossal Nerve/cytology , Hypoglossal Nerve/metabolism , Membrane Proteins/pharmacology , Mesenchymal Stem Cells/metabolism , Rats , Rats, Wistar , Receptors, CXCR4/metabolism
15.
Circ Res ; 94(6): 776-84, 2004 Apr 02.
Article in English | MEDLINE | ID: mdl-14963004

ABSTRACT

Leukocyte recruitment is crucial for the response to vascular injury in spontaneous and accelerated atherosclerosis. Whereas the mechanisms of leukocyte adhesion to endothelium or matrix-bound platelets have been characterized, less is known about the proadhesive role of smooth muscle cells (SMCs) exposed after endothelial denudation. In laminar flow assays, neointimal rat SMCs (niSMCs) supported a 2.5-fold higher arrest of monocytes and "memory" T lymphocytes than medial SMCs, which was dependent on both P-selectin and VLA-4, as demonstrated by blocking antibodies. The increase in monocyte arrest on niSMCs was triggered by the CXC chemokine GRO-alpha and fractalkine, whereas "memory" T cell arrest was mediated by stromal cell-derived factor (SDF)-1alpha. This functional phenotype was paralleled by a constitutively increased mRNA and surface expression of P-selectin and of relevant chemokines in niSMCs, as assessed by real-time PCR and flow cytometry. The increased expression of P-selectin in niSMCs versus medial SMCs was associated with enhanced NF-kappaB activity, as revealed by immunofluorescence staining for nuclear p65 in vitro. Inhibition of NF-kappaB by adenoviral IkappaBalpha in niSMCs resulted in a marked reduction of increased leukocyte arrest in flow. Furthermore, P-selectin expression by niSMCs in vivo was confirmed in a hypercholesterolemic mouse model of vascular injury by double immunofluorescence and by RT-PCR after laser microdissection. In conclusion, we have identified a NF-kappaB-mediated proinflammatory phenotype of niSMCs that is characterized by increased P-selectin and chemokine expression and thereby effectively supports leukocyte recruitment.


Subject(s)
Chemotaxis, Leukocyte/physiology , Inflammation/pathology , Integrin alpha4beta1/physiology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/physiology , P-Selectin/physiology , Animals , Aorta, Thoracic/injuries , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Cell Adhesion , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Chemokine CX3CL1 , Chemokines, CX3C/pharmacology , Constriction, Pathologic , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/physiology , Endothelium, Vascular/injuries , Gene Expression Regulation , Hypercholesterolemia/metabolism , I-kappa B Proteins/physiology , Inflammation/metabolism , Integrin alpha4beta1/biosynthesis , Integrin alpha4beta1/genetics , Membrane Proteins/pharmacology , Mice , Mice, Knockout , Monocytes/cytology , Muscle, Smooth, Vascular/cytology , NF-KappaB Inhibitor alpha , P-Selectin/biosynthesis , P-Selectin/genetics , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-8B/physiology , Recombinant Fusion Proteins/physiology , Recurrence , Rheology , T-Lymphocyte Subsets/cytology
16.
Stem Cells ; 22(1): 109-18, 2004.
Article in English | MEDLINE | ID: mdl-14688397

ABSTRACT

Many cell types in the brain express chemokines and chemokine receptors under homeostatic conditions, arguing for a role of these proteins in normal brain processes. Because chemokines have been shown to regulate hematopoietic progenitor cell proliferation, we hypothesized that chemokines would regulate neural progenitor cell (NPC) proliferation as well. Here we show that chemokines activating CXCR4 or CCR3 reversibly inhibit NPC proliferation in isolated cells, neurospheres, and in hippocampal slice cultures. Cells induced into quiescence by chemokines maintain their multipotential ability to form both neurons and astrocytes. The mechanism of chemokine action appears to be a reduction of extracellular signal-related kinase phosphorylation as well as an increase in Reelin expression. The inhibitory effects of chemokines are blocked by heparan sulfate and apolipoprotein E3 but not apolipoprotein E4, suggesting a regulatory role of these molecules on the effects of chemokines. Additionally, we found that the chemokine fractalkine promotes survival of NPCs. In addition to their role in chemotaxis, chemokines affect both the survival and proliferation of human NPCs in vitro. The presence of constitutively expressed chemokines in the brain argues that under homeostatic conditions, chemokines promote survival but maintain NPCs in a quiescent state. Our studies also suggest a link between inflammatory chemokine production and the inhibition of neurogenesis.


Subject(s)
Cell Culture Techniques/methods , Cell Division/drug effects , Cell Survival/drug effects , Chemokines/pharmacology , Neurons/drug effects , Stem Cells/drug effects , Apolipoprotein E3 , Apolipoproteins E/metabolism , Apolipoproteins E/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/immunology , Cell Adhesion Molecules, Neuronal/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cell Division/immunology , Cell Line , Cell Survival/immunology , Chemokine CX3CL1 , Chemokines/immunology , Chemokines, CX3C/immunology , Chemokines, CX3C/pharmacology , Encephalitis/immunology , Extracellular Matrix Proteins/metabolism , Fetus , Heparitin Sulfate/metabolism , Heparitin Sulfate/pharmacology , Humans , Membrane Proteins/immunology , Membrane Proteins/pharmacology , Nerve Tissue Proteins , Neurons/cytology , Neurons/immunology , Phosphorylation/drug effects , Receptors, CCR3 , Receptors, CXCR4/drug effects , Receptors, CXCR4/immunology , Receptors, Chemokine/drug effects , Receptors, Chemokine/immunology , Reelin Protein , Serine Endopeptidases , Stem Cells/cytology , Stem Cells/immunology
17.
Circulation ; 108(20): 2498-504, 2003 Nov 18.
Article in English | MEDLINE | ID: mdl-14581400

ABSTRACT

BACKGROUND: Chemokines are important mediators of inflammatory cell recruitment that play a significant role in atherosclerosis. Fractalkine (CX3CL1) is an unusual membrane-bound chemokine that mediates chemotaxis through the CX3CR1 receptor. Recently, functional polymorphisms in the human CX3CR1 gene have been described that are associated with coronary artery disease. METHODS AND RESULTS: We investigated the expression of the CX3C chemokine fractalkine and its receptor CX3CR1 in human coronary artery plaques by immunocytometry. We show that a subset of mononuclear cells expresses high levels of fractalkine in human coronary atherosclerotic plaques and that smooth muscle cells within the neointima express the fractalkine receptor CX3CR1. There is a positive correlation between the number of fractalkine-expressing cells and the number of CX3CR1-positive cells in human atherosclerotic plaques (r=0.70, n=15 plaques). Furthermore, we demonstrate that cultured vascular smooth muscle cells express the CX3CR1 receptor and undergo chemotaxis to fractalkine that can be inhibited by G protein inactivation by pertussis toxin. CONCLUSIONS: These results suggest that in human atherosclerosis, fractalkine, rather than mediating inflammatory cell recruitment, can act as a mediator of smooth muscle cell migration.


Subject(s)
Arteriosclerosis/metabolism , Chemokines, CX3C/metabolism , Chemotaxis/physiology , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Adult , Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , Arteriosclerosis/pathology , CD3 Complex/biosynthesis , Cell Count , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/pharmacology , Chemotaxis/drug effects , Female , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Lipopolysaccharide Receptors/biosynthesis , Male , Membrane Proteins/pharmacology , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Pertussis Toxin/pharmacology
18.
Immunobiology ; 207(3): 187-96, 2003.
Article in English | MEDLINE | ID: mdl-12777060

ABSTRACT

Chemokines are important mediators of leukocyte recruitment and activation that play critical roles in the pathology of inflammatory diseases such as atherosclerosis, rheumatoid arthritis and asthma. The vaccinia virus (strain Lister) expresses a 35 kDa soluble protein ('35K') that binds and inactivates a wide range of CC chemokines. We generated a recombinant adenovirus encoding soluble 35K (Ad35K). Ad35K-infected cell culture medium, containing recombinant 35K, potently reduced migration of CCR5-transfected 293 cells by 95% in response to the CC-chemokine RANTES, but had no effect on cells transfected with the CX3CR1 fractalkine receptor. Delivery of Ad35K to mice in vivo via tail vein injection resulted in expression of recombinant 35K in plasma and increased serum RANTES and MIP-1alpha levels when quantified by ELISA. However, chemotaxis of both CCR5-transfected cells and primary macrophages was inhibited by more than 90% by plasma from Ad35K-infected animals compared with control plasma from animals injected with AdGFP. Furthermore, 35K delivered by intra-peritoneal injection more than halved biogel-induced inflammatory cell recruitment in peritoneal exudates compared to AdGFP medium. These studies identify broad-spectrum CC-chemokine blockade using in vivo adenoviral-mediated recombinant 35K expression as a promising strategy to reduce local and systemic inflammation.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Receptors, Interleukin-8A/genetics , Animals , Blotting, Western , Cell Line , Cell Movement , Chemokine CCL5/pharmacology , Chemokine CX3CL1 , Chemokines, CX3C/pharmacology , Cloning, Molecular , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Green Fluorescent Proteins , Humans , Inflammation , Luminescent Proteins/metabolism , Macrophages/metabolism , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Receptors, CCR5/chemistry , Transfection
19.
Kidney Int ; 63(2): 474-86, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12631113

ABSTRACT

BACKGROUND: Fractalkine is a CX3C chemokine for mononuclear cells that has been implicated in the recruitment and accumulation of monocytes seen in glomerular diseases. We investigated the mechanisms by which tumor necrosis factor (TNF)-alpha stimulates mesangial cell (MC) fractalkine expression, and the effects of MC-derived fractalkine on monocyte transmigration. METHODS: Cultured rat MCs were incubated with TNF-alpha, with or without pretreatment with pharmacologic inhibitors of protein kinases or transcriptional factors downstream to TNF-alpha. Fractalkine mRNA and protein were analyzed by Northern and Western blotting. Translocation of nuclear factor (NF)-kappaB was evaluated by immunocytochemical staining. Monocyte transmigration was determined by in vitro chemotaxis assay. RESULTS: TNF-alpha stimulated MC fractalkine mRNA as well as cell-bound and soluble protein expression in a dose- and time-dependent manner. The soluble fractalkine was shed from the cell-bound form via metalloproteinase-dependent cleavage, and mediated in part TNF-alpha-induced monocyte transmigration in vitro. The incubation of MCs with calphostin C [a selective inhibitor of protein kinase C (PKC)] or PD98059 [a selective inhibitor of p42/44 mitogen-activated protein kinase (MAPK) kinase] attenuated TNF-alpha-stimulated fractalkine mRNA and protein expression. Coincubation of MCs with calphostin C and PD98059 resulted in a synergistic inhibition of TNF-alpha-stimulated fractalkine mRNA and protein expression. Incubation of MCs with phorbol myristate acetate (PMA) for four hours resulted in an increase in fractalkine mRNA expression that could be suppressed by calphostin C or depletion of PKC by pretreatment with PMA for 24 hours. Further, activation of PKC-depleted MCs with TNF-alpha stimulated fractalkine mRNA expression that could be blocked by calphostin C. PD 98059, but not calphostin C, inhibited TNF-alpha-activated phospho-p42/44 MAPK and phospho-c-Jun levels, whereas only calphostin C inhibited TNF-alpha-activated phosphorylation of PKCzeta/iota. The incubation of MCs with MG132, a NF-kappaB inhibitor, abolished TNF-alpha-induced degradation of inhibitory protein of NF-kappaB (I-kappaB)alpha, nuclear translocation of NF-kappaB, and fractalkine expression, without affecting phospho-c-Jun levels. In contrast, curcumin, an activating protein (AP)-1 inhibitor, attenuated TNF-alpha-stimulated phospho-c-Jun levels and fractalkine expression without discernible effects on TNF-alpha-induced degradation of I-kappaBalpha or NF-kappaB nuclear translocation. Neither PD 98059 nor calphostin C affected TNF-alpha-induced degradation of I-kappaBalpha or NF-kappaB nuclear translocation. Additional experiments examining the role of cAMP on MC fractalkine expression showed that the incubation of MCs with TNF-alpha and either db-cAMP or forskolin attenuated TNF-alpha-stimulated fractalkine mRNA and protein expression, preceded by attenuation of TNF-alpha-activated phosphorylation of p42/44 MAPK, and c-Jun, but not phosphorylation of PKCzeta/iota or nuclear translocation of NF-kappaB. CONCLUSION: The present data indicate that TNF-alpha activation of PKCzeta/iota, p42/44 MAPK, c-Jun/AP-1, and p65/NF-kappaB are involved in TNF-alpha-stimulated MC fractalkine expression, with the soluble fractalkine mediating in part the TNF-alpha-induced monocyte transmigration in vitro. Uncoupling of p42/44 MAPK or c-Jun/AP-1 signals may contribute to cAMP inhibition of MC fractalkine expression activated by TNF-alpha.


Subject(s)
Chemokines, CX3C/biosynthesis , Glomerular Mesangium/metabolism , Membrane Proteins/biosynthesis , Monocytes/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Movement/drug effects , Cells, Cultured , Chemokine CX3CL1 , Chemokines, CX3C/chemistry , Chemokines, CX3C/genetics , Chemokines, CX3C/pharmacology , Cyclic AMP/physiology , Down-Regulation , Glomerular Mesangium/cytology , Intracellular Membranes/physiology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Metalloproteases/physiology , Monocytes/drug effects , Monocytes/metabolism , RNA, Messenger/metabolism , Rats , Recombinant Proteins/pharmacology , Signal Transduction , Solubility
20.
J Biol Chem ; 278(21): 19473-82, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12646575

ABSTRACT

US28 is one of four 7 transmembrane (7TM) chemokine receptors encoded by human cytomegalovirus and has been shown to both signal and endocytose in a ligand-independent, constitutively active manner. Here we show that the constitutive activity and constitutive endocytosis properties of US28 are separable entities in this viral chemokine receptor. We generated chimeric and mutant US28 proteins that were altered in either their constitutive endocytic (US28 Delta 300, US28 Delta 317, US28-NK1-ctail, and US28-ORF74-ctail) or signaling properties (US28R129A). By using this series of mutants, we show that the cytoplasmic tail domain of US28 per se regulates receptor endocytosis, independent of the signaling ability of the core domain of US28. The constitutive endocytic property of the US28 c-tail was transposable to other 7TM receptors, the herpes virus 8-encoded ORF74 and the tachykinin NK1 receptor (ORF74-US28-ctail and NK1-US28-ctail). Deletion of the US28 C terminus resulted in reduced constitutive endocytosis and consequently enhanced signaling capacity of all receptors tested as assessed by inositol phosphate turnover, NF-kappa B, and cAMP-responsive element-binding protein transcription assays. We further show that the constitutive endocytic property of US28 affects the action of its chemokine ligand fractalkine/CX3CL1 and show that in the absence of the US28 C terminus, fractalkine/CX3CL1 acts as an agonist on US28. This demonstrates for the first time that the endocytic properties of a 7TM receptor can camouflage the agonist properties of a ligand.


Subject(s)
Endocytosis/physiology , Receptors, Chemokine/physiology , Viral Proteins/physiology , Amino Acid Sequence , Animals , COS Cells , Chemokine CX3CL1 , Chemokines, CX3C/pharmacology , Cyclic AMP Response Element-Binding Protein/genetics , Cytoplasm/chemistry , Gene Deletion , Gene Expression , Inositol Phosphates/metabolism , Iodine Radioisotopes , Membrane Proteins/pharmacology , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-kappa B/genetics , Radioligand Assay , Receptors, Chemokine/chemistry , Receptors, Chemokine/genetics , Receptors, Neurokinin-1/genetics , Recombinant Fusion Proteins , Signal Transduction , Structure-Activity Relationship , Transcription, Genetic , Transfection , Viral Proteins/chemistry , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...