Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Circ Cardiovasc Interv ; 5(6): 821-30, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23192920

ABSTRACT

BACKGROUND: Critical limb ischemia portends a risk of major amputation of 25% to 35% within 1 year of diagnosis. Preclinical studies provide evidence that intramuscular injection of autologous CD34+ cells improves limb perfusion and reduces amputation risk. In this randomized, double-blind, placebo-controlled pilot study, we evaluated the safety and efficacy of intramuscular injections of autologous CD34+ cells in subjects with moderate or high-risk critical limb ischemia, who were poor or noncandidates for surgical or percutaneous revascularization (ACT34-CLI). METHODS AND RESULTS: Twenty-eight critical limb ischemia subjects were randomized and treated: 7 to 1 × 10(5) (low-dose) and 9 to 1 × 10(6) (high-dose) autologous CD34+ cells/kg; and 12 to placebo (control). Intramuscular injections were distributed into 8 sites within the ischemic lower extremity. At 6 months postinjection, 67% of control subjects experienced a major or minor amputation versus 43% of low-dose and 22% of high-dose cell-treated subjects (P=0.137). This trend continued at 12 months, with 75% of control subjects experiencing any amputation versus 43% of low-dose and 22% of high-dose cell-treated subjects (P=0.058). Amputation incidence was lower in the combined cell-treated groups compared with control group (6 months: P=0.125; 12 months: P=0.054), with the low-dose and high-dose groups individually showing trends toward improved amputation-free survival at 6 months and 12 months. No adverse safety signal was associated with cell administration. CONCLUSIONS: This study provides evidence that intramuscular administration of autologous CD34+ cells was safe in this patient population. Favorable trends toward reduced amputation rates in cell-treated versus control subjects were observed. These findings warrant further exploration in later-phase clinical trials. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT00616980.


Subject(s)
Antigens, CD34/analysis , Ischemia/surgery , Lower Extremity/blood supply , Stem Cell Transplantation , Stem Cells/immunology , Aged , Aged, 80 and over , Amputation, Surgical , Analysis of Variance , Biomarkers/analysis , Critical Illness , Disease-Free Survival , Double-Blind Method , Female , Humans , Injections, Intramuscular , Ischemia/physiopathology , Limb Salvage , Male , Middle Aged , Pilot Projects , Prospective Studies , Quality of Life , Recovery of Function , Stem Cell Transplantation/adverse effects , Time Factors , Transplantation, Autologous , Treatment Outcome , United States , Wound Healing
2.
Am Heart J ; 164(5): 689-697.e3, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23137499

ABSTRACT

BACKGROUND: Cell therapy is a promising therapeutic for a variety of cardiovascular conditions including refractory angina. Elevation of cardiac biomarkers during cell delivery has been frequently described, but the clinical implications have never been studied. METHODS: ACT34-CMI was a randomized double-blind study assessing the use of intramyocardial delivery of autologous CD34(+) cells for the treatment of refractory angina. Patients (n = 167) underwent G-CSF-mediated (5 µg/[kg day] × 5 days) stem cell mobilization, apheresis, and intramyocardial injection of 1 × 10(5)/kg or 5 × 10(5)/kg CD34(+) cells or placebo. Troponin and creatinine kinase MB were assessed at baseline (n = 161), after cell mobilization and apheresis (n = 153 and 143, respectively), and post-intramyocardial injection (n = 155 and 141, respectively). Major adverse cardiac events (MACE) included death, myocardial infarction, acute congestive heart failure, urgent revascularization, or sustained ventricular arrhythmia. RESULTS: Seven (4.3%) subjects had troponin above the upper limits of normal (ULN) at baseline. Thirty-four (22.2%) and 11 (7.2%) subjects had troponin levels > ULN or >3× ULN after cell mobilization and apheresis, whereas 72 (46.1%) and 39 (25.2%) subjects had troponin elevations > ULN or >3× ULN, respectively, after intramyocardial injections. Age, but no other preprocedural factors, was predictive of troponin elevation. Periprocedural troponin elevation was not associated with an increased risk of MACE during 1 year, especially in cell therapy-treated patients. CONCLUSIONS: Troponin elevation is common during stem cell harvesting and intramyocardial administration, is usually asymptomatic, and does not appear to be associated with long-term MACE in subjects undergoing stem cell mobilization and intramyocardial injection.


Subject(s)
Angina Pectoris/therapy , Antigens, CD34 , Blood Component Removal , Creatinine/blood , Stem Cell Transplantation/adverse effects , T-Lymphocytes , Troponin I/blood , Adult , Aged , Aged, 80 and over , Angina Pectoris/blood , Biomarkers/blood , Coronary Disease/epidemiology , Coronary Disease/etiology , Female , Follow-Up Studies , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Incidence , Logistic Models , Male , Middle Aged , Myocardium , Randomized Controlled Trials as Topic , Transplantation, Autologous
3.
Mol Brain ; 4: 32, 2011 Aug 10.
Article in English | MEDLINE | ID: mdl-21827709

ABSTRACT

Protease activated receptor-1 (PAR1) is expressed in multiple cell types in the CNS, with the most prominent expression in glial cells. PAR1 activation enhances excitatory synaptic transmission secondary to the release of glutamate from astrocytes following activation of astrocytically-expressed PAR1. In addition, PAR1 activation exacerbates neuronal damage in multiple in vivo models of brain injury in a manner that is dependent on NMDA receptors. In the hippocampal formation, PAR1 mRNA appears to be expressed by a subset of neurons, including granule cells in the dentate gyrus. In this study we investigate the role of PAR activation in controlling neuronal excitability of dentate granule cells. We confirm that PAR1 protein is expressed in neurons of the dentate cell body layer as well as in astrocytes throughout the dentate. Activation of PAR1 receptors by the selective peptide agonist TFLLR increased the intracellular Ca2+ concentration in a subset of acutely dissociated dentate neurons as well as non-neuronal cells. Bath application of TFLLR in acute hippocampal slices depolarized the dentate gyrus, including the hilar region in wild type but not in the PAR1-/- mice. PAR1 activation increased the frequency of action potential generation in a subset of dentate granule neurons; cells in which PAR1 activation triggered action potentials showed a significant depolarization. The activation of PAR1 by thrombin increased the amplitude of NMDA receptor-mediated component of EPSPs. These data suggest that activation of PAR1 during normal function or pathological conditions, such as during ischemia or hemorrhage, can increase the excitability of dentate granule cells.


Subject(s)
Action Potentials/physiology , Dentate Gyrus/cytology , Hippocampus/cytology , Neurons/physiology , Receptor, PAR-1/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptor, PAR-1/genetics , Synaptic Transmission/physiology
4.
J Biol Chem ; 284(19): 12862-73, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19240037

ABSTRACT

Zinc is hypothesized to be co-released with glutamate at synapses of the central nervous system. Zinc binds to NR1/NR2A N-methyl-d-aspartate (NMDA) receptors with high affinity and inhibits NMDAR function in a voltage-independent manner. The serine protease plasmin can cleave a number of substrates, including protease-activated receptors, and may play an important role in several disorders of the central nervous system, including ischemia and spinal cord injury. Here, we demonstrate that plasmin can cleave the native NR2A amino-terminal domain (NR2A(ATD)), removing the functional high affinity Zn(2+) binding site. Plasmin also cleaves recombinant NR2A(ATD) at lysine 317 (Lys(317)), thereby producing a approximately 40-kDa fragment, consistent with plasmin-induced NR2A cleavage fragments observed in rat brain membrane preparations. A homology model of the NR2A(ATD) predicts that Lys(317) is near the surface of the protein and is accessible to plasmin. Recombinant expression of NR2A with an amino-terminal deletion at Lys(317) is functional and Zn(2+) insensitive. Whole cell voltage-clamp recordings show that Zn(2+) inhibition of agonist-evoked NMDA receptor currents of NR1/NR2A-transfected HEK 293 cells and cultured cortical neurons is significantly reduced by plasmin treatment. Mutating the plasmin cleavage site Lys(317) on NR2A to alanine blocks the effect of plasmin on Zn(2+) inhibition. The relief of Zn(2+) inhibition by plasmin occurs in PAR1(-/-) cortical neurons and thus is independent of interaction with protease-activated receptors. These results suggest that plasmin can directly interact with NMDA receptors, and plasmin may increase NMDA receptor responses through disruption or removal of the amino-terminal domain and relief of Zn(2+) inhibition.


Subject(s)
Fibrinolysin/pharmacology , Fibrinolytic Agents/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Trace Elements/pharmacology , Zinc/pharmacology , Amino Acid Sequence , Animals , Brain/cytology , Brain/drug effects , Brain/metabolism , Humans , Immunoblotting , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Protein Conformation , Protein Subunits , Rats , Receptor, PAR-1/physiology , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Xenopus laevis
5.
J Biol Chem ; 283(29): 20600-11, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18474593

ABSTRACT

Protease-activated receptor-1 (PAR1) is activated by a number of serine proteases, including plasmin. Both PAR1 and plasminogen, the precursor of plasmin, are expressed in the central nervous system. In this study we examined the effects of plasmin in astrocyte and neuronal cultures as well as in hippocampal slices. We find that plasmin evokes an increase in both phosphoinositide hydrolysis (EC(50) 64 nm) and Fura-2/AM fluorescence (195 +/- 6.7% above base line, EC(50) 65 nm) in cortical cultured murine astrocytes. Plasmin also activates extracellular signal-regulated kinase (ERK1/2) within cultured astrocytes. The plasmin-induced rise in intracellular Ca(2+) concentration ([Ca(2+)](i)) and the increase in phospho-ERK1/2 levels were diminished in PAR1(-/-) astrocytes and were blocked by 1 microm BMS-200261, a selective PAR1 antagonist. However, plasmin had no detectable effect on ERK1/2 or [Ca(2+)](i) signaling in primary cultured hippocampal neurons or in CA1 pyramidal cells in hippocampal slices. Plasmin (100-200 nm) application potentiated the N-methyl-D-aspartate (NMDA) receptor-dependent component of miniature excitatory postsynaptic currents recorded from CA1 pyramidal neurons but had no effect on alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionate- or gamma-aminobutyric acid receptor-mediated synaptic currents. Plasmin also increased NMDA-induced whole cell receptor currents recorded from CA1 pyramidal cells (2.5 +/- 0.3-fold potentiation over control). This effect was blocked by BMS-200261 (1 microm; 1.02 +/- 0.09-fold potentiation over control). These data suggest that plasmin may serve as an endogenous PAR1 activator that can increase [Ca(2+)](i) in astrocytes and potentiate NMDA receptor synaptic currents in CA1 pyramidal neurons.


Subject(s)
Fibrinolysin/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Receptor, PAR-1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Calcium/metabolism , Cells, Cultured , Enzyme Activation , Humans , Magnesium/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Receptor, PAR-1/deficiency , Receptor, PAR-1/genetics , Signal Transduction/drug effects
6.
Exp Neurol ; 188(1): 94-103, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15191806

ABSTRACT

Protease-activated receptor-1 (PAR1) is a G-protein coupled receptor that is proteolytically activated by blood-derived serine proteases. Although PAR1 is best known for its role in coagulation and hemostasis, recent findings demonstrate that PAR1 activation has actions in the central nervous system (CNS) apart from its role in the vasculature. Rodent studies have demonstrated that PAR1 is expressed throughout the brain on neurons and astrocytes. PAR1 activation in vitro and in vivo appears to influence neurodegeneration and neuroprotection in animal models of stroke and brain injury. Because of increasing evidence that PAR1 has important and diverse roles in the CNS, we explored the protein localization and function of PAR1 in human brain. PAR1 is most intensely expressed in astrocytes of white and gray matter and moderately expressed in neurons. PAR1 and GFAP co-localization demonstrates that PAR1 is expressed on the cell body and on astrocytic endfeet that invest capillaries. PAR1 activation in the U178MG human glioblastoma cell line increased PI hydrolysis and intracellular Ca(2+), indicating that PAR1 is functional in human glial-derived tumor cells. Primary cultures of human astrocytes and human glioblastoma cells respond to PAR1 activation by increasing intracellular Ca(2+). Together, these results demonstrate that PAR1 is expressed in human brain and functional in glial tumors and cultures derived from it. Because serine proteases may enter brain tissue and activate PAR1 when the blood brain barrier (BBB) breaks down, pharmacological manipulation of PAR1 signaling may provide a potential therapeutic target for neuroprotection in human neurological disorders.


Subject(s)
Astrocytes/metabolism , Brain Neoplasms/metabolism , Brain/metabolism , Glioblastoma/metabolism , Glioma/metabolism , Receptor, PAR-1/metabolism , Serine Endopeptidases/metabolism , Astrocytes/cytology , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/cytology , Brain Neoplasms/pathology , Calcium/metabolism , Calcium Signaling/physiology , Capillaries/metabolism , Capillaries/ultrastructure , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/pathology , Glioma/pathology , Humans , Immunohistochemistry , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/ultrastructure , Neurons/cytology , Neurons/metabolism , Phosphatidylinositols/metabolism , Tumor Cells, Cultured
7.
Proc Natl Acad Sci U S A ; 100(22): 13019-24, 2003 Oct 28.
Article in English | MEDLINE | ID: mdl-14559973

ABSTRACT

The serine proteases tissue plasminogen activator, plasmin, and thrombin and their receptors have previously been suggested to contribute to neuronal damage in certain pathological situations. Here we demonstrate that mice lacking protease-activated receptor 1 (PAR1) have a 3.1-fold reduction in infarct volume after transient focal cerebral ischemia. Intracerebroventricular injection of PAR1 antagonist BMS-200261 reduced infarct volume 2.7-fold. There are no detectable differences between PAR1-/- and WT mice in cerebrovascular anatomy, capillary density, or capillary diameter, demonstrating that the neuroprotective phenotype is not likely related to congenital abnormalities in vascular development. We also show that the exogenously applied serine proteases thrombin, plasmin, and tissue plasminogen activator can activate PAR1 signaling in brain tissue. These data together suggest that if blood-derived serine proteases that enter brain tissue in ischemic situations can activate PAR1, this sequence of events may contribute to the harmful effects observed. Furthermore, PAR1 immunoreactivity is present in human brain, suggesting that inhibition of PAR1 may provide a novel potential therapeutic strategy for decreasing neuronal damage associated with ischemia and blood-brain barrier breakdown.


Subject(s)
Guanidines/pharmacology , Ischemic Attack, Transient/pathology , Neurons/pathology , Oligopeptides/pharmacology , Receptor, PAR-1/physiology , Animals , Cerebrovascular Circulation/physiology , Disease Models, Animal , Fibrinolysin/metabolism , Guanidines/administration & dosage , Hippocampus/metabolism , Hippocampus/pathology , Humans , Hydrolysis , Injections, Intra-Articular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligopeptides/administration & dosage , Phosphatidylinositols/metabolism , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/deficiency , Receptor, PAR-1/genetics , Thrombin/metabolism , Tissue Plasminogen Activator/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...