Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
Proc Natl Acad Sci U S A ; 119(15): e2116097119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35377786

ABSTRACT

Confining the activity of a designed protein to a specific microenvironment would have broad-ranging applications, such as enabling cell type-specific therapeutic action by enzymes while avoiding off-target effects. While many natural enzymes are synthesized as inactive zymogens that can be activated by proteolysis, it has been challenging to redesign any chosen enzyme to be similarly stimulus responsive. Here, we develop a massively parallel computational design, screening, and next-generation sequencing-based approach for proenzyme design. For a model system, we employ carboxypeptidase G2 (CPG2), a clinically approved enzyme that has applications in both the treatment of cancer and controlling drug toxicity. Detailed kinetic characterization of the most effectively designed variants shows that they are inhibited by ∼80% compared to the unmodified protein, and their activity is fully restored following incubation with site-specific proteases. Introducing disulfide bonds between the pro- and catalytic domains based on the design models increases the degree of inhibition to 98% but decreases the degree of restoration of activity by proteolysis. A selected disulfide-containing proenzyme exhibits significantly lower activity relative to the fully activated enzyme when evaluated in cell culture. Structural and thermodynamic characterization provides detailed insights into the prodomain binding and inhibition mechanisms. The described methodology is general and could enable the design of a variety of proproteins with precise spatial regulation.


Subject(s)
Computer-Aided Design , Drug Design , Enzyme Precursors , Protein Engineering , gamma-Glutamyl Hydrolase , Catalytic Domain , Drug Design/methods , Enzyme Precursors/chemistry , Enzyme Precursors/pharmacology , Humans , PC-3 Cells , Protein Engineering/methods , gamma-Glutamyl Hydrolase/chemistry , gamma-Glutamyl Hydrolase/pharmacology
2.
Int J Biol Macromol ; 113: 996-1007, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29524494

ABSTRACT

Prophenoloxidase is a conserved Cu-containing enzyme acting as a major defense molecule in the immune response of crustaceans. In the present research, we purified prophenoloxidase from the haemolymph of Portunus pelagicus (Pp-proPO) by Blue Sepharose CL-6B chromatography. Pp-proPO exhibited only one band with molecular weight of 75kDa on SDS-PAGE. The purified Pp-proPO was characterized through X-ray diffraction (XRD) and high-performance liquid chromatography (HPLC). Pp-proPO showed phagocytic activity on the yeast Saccharomyces cerevisiae as well as encapsulation on sepharose CL-6B beads associated with CM sepharose and beads of sodium alginate. Pp-proPO also led to strong agglutination on human erythrocytes. Furthermore, Pp-proPO showed magnified PO activity when altered with activated particles acting as pathogen combined molecular patterns (PAMPs), metal ions or other chemicals. Pp-proPO showed relevant antibiofilm activity on Gram negative bacteria Pseudomonas aeruginosa and Escherichia coli. Overall, the above results allowed us to claim that Pp-proPO play a key role in immune defense mechanisms of P. pelagicus crabs, in particular towards microbial pathogens; notably we added basic information to the functional characterization of Pp-proPO, as well as to understand its immunological role in crustaceans defense systems.


Subject(s)
Biofilms/drug effects , Brachyura/immunology , Catechol Oxidase/immunology , Catechol Oxidase/pharmacology , Enzyme Precursors/immunology , Enzyme Precursors/pharmacology , Animals , Biofilms/growth & development , Brachyura/enzymology , Catechol Oxidase/chemistry , Enzyme Precursors/chemistry , Hemagglutination , Hydrophobic and Hydrophilic Interactions , Phagocytosis
3.
Biomol Concepts ; 9(1): 169-175, 2018 Dec 26.
Article in English | MEDLINE | ID: mdl-30864392

ABSTRACT

Meizothrombin is an active intermediate generated during the proteolytic activation of prothrombin to thrombin in the penultimate step of the coagulation cascade. Structurally, meizothrombin differs from thrombin because it retains the auxiliary Gla domain and two kringles. Functionally, meizothrombin shares with thrombin the ability to cleave procoagulant (fibrinogen), prothrombotic (PAR1) and anticoagulant (protein C) substrates, although its specificity toward fibrinogen and PAR1 is less pronounced. In this study we report information on the structural architecture of meizothrombin resolved by SAXS and single molecule FRET as an elongated arrangement of its individual domains. In addition, we show the properties of a meizothrombin construct analogous to the anticoagulant thrombin mutant W215A/E217A currently in Phase I for the treatment of thrombotic complications and stroke. The findings reveal new structural and functional aspects of meizothrombin that advance our understanding of a key intermediate of the prothrombin activation pathway.


Subject(s)
Anticoagulants/chemistry , Enzyme Precursors/chemistry , Thrombin/chemistry , Amino Acid Substitution , Animals , Anticoagulants/pharmacology , Cell Line , Cricetinae , Cricetulus , Enzyme Precursors/pharmacology , Fibrinogen/metabolism , Protein C/metabolism , Protein Domains , Proteolysis/drug effects , Thrombin/pharmacology
4.
Eur J Pharmacol ; 818: 429-434, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29154937

ABSTRACT

We evaluated the efficacy and safety of human recombinant prourokinase ( rhpro-UK) on thromboembolic stroke in rats. 60 rats with thromboembolic stroke were divided into 6 groups (n = 10). The model group was given saline, the reagent groups were given rhpro-UK (5, 10, 20 × 104U/kg), and positive control groups were given urokinase (UK) 10 × 104U/kg and recombinant tissue plasminogen activator (rt-PA) 9mg/kg through intravenous infusion at 1.5h after embolism. And other 10 rats without occluded by autologous blood clots as the sham group were given saline. At 6h after treatment, neurological deficit score and Magnetic Resonance Imaging(MRI) including T1WI and T2WI sequence scanning were measured. At 24h after treatment, the brain was cut for 2,3,5-triphenyltetrazolium chloride (TTC) staining and aspectrophotometric assay to measure the infarct area and intracerebral hemorrhage after neurological deficit detection. rhpro-UK (5, 10, 20 × 104 U/kg) improved neurological disorder by 39.1 ± 19.7% (n = 10, P > 0.05), 65.2 ± 14.2% (n = 10, P < 0.01) and 65.2 ± 14.2% (n = 10, P < 0.01) maximally; decreased brain lesion volume by 36.7 ± 34.8% (n = 10, P < 0.05), 77.6 ± 7.7% (n = 10, P < 0.01) and 80.5 ± 6.9% (n = 10, P < 0.01); decreased infarction area by 38.2 ± 24.0% (n = 10, P < 0.01), 73.9 ± 5.2% (n = 10, P < 0.001) and 79.7 ± 4.0% (n = 10, P < 0.001) respectively, and there were no statistics difference between rhpro-UK (5, 10, 20 × 104 U/kg) and each positive groups at intracerebral hemorrhage (P > 0.05). Rhpro-UK improved the damaged neural function, decreased the extent of the disease and did not raise bleeding, had protective effects for cerebral ischemia in rats.


Subject(s)
Enzyme Precursors/pharmacology , Recombinant Proteins/pharmacology , Stroke/complications , Stroke/drug therapy , Thromboembolism/complications , Urokinase-Type Plasminogen Activator/pharmacology , Animals , Cerebral Hemorrhage/complications , Enzyme Precursors/therapeutic use , Humans , Male , Rats , Rats, Sprague-Dawley , Recombinant Proteins/therapeutic use , Stroke/pathology , Urokinase-Type Plasminogen Activator/therapeutic use
5.
Am J Physiol Heart Circ Physiol ; 313(3): H548-H557, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28626074

ABSTRACT

Recent studies have supported an important contribution of prorenin (PR) and its receptor (PRR) to the regulation of hypothalamic, sympathetic, and neurosecretory outflows to the cardiovascular system, including systemic release of vasopressin (VP), both under physiological and cardiovascular disease conditions. Still, the identification of precise cellular mechanisms and neuronal/molecular targets remain unknown. We have recently shown that PRR is expressed in VP neurons and that their activation increases neuronal activity. However, the underlying ionic channel mechanisms are undefined. Here, we performed patch-clamp electrophysiology from identified VP neurons in acute hypothalamic slices obtained from enhanced green fluorescent protein-VP transgenic rats. Voltage-clamp recordings showed that PR inhibited the magnitude of A-type K+ current (IA; ~50% at -25 mV), a subthreshold voltage-dependent current that restrains VP firing activity. PR also increased the inactivation rate of IA and shifted the steady-state voltage-dependent inactivation function toward more hyperpolarized membrane potential (~7 mV shift), thus resulting in less channel availability to be activated at any given membrane potential. PR also inhibited a sustained component of IA ("window" current). PR-mediated changes in action potential waveform and increased firing activity were occluded when IA was blocked by 4-aminopyridine. Finally, PR failed to increase superoxide production within the supraoptic nucleus/paraventricular nucleus, and PR excitatory effects persisted in slices treated with the SOD mimetic tempol. Taken together, these experiments indicated that PR excitatory effects on vasopressin neurons involve inhibition of IA, due, in part, to increases in its voltage-dependent inactivation properties. Moreover, our results indicate that PR effects did not involve an increase in oxidative stress.NEW & NOTEWORTHY Here, we demonstrate that prorenin/the prorenin receptor is an important signaling unit for the regulation of vasopressin firing activity and, thus, systemic hormonal release. We identified A-type K+ channels as key molecular targets mediating prorenin stimulation of vasopressin neuronal activity, thus standing as a potential therapeutic target for neurohumoral activation in cardiovascular disease.


Subject(s)
Enzyme Precursors/pharmacology , Hypothalamus/drug effects , Neurons/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Renin/pharmacology , Vasopressins/metabolism , Action Potentials , Animals , Genotype , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypothalamus/cytology , Hypothalamus/metabolism , In Vitro Techniques , Ion Channel Gating/drug effects , Male , Neurons/metabolism , Neurosecretion , Patch-Clamp Techniques , Phenotype , Potassium Channels, Voltage-Gated/metabolism , Rats, Transgenic , Rats, Wistar , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/metabolism , Time Factors , Vasopressins/genetics
6.
Clin Exp Metastasis ; 34(2): 155-169, 2017 02.
Article in English | MEDLINE | ID: mdl-28124276

ABSTRACT

Thrombin-activatable fibrinolysis inhibitor (TAFI) is a basic carboxypeptidase zymogen present in blood plasma. Proteolytic activation of TAFI by thrombin, thrombin in complex with the endothelial cell cofactor thrombomodulin, or plasmin results in an enzyme (TAFIa) that removes carboxyl-terminal lysine residues from protein and peptide substrates, including cell-surface plasminogen receptors. TAFIa is therefore capable of inhibiting plasminogen activation in the pericellular milieu. Since plasminogen activation has been linked to angiogenesis, TAFIa could therefore have anti-angiogenic properties, and indeed TAFIa has been shown to inhibit endothelial tube formation in a fibrin matrix. In this study, the TAFI pathway was manipulated by providing exogenous TAFI or TAFIa or by adding a potent and specific inhibitor of TAFIa. We found that TAFIa elicited a series of anti-angiogenic responses by endothelial cells, including decreased endothelial cell proliferation, cell invasion, cell migration, tube formation, and collagen degradation. Moreover, TAFIa decreased tube formation and proteolysis in endothelial cell culture grown alone and in co-culture with breast cancer cell lines. In accordance with these findings, inhibition of TAFIa increased secretion of matrix metalloprotease proenzymes by endothelial and breast cancer cells. Finally, treatment of endothelial cells with TAFIa significantly inhibited plasminogen activation. Taken together our results suggest a novel role for TAFI in inhibiting tumour angiogenic behaviors in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Carboxypeptidase B2/physiology , Endothelial Cells/drug effects , Neovascularization, Pathologic/drug therapy , Carboxypeptidase B2/antagonists & inhibitors , Carboxypeptidase B2/pharmacology , Cell Division/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Collagen Type IV/metabolism , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Enzyme Precursors/pharmacology , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Plasminogen/antagonists & inhibitors , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
7.
Fish Shellfish Immunol ; 51: 220-228, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26899630

ABSTRACT

In the present study, we purified two prophenoloxidases (proPO) from haemolymph of green tiger shrimp, Penaeus semisulcatus by gel fermentation chromatography using blue Sepharose matrix. The two purified prophenoloxidase macromolecules are of about 76 and 75 kDa determined through SDS-PAGE and named as Penaeus semisulcatus prophenoloxidase I (PSproPO I) and Penaeus semisulcatus prophenoloxidase II (PSproPO II). It was further characterized by X-Ray Diffraction (XRD), Fourier Transform Infrared Spectroscopy (FTIR), Circular Dichroism (CD) and High Performance Liquid Chromatography (HPLC) analysis. The purified PSproPO I and PSproPO II showed the strongest agglutination titre against human erythrocytes compared to goat RBC. The PSproPO I and PSproPO II showed phagocytic activity against yeast Saccharomyces cerevisiae and encapsulation activity against Sepharose CL 6B beads compared to CM Sepharose and Sodium alginate beads. The functional analysis of purified PSproPO I and PSproPO II showed enhanced PO activity when added with the triggering molecules such as pathogen associated molecular patterns (PAMPs), metals and chemicals. In addition, eluted fraction containing PSproPO I and PSproPO II showed antibiofilm activity against Gram positive and Gram negative bacteria. The above results concluded that no significant differences were found between the purified PSproPO I and PSproPO II immune indices and functions. This study might provide a sensitive platform to understand more about the critical roles of PSproPO I and PSproPO II in crustacean immune system.


Subject(s)
Biofilms/drug effects , Catechol Oxidase/pharmacology , Enzyme Precursors/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Hemolymph/enzymology , Penaeidae , Animals , Catechol Oxidase/isolation & purification , Enzyme Precursors/isolation & purification , Erythrocytes/drug effects , Goats , Gram-Negative Bacteria/physiology , Gram-Positive Bacteria/physiology , Hemagglutination Tests , Humans , Metals/pharmacology , Phagocytosis , Saccharomyces cerevisiae/drug effects
9.
J Dent Res ; 92(12): 1113-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24130220

ABSTRACT

Inferior alveolar nerve (IAN) injury induces persistent ectopic pain which spreads to a wide area in the orofacial region. Its exact mechanism remains unclear. We investigated the involvement of nitric oxide (NO) in relation to ectopic orofacial pain caused by IAN transection (IANX). We assessed the changes in mechanical sensitivity of the whisker pad skin following IANX, neuronal nitric oxide synthase (nNOS) expression in the trigeminal ganglion (TG), and the functional significance of NO in relation to the mechanical allodynia following intra-TG administration of a chemical precursor to NO and selective nNOS inhibitors. IANX induced mechanical allodynia, which was diminished by intra-TG administration of selective nNOS inhibitors. NO metabolites and nNOS immunoreactive neurons innervating the lower lip were also increased in the TG. Intra-TG administration of nNOS substrate induced the mechanical allodynia. The present findings suggest that NO released from TG neurons regulates the excitability of TG neurons innervating the whisker pad skin, and the enhancement of TG neuronal excitability may underlie ectopic mechanical allodynia.


Subject(s)
Facial Pain/etiology , Mandibular Nerve/physiopathology , Neuralgia/etiology , Nitric Oxide/physiology , Signal Transduction/physiology , Trigeminal Nerve Injuries/complications , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Enzyme Inhibitors/pharmacology , Enzyme Precursors/pharmacology , Hyperalgesia/etiology , Indazoles/pharmacology , Lip/innervation , Male , Mandibular Nerve/pathology , Neural Pathways/pathology , Neural Pathways/physiopathology , Neurons/drug effects , Neurons/pathology , Nitric Oxide/analysis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I/analysis , Nitric Oxide Synthase Type I/antagonists & inhibitors , Pain Threshold/physiology , Rats , Rats, Sprague-Dawley , Sensory Thresholds/physiology , Signal Transduction/drug effects , Touch/physiology , Trigeminal Ganglion/drug effects , Trigeminal Ganglion/pathology
10.
Neurosci Lett ; 533: 60-4, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23178475

ABSTRACT

Enterostatin, a gut-brain pentapeptide cleaved from procolipase has been shown to inhibit fat intake in rodents after both peripheral and central administration. In this study, the effect of intracerebroventricular (ICV) injection of enterostatin on fat intake was investigated in neonatal chicks. In Experiment 1, 3-h-fasted chicks fed a low-fat diet were injected with the various doses of enterostatin. Experiment 2 was similar to experiment 1 except that the birds were fasted overnight. In Experiment 3, the 3-h-fasted and in Experiment 4, the overnight fasted chicks adapted to a high-fat diet received different doses of enterostatin. ICV injection of enterostatin caused a dose-dependent increase in high-fat diet intake in 3-h-fasted chicks whereas a decrease in high-fat intake was observed in chicks that were fasted overnight. However, low-fat diet intake was not affected by enterostatin in either 3-h or overnight fasted chicks. These results suggest that enterostatin acts within the brain of chicks to influence fat intake. It appears that in chicks, the eating effect of enterostatin has a biphasic nature similar to those seen in rodents.


Subject(s)
Colipases/physiology , Dietary Fats/administration & dosage , Eating , Enzyme Precursors/physiology , Animals , Animals, Newborn , Chickens , Colipases/pharmacology , Eating/drug effects , Enzyme Precursors/pharmacology , Fasting , Female , Injections, Intraventricular , Male
11.
Vet Microbiol ; 159(3-4): 479-84, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22633172

ABSTRACT

Microsporum canis is a pathogenic fungus that causes a superficial cutaneous infection called dermatophytosis, mainly in cats, dogs and humans. Proteolytic enzymes have been postulated to be key factors involved in the invasion of the stratum corneum and keratinized epidermal structures. Among these proteases, the secreted subtilisin protease Sub3 was found to be required for adherence of M. canis arthroconidia to feline epidermis. This protease is synthetized as a preproenzyme consisting of a signal peptide followed by the propeptide and the protease domain. In order to assess whether the enzymatic activity of Sub3 could be responsible for the role of the protease in the adherence process, we expressed and characterized the propeptide of Sub3 and demonstrated that this propeptide is a strong inhibitor of its mature enzyme. This propeptide acts as a noncompetitive inhibitor with dissociation constants, K(I) and [Formula: see text] of 170 and 130 nM respectively. When tested for its capacity to inhibit adherence of M. canis to feline epidermis using an ex vivo adherence model made of feline epidermis, the propeptide does not prevent adherence of M. canis arthroconidia because it loses its capacity to inhibit rSub3 following a direct contact with living arthroconidia, presumably through inactivation by fungal membrane-bound proteases.


Subject(s)
Cat Diseases/microbiology , Dermatomycoses/veterinary , Enzyme Precursors/pharmacology , Epidermis/microbiology , Microsporum/physiology , Peptide Hydrolases/pharmacology , Animals , Cats , Dermatomycoses/microbiology , Dermatomycoses/pathology , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Epidermis/pathology , Escherichia coli/genetics , In Vitro Techniques , Microsporum/enzymology , Microsporum/pathogenicity , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Subtilisin/chemistry
12.
J Biol Chem ; 285(45): 34803-12, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20826780

ABSTRACT

We recently demonstrated that the Gla domain-dependent interaction of protein C with endothelial protein C receptor (EPCR) leads to dissociation of the receptor from caveolin-1 and recruitment of PAR-1 to a protective signaling pathway. Thus, the activation of PAR-1 by either thrombin or PAR-1 agonist peptide elicited a barrier-protective response if endothelial cells were preincubated with protein C. In this study, we examined whether other vitamin K-dependent coagulation protease zymogens can modulate PAR-dependent signaling responses in endothelial cells. We discovered that the activation of both PAR-1 and PAR-2 in endothelial cells pretreated with factor FX (FX)-S195A, but not other procoagulant protease zymogens, also results in initiation of protective intracellular responses. Interestingly, similar to protein C, FX interaction with endothelial cells leads to dissociation of EPCR from caveolin-1 and recruitment of PAR-1 to a protective pathway. Further studies revealed that, FX activated by factor VIIa on tissue factor bearing endothelial cells also initiates protective signaling responses through the activation of PAR-2 independent of EPCR mobilization. All results could be recapitulated by the receptor agonist peptides to both PAR-1 and PAR-2. These results suggest that a cross-talk between EPCR and an unknown FX/FXa receptor, which does not require interaction with the Gla domain of FX, recruits PAR-1 to protective signaling pathways in endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Enzyme Precursors/metabolism , Factor Xa/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Signal Transduction/physiology , Amino Acid Substitution , Antigens, CD/genetics , Antigens, CD/metabolism , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line , Endothelial Cells/cytology , Endothelial Protein C Receptor , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Precursors/genetics , Enzyme Precursors/pharmacology , Factor VIIa/genetics , Factor VIIa/metabolism , Factor Xa/genetics , Factor Xa/pharmacology , Humans , Mutation, Missense , Protein C/genetics , Protein C/metabolism , Protein C/pharmacology , Protein Structure, Tertiary , Receptor, PAR-1/genetics , Receptor, PAR-2/genetics , Receptors, Cell Surface/agonists , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects
13.
Brain Res ; 1349: 48-55, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20599830

ABSTRACT

Studies have demonstrated defects of DA and 5HT neurotransmission in dietary fat induced obese animals. In the present study, we used a perfusion system to assay the release of DA and 5HT from striatal slices preloaded with [(3)H]-DA or [(3)H]-5HT. The release of both DA and 5HT from striatal slices of rats fed a high fat diet for 10 days, but not 3 days, was reduced when compared to striatal slices taken from rats fed a low fat diet. Enterostatin, an endogenous pentapeptide inhibits dietary fat intake when administered peripherally and centrally in animals. The central mechanism for the action of enterostatin is not yet determined even though several mechanisms have been suggested. We have shown that enterostatin enhanced [(3)H]-DA release, but not [(3)H]-5HT release from striatal slices of rats that had been adapted to high fat diet for 10 days. The enterostatin-induced increase in [(3)H]-DA release was blocked by nomifensine. Enterostatin did not alter [(3)H]-DA or [(3)H]-5HT release from striatal slices of rats adapted to high fat or low fat diet feeding for 3 days. These findings suggest that enterostatin may inhibit dietary fat intake by blocking dopamine reuptake transport to increase central striatal DA release from rats that have acquired diminished dopamine signal after an adaptive period of fat consumption.


Subject(s)
Colipases/pharmacology , Corpus Striatum/drug effects , Dietary Fats/pharmacology , Dopamine/metabolism , Enzyme Precursors/pharmacology , Serotonin/metabolism , Analysis of Variance , Animals , Corpus Striatum/metabolism , Dose-Response Relationship, Drug , In Vitro Techniques , Male , Rats , Rats, Sprague-Dawley , Tritium/metabolism
14.
Blood ; 115(25): 5241-8, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20410503

ABSTRACT

Plasminogen activators (PAs) are used to treat life-threatening thrombosis, but not for thromboprophylaxis because of rapid clearance, risk of bleeding, and central nervous system (CNS) toxicity. We describe a novel strategy that may help to overcome these limitations by targeting a thrombin-activated PA pro-drug to circulating red blood cells (RBCs). We fused a single chain antibody (scFv Ter-119) that binds to mouse glycophorin A (GPA) with a variant human single-chain low molecular weight urokinase construct that can be activated selectively by thrombin (scFv/uPA-T). scFv/uPA-T bound specifically to mouse RBCs without altering their biocompatibility and retained its zymogenic properties until converted by thrombin into an active 2-chain molecule. As a result, RBC-bound scFv/uPA-T caused thrombin-induced fibrinolysis. One hour and 48 hours after intravenous (IV) injection in mice, approximately 70% and approximately 35% of scFv/uPA-T was retained in the blood, respectively, and approximately 95% of the circulating scFv/uPA-T remained bound to RBCs. A single IV injection of scFv/uPA-T provided effective prophylaxis against arterial and venous thrombosis for up to 24 hours. Thus, prophylactic delivery of RBC-targeted PA pro-drugs activated selectively at the site of clot formation represents a new approach to prevent thrombosis in clinical settings where the risk of clotting is high.


Subject(s)
Drug Delivery Systems , Enzyme Precursors/pharmacology , Erythrocytes , Fibrinolytic Agents/pharmacology , Prodrugs/pharmacology , Recombinant Fusion Proteins/pharmacology , Single-Chain Antibodies/pharmacology , Thrombosis/prevention & control , Urokinase-Type Plasminogen Activator/pharmacology , Animals , Humans , Mice , Recombinant Proteins/pharmacology , Time Factors
15.
Adv Exp Med Biol ; 662: 439-45, 2010.
Article in English | MEDLINE | ID: mdl-20204827

ABSTRACT

This is the first case (November 12, 2007) of peri-operative use of zymogen protein C (ZPC) for a heterozygote Protein C deficient (50%) patient with heterozygosity for the prothrombin gene mutation. The surgery involved total left hip replacement. The patient was 74 years of age and at very high-risk for Deep Vein Thrombosis (DVT) thus possible lung emboli. He was a survivor of Venous thrombo-embolism (VTE) in 1999.For this case the need for increased heparin to prevent a reoccurrence of thrombosis led to a further concern of internal bleeding and possible infection. As proposed by the authors in previous publications the use of a ZPC concentrate would decrease the chance of a thrombosis, with minimum or no internal bleeding. ZPC is activated at the endothelium cell surface and on the surface of the platelets where and when it is needed, therefore providing a safer procedure.This protocol was implemented by administering ZPC concentrate 1 day prior to surgery and continuing for 10 days after. Slightly higher than normal blood loss occurred, however, a safe procedure was achieved without dangerous side effects while several additional benefits were experienced (level of PC activity as high as 235% above normal were measured during the procedure.). It is felt that positive effects were the result of increased blood flow and oxygen transport to the tissues by reducing blood sludging in the microcirculation. Further studies on Protein C dose levels will be necessary to provide economical, safe and efficacious use of Protein C products.It is suggested that ZPC should be considered for patients with high probability of VTE which could cause thrombotic occlusion of the pulmonary and peripheral vasculature. These phenomena can result in hypoxia and hypo perfusion causing organ failure and death.Our research includes a focused effort to optimize upstream and downstream bio-processing to produce both zymogen and activated Protein C at a lower cost and to examine the medical indications that could benefit from a more available and affordable Protein C product.


Subject(s)
Arthroplasty, Replacement, Hip/methods , Enzyme Precursors/pharmacology , Protein C/pharmacology , Aged , Humans , Male
16.
J Biol Chem ; 285(11): 8278-89, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20053992

ABSTRACT

Inactivation of thrombin (T) by the serpins heparin cofactor II (HCII) and antithrombin (AT) is accelerated by a heparin template between the serpin and thrombin exosite II. Unlike AT, HCII also uses an allosteric interaction of its NH(2)-terminal segment with exosite I. Sucrose octasulfate (SOS) accelerated thrombin inactivation by HCII but not AT by 2000-fold. SOS bound to two sites on thrombin, with dissociation constants (K(D)) of 10 +/- 4 microm and 400 +/- 300 microm that were not kinetically resolvable, as evidenced by single hyperbolic SOS concentration dependences of the inactivation rate (k(obs)). SOS bound HCII with K(D) 1.45 +/- 0.30 mm, and this binding was tightened in the T.SOS.HCII complex, characterized by K(complex) of approximately 0.20 microm. Inactivation data were incompatible with a model solely depending on HCII.SOS but fit an equilibrium linkage model employing T.SOS binding in the pathway to higher order complex formation. Hirudin-(54-65)(SO(3)(-)) caused a hyperbolic decrease of the inactivation rates, suggesting partial competitive binding of hirudin-(54-65)(SO(3)(-)) and HCII to exosite I. Meizothrombin(des-fragment 1), binding SOS with K(D) = 1600 +/- 300 microm, and thrombin were inactivated at comparable rates, and an exosite II aptamer had no effect on the inactivation, suggesting limited exosite II involvement. SOS accelerated inactivation of meizothrombin 1000-fold, reflecting the contribution of direct exosite I interaction with HCII. Thrombin generation in plasma was suppressed by SOS, both in HCII-dependent and -independent processes. The ex vivo HCII-dependent process may utilize the proposed model and suggests a potential for oversulfated disaccharides in controlling HCII-regulated thrombin generation.


Subject(s)
Anticoagulants/metabolism , Blood Coagulation/physiology , Heparin Cofactor II/metabolism , Sucrose/analogs & derivatives , Thrombin/metabolism , Allosteric Regulation , Anticoagulants/chemistry , Anticoagulants/pharmacology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Blood Coagulation/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Precursors/metabolism , Enzyme Precursors/pharmacology , Heparin Cofactor II/chemistry , Hirudins/metabolism , Hirudins/pharmacology , Humans , Kinetics , Models, Chemical , Plasma , Protease Inhibitors/metabolism , Protease Inhibitors/pharmacology , Prothrombin/metabolism , Spectrometry, Fluorescence , Sucrose/chemistry , Sucrose/metabolism , Sucrose/pharmacology , Thrombin/chemistry , Thrombin/pharmacology
17.
J Drug Target ; 18(2): 115-24, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19751096

ABSTRACT

Novel phage-displayed random linear dodecapeptide (X(12)) and cysteine-constrained decapeptide (CX(10)C) libraries constructed in fusion to the amino-terminus of P99 beta-lactamase molecules were used for identifying beta-lactamase-linked cancer cell-specific ligands. The size and quality of both libraries were comparable to the standards of other reported phage display systems. Using the single-round panning method based on phage DNA recovery, we identified several beta-lactamase fusion peptides that specifically bind to live human breast cancer MDA-MB-361 cells. The beta-lactamase fusion to the peptides helped in conducting the enzyme activity-based clone normalization and cell-binding screening in a very time- and cost-efficient manner. The methods were suitable for 96-well readout as well as microscopic imaging. The success of the biopanning was indicated by the presence of approximately 40% cancer cell-specific clones among recovered phages. One of the binding clones appeared multiple times. The cancer cell-binding fusion peptides also shared several significant motifs. This opens a new way of preparing and selecting phage display libraries. The cancer cell-specific beta-lactamase-linked affinity reagents selected from these libraries can be used for any application that requires a reporter for tracking the ligand molecules. Furthermore, these affinity reagents have also a potential for their direct use in the targeted enzyme prodrug therapy of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Enzyme Precursors/pharmacology , Peptide Library , Prodrugs/pharmacology , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Cloning, Molecular , Consensus Sequence , DNA , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Oligopeptides , Peptides
18.
Surgery ; 147(2): 295-302, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20004432

ABSTRACT

BACKGROUND: Metalloproteinase-9 (MMP-9) is a type IV collagenase found at elevated levels in chronic wounds. As wounds heal, MMP-9 diminishes. In this study, we investigated whether MMP-9 directly contributes to chronic wound pathogenesis. METHODS: Recombinant proMMP-9 was prepared using immortalized keratinocytes transduced by a lentivirus. ProMMP-9 was purified from cell culture media and activated using 4-aminophenylmercuric acetate. Active MMP-9 was then suspended in xanthan gum to a concentration paralleling that found in human chronic wounds. Two parallel 6-mm punch biopsies were made on the backs of C57BL mice. Wounds were treated daily with MMP-9 or vehicle. Wound areas were measured and tissues examined by densitometry, real-time RT-PCR, histology, and immunohistochemistry at days 7, 10, and 12. RESULTS: Exogenous MMP-9, at the level found within chronic wounds, delayed wound healing in this animal model. By 7 days, wounds in the MMP-9-injected group were 12% larger than control wounds (P = .008). By day 12, wounds in the MMP-9-injected group were 25% larger than those of the control group (P = .03). Histologic examination shows that high levels of active MMP-9-impaired epithelial migrating tongues (P = .0008). Moreover, consistent with elevated MMP-9, the collagen IV in the leading edge of the epithelial tongue was diminished. CONCLUSION: MMP-9 appears to directly delay wound healing. Our data suggests that this may occur through interference with re-epithelialization. We propose that MMP-9 interferes with the basement membrane protein structure, which in turn impedes keratinocyte migration, attachment, and the reestablishment of the epidermis.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Skin/injuries , Wound Healing/physiology , Animals , Basement Membrane/drug effects , Basement Membrane/pathology , Blotting, Western , Cell Movement/drug effects , Electrophoresis, Polyacrylamide Gel , Enzyme Precursors/pharmacology , Epithelium/drug effects , Epithelium/pathology , Matrix Metalloproteinase 9/pharmacology , Mice , Mice, Inbred C57BL , Recombinant Proteins/pharmacology , Wound Healing/drug effects
19.
Oncogene ; 28(38): 3390-400, 2009 Sep 24.
Article in English | MEDLINE | ID: mdl-19597471

ABSTRACT

Enhanced RAS signaling and decreased androgen dependence of prostate cancer cells accompany poor clinical outcomes. Elevated autocrine fibroblast growth factors 2 (FGF-2) signaling promotes prostate cancer cell growth and survival. Expression of lysyl oxidase (LOX) inhibits RAS transforming activity. LOX is secreted as 50 kDa pro-LOX protein and then undergoes extracellular proteolytic processing to form approximately 30 kDa LOX enzyme and approximately 18 kDa propeptide (LOX-PP). We have previously shown that LOX-PP inhibits breast cancer cell transformation and tumor formation, but mechanisms of action of LOX-PP have not been fully elucidated. Here we report that LOX expression is reduced in prostate cancer cell lines and that recombinant LOX-PP protein inhibits serum-stimulated DNA synthesis and MEK/ERK and PI3K/AKT pathways in DU 145 and PC-3 androgen-independent cell lines. In DU 145 cells, treatment with a pharmacologic FGF-receptor inhibitor or a neutralizing anti-FGFR1 antibody mimicked LOX-PP inhibition of serum-stimulated DNA synthesis. FGF-2-stimulated DNA synthesis, ERK1/2, AKT and FRS2alpha activation were found all to be inhibited by LOX-PP in DU 145 cells. LOX-PP reduced specific binding of FGF-2 to DU 145 cells, suggesting that LOX-PP targets FGF signaling at the receptor. Interestingly, PC-3 cells did not respond to FGF-2, consistent with previous reports. We conclude that LOX-PP inhibits proliferation of DU 145 cells by interfering with FGFR(s) binding and signaling, and that LOX-PP has other mechanisms of action in PC-3 cells.


Subject(s)
Enzyme Precursors/pharmacology , Fibroblast Growth Factor 2/antagonists & inhibitors , Prostatic Neoplasms/prevention & control , Protein-Lysine 6-Oxidase/pharmacology , Signal Transduction , Cell Line, Tumor , Cell Proliferation , DNA/biosynthesis , Fibroblast Growth Factor 2/metabolism , Humans , Male , Prostate/enzymology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Receptors, Fibroblast Growth Factor/metabolism , Recombinant Proteins/pharmacology
20.
J Biol Chem ; 284(38): 25854-66, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19608737

ABSTRACT

The structural and catalytic requirements for neutrophil MMP-9 proenzyme (proMMP-9) to induce angiogenesis were investigated using a quantitative angiogenesis model based on grafting of collagen onplants onto the chorioallantoic membrane of chick embryos. Both physiological activation of neutrophil proMMP-9 and proteolytic activity of the generated MMP-9 enzyme were critically dependent on the tissue inhibitor of metalloproteinase (TIMP)-free status of the zymogen. The presence of an intact active site and hemopexin domain were required for full angiogenesis-inducing activity of the MMP-9 enzyme. Timed additions of TIMP-1 to the onplants containing TIMP-free neutrophil proMMP-9 indicated that in vivo activation of the zymogen occurred during the first 24 h after grafting. Within the onplant tissue, MMP-9 activation was accompanied by proteolytic modifications of fibrillar collagen and an influx of host proteins, the rate of which depended on the TIMP-free status of the zymogen. By quantifying the levels of host angiogenic factors, we demonstrated that basic fibroblast growth factor (FGF-2) was a major cytokine becoming bioavailable in the onplant tissue undergoing a neutrophil proMMP-9-mediated angiogenic switch. Inhibition of angiogenesis with specific function-blocking antibodies further indicated an involvement of a FGF-2/FGFR-2 pathway in neutrophil proMMP-9-induced angiogenesis. The enhanced angiogenesis catalyzed by neutrophil MMP-9 appears to evoke also a localized, low threshold level vascular endothelial growth factor (VEGF)/VEGFR-2 pathway, likely functioning in the formation and/or stabilization of blood vessels. That neutrophil proMMP-9, unencumbered by TIMP-1, directly mediates FGF-2-dependent angiogenesis was also demonstrated in our quantitative mouse angiogenesis model employing subcutaneous collagen implants, thus implicating the novel TIMP-free MMP-9/FGF-2/FGFR-2 pathway in proMMP-9-induced angiogenesis in a mammalian setting.


Subject(s)
Enzyme Precursors/metabolism , Fibroblast Growth Factor 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Physiologic/physiology , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction/physiology , Tissue Inhibitor of Metalloproteinase-1/metabolism , Animals , Chick Embryo , Enzyme Precursors/genetics , Enzyme Precursors/pharmacology , Fibroblast Growth Factor 2/genetics , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/pharmacology , Mice , Neovascularization, Physiologic/drug effects , Neutrophils/enzymology , Protein Structure, Tertiary/physiology , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...