Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 76
1.
Lancet Neurol ; 20(9): 721-728, 2021 09.
Article En | MEDLINE | ID: mdl-34418399

BACKGROUND: Non-immunogenic staphylokinase is modified recombinant staphylokinase with low immunogenicity, high thrombolytic activity, and selectivity to fibrin. We aimed to assess the safety and efficacy of a single intravenous bolus of non-immunogenic staphylokinase compared with alteplase in patients with acute ischaemic stroke within 4·5 h after symptom onset. METHODS: We did a randomised, open-label, multicentre, parallel-group, non-inferiority trial in 18 clinical sites in Russia. We included patients aged 18 years and older with a diagnosis of acute ischaemic stroke (up to 25 points on the National Institutes of Health Stroke Scale). The study drug had to be administered within 4·5 h after the onset of symptoms. Patients were randomly assigned to receive either non-immunogenic staphylokinase (10 mg) or alteplase (0·9 mg/kg, maximum 90 mg), both administered intravenously. The randomisation sequence was created by an independent biostatistician using computer-generated random numbers. 84 blocks (block size of four) of opaque sealed envelopes were numbered sequentially from 1 to 336 and were opened in numerical order. Patients were unaware of their assigned treatment and were assessed by the study investigators who were also unaware of the treatment assignment on all trial days. Emergency department staff, who administered the assigned drug and opened the envelopes, were not masked to treatment. The primary efficacy endpoint was a favourable outcome, defined as a modified Rankin scale (mRS) score of 0-1 on day 90. The margin of non-inferiority was established as 16% for the difference in mRS score of 0-1 on day 90. Non-inferiority was tested using Welch's t-test for the primary outcome only. Endpoints were analysed in the per-protocol population, which comprised all randomly assigned patients who completed treatment without any protocol violations; this population was identical to the intention-to-treat population. This trial is completed and registered at ClinicalTrials.gov, NCT03151993. FINDINGS: Of 385 patients recruited from March 18, 2017, to March 23, 2019, 336 (87%) were included in the trial. 168 (50%) patients were randomly assigned to receive non-immunogenic staphylokinase and 168 (50%) to receive alteplase. The median duration of follow-up was 89 days (IQR 89-89). 84 (50%) of 168 patients in the non-immunogenic staphylokinase group had a favourable outcome at day 90 compared with 68 (40%) of 168 patients in the alteplase group (odds ratio [OR] 1·47, 95% CI 0·93 to 2·32; p=0·10). The difference in the rate of favourable outcome at day 90 was 9·5% (95% CI -1·7 to 20·7) and the lower limit did not cross the margin of non-inferiority (pnon-inferiority <0·0001). Symptomatic intracranial haemorrhage occurred in five (3%) patients in the non-immunogenic staphylokinase group and in 13 (8%) patients in the alteplase group (p=0·087). On day 90, 17 (10%) patients in the non-immunogenic staphylokinase group and 24 (14%) patients in the alteplase group had died (p=0·32). 22 (13%) patients in the non-immunogenic staphylokinase group had serious adverse events, compared with 37 (22%) patients in the alteplase group (p=0·044). INTERPRETATION: Non-immunogenic staphylokinase was non-inferior to alteplase for patients with acute ischaemic stroke. Mortality, symptomatic intracranial haemorrhage, and serious adverse events did not differ significantly between groups. Future studies are needed to continue to assess the safety and efficacy of non-immunogenic staphylokinase in patients with acute ischaemic stroke within the 4·5 h time window, and to assess the drug in patients with acute ischaemic stroke outside this time window with reperfusion CT or magnetic resonance angiography followed by thrombectomy if necessary. FUNDING: The Russian Academy of Sciences.


Fibrinolytic Agents/pharmacology , Ischemic Stroke/drug therapy , Metalloendopeptidases/pharmacology , Time-to-Treatment , Tissue Plasminogen Activator/pharmacology , Aged , Aged, 80 and over , Double-Blind Method , Female , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/adverse effects , Humans , Male , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/adverse effects , Metalloendopeptidases/immunology , Middle Aged , Outcome Assessment, Health Care , Recombinant Proteins , Russia , Tissue Plasminogen Activator/administration & dosage , Tissue Plasminogen Activator/adverse effects
2.
Commun Biol ; 4(1): 585, 2021 05 14.
Article En | MEDLINE | ID: mdl-33990686

G protein-coupled receptor (GPR)35 is highly expressed in the gastro-intestinal tract, predominantly in colon epithelial cells (CEC), and has been associated with inflammatory bowel diseases (IBD), suggesting a role in gastrointestinal inflammation. The enterotoxigenic Bacteroides fragilis (ETBF) toxin (BFT) is an important virulence factor causing gut inflammation in humans and animal models. We identified that BFT signals through GPR35. Blocking GPR35 function in CECs using the GPR35 antagonist ML145, in conjunction with shRNA knock-down and CRISPRcas-mediated knock-out, resulted in reduced CEC-response to BFT as measured by E-cadherin cleavage, beta-arrestin recruitment and IL-8 secretion. Importantly, GPR35 is required for the rapid onset of ETBF-induced colitis in mouse models. GPR35-deficient mice showed reduced death and disease severity compared to wild-type C57Bl6 mice. Our data support a role for GPR35 in the CEC and mucosal response to BFT and underscore the importance of this molecule for sensing ETBF in the colon.


Bacterial Toxins/administration & dosage , Bacteroides fragilis/pathogenicity , Colitis/pathology , Colon/pathology , Epithelial Cells/pathology , Gastrointestinal Tract/pathology , Metalloendopeptidases/administration & dosage , Receptors, G-Protein-Coupled/physiology , Animals , Bacteroides fragilis/genetics , Bacteroides fragilis/metabolism , Colitis/etiology , Colitis/metabolism , Colon/drug effects , Colon/metabolism , Colon/microbiology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Mice , Mice, Inbred C57BL
3.
Spinal Cord Ser Cases ; 6(1): 9, 2020 02 17.
Article En | MEDLINE | ID: mdl-32066656

STUDY DESIGN: Case series on four dogs. OBJECTIVES: To determine the alleviation of motor symptoms in spinal cord injury (SCI) by tetanus neurotoxin (TeNT). SETTING: Different Berlin veterinary clinics, Germany. METHODS: We report on the effect of intramuscular injections of low-dose TeNT into paretic hind limb muscles 2-157 weeks after SCI due to lumbar disc herniation in a clinical case series on four dogs. All dogs underwent unsuccessful or incomplete surgical decompression prior to TeNT treatment. TeNT was injected on a compassionate basis. Stance, gait ability and the diameter of the rectus femoris muscle were assessed as parameters. RESULTS: All four dogs improved their stance and three of these dogs improved in gait at 4 and 6 weeks after TeNT injections without evidence of side effects or spreading of TeNT effects. At the same time, the size of the rectus femoris muscle diameter increased considerably as compared with baseline (baseline: 100%; 4 weeks: 148.7% ± 10.9%; 6 weeks: 137.1% ± 7.9%). CONCLUSIONS: Facilitation of α-motor neurons by TeNT injections into paretic hind limb muscles of four dogs improved standing and/or gait abilities and partly reversed muscle atrophy after SCI. The absence of generalized or painful muscle spasms supports the safety of low-dose TeNT. Therefore, TeNT might evolve as a promising therapeutic option for muscle paresis of central origin, e.g. in individuals with SCI, stroke or multiple sclerosis.


Gait/drug effects , Metalloendopeptidases/administration & dosage , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Tetanus Toxin/administration & dosage , Animals , Dogs , Gait/physiology , Injections, Intramuscular , Lumbar Vertebrae/injuries , Pilot Projects , Spinal Cord Injuries/physiopathology , Thoracic Vertebrae/injuries , Treatment Outcome
4.
BMC Microbiol ; 17(1): 187, 2017 Aug 24.
Article En | MEDLINE | ID: mdl-28836948

BACKGROUND: The bronchial epithelium serves as the first defendant line of host against respiratory inhaled pathogens, mainly through releasing chemokines (e.g. interleukin-8 (IL-8), interferon-induced protein 10 (IP-10) etc.) responsible for neutrophil or lymphocyte recruitment to promote the clearance of inhaled pathogens including Streptococcus pneumoniae (S. pneumoniae). Previous studies have shown that IL-8 expression is induced by pneumococcal virulence factors (e.g. pneumolysin, peptidoglycan-polysaccharides, pneumococcal surface protein A (PspA) etc.), which contributes to the pathogenesis of pneumonia. Whether other pneumococcal virulence factors are involved in inducing chemokines expression in epithelium is still unknown. RESULTS: We studied the effect of PepO, a widely expressed and newly discovered pneumococcal virulence protein, on the release of proinflammatory cytokines, IL-8 and IP-10, from human bronchial epithelial cell line BEAS-2B and identified the relevant signaling pathways. Incubation of BEAS-2B with PepO resulted in increased synthesis and release of IL-8 and IP-10 in a dose and time independent manner. We also detected the increased and sustained expression of TLR2 and TLR4 transcripts in BEAS-2B stimulated by PepO. PepO activation leaded to the phosphorylation of MAPKs, Akt and p65. Pharmacologic inhibitors of MAPKs, PI3K and IκB-α phosphorylation attenuated IL-8 release, while IP-10 production was just suppressed by inhibitors of IκB-α phosphorylation, PI3K and P38 MAPK. CONCLUSION: These results suggest that PepO enhances IL-8 and IP-10 production in BEAS-2B in a MAPKs-PI3K/Akt-p65 dependent manner, which may play critical roles in the pathogenesis of pneumonia.


Bacterial Proteins/pharmacology , Bronchi/metabolism , Chemokine CXCL10/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Interleukin-8/metabolism , Metalloendopeptidases/pharmacology , Streptococcus pneumoniae/metabolism , Bacterial Proteins/administration & dosage , Cell Line , Chemokine CXCL10/genetics , Cytokines/metabolism , Gene Expression Regulation, Enzymologic , Humans , Interleukin-8/genetics , Metalloendopeptidases/administration & dosage , Mitogen-Activated Protein Kinase Kinases/drug effects , NF-KappaB Inhibitor alpha/metabolism , Phosphatidylinositol 3-Kinases , Phosphorylation , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Streptococcus pneumoniae/pathogenicity , Time Factors , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Transcription, Genetic , Virulence Factors , eIF-2 Kinase/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Theranostics ; 7(6): 1749-1769, 2017.
Article En | MEDLINE | ID: mdl-28529649

The accumulations of excess lipids within liver and serum are defined as non-alcoholic fatty liver disease (NAFLD) and hyperlipemia respectively. Both of them are components of metabolic syndrome that greatly threaten human health. Here, a recombinant fusion protein (SAK-HV) effectively treated NAFLD and hyperlipemia in high-fat-fed ApoE-/- mice, quails and rats within just 14 days. Its triglyceride and cholesterol-lowering effects were significantly better than that of atorvastatin during the observation period. We explored the lipid-lowering mechanism of SAK-HV by the hepatic transcriptome analysis and serials of experiments both in vivo and in vitro. Unexpectedly, SAK-HV triggered a moderate energy and material-consuming liver proliferation to dramatically decrease the lipids from both serum and liver. We provided the first evidence that PGC-1α mediated the hepatic synthesis of female hormones during liver proliferation, and proposed the complement system-induced PGC-1α-estrogen axis via the novel STAT3-C/EBPß-PGC-1α pathway in liver as a new energy model for liver proliferation. In this model, PGC-1α ignited and fueled hepatocyte activation as an "igniter"; PGC-1α-induced estrogen augmented the energy supply of PGC-1α as an "ignition amplifier", then triggered the hepatocyte state transition from activation to proliferation as a "starter", causing triglyceride and cholesterol-lowering effects via PPARα-mediated fatty acid oxidation and LDLr-mediated cholesterol uptake, respectively. Collectively, the SAK-HV-triggered distinctive lipid-lowering strategy based on the new energy model of liver proliferation has potential as a novel short-period biotherapy against NAFLD and hyperlipemia.


Anticholesteremic Agents/administration & dosage , Biological Therapy/methods , Hyperlipidemias/therapy , Non-alcoholic Fatty Liver Disease/therapy , Recombinant Fusion Proteins/administration & dosage , Animals , Disease Models, Animal , Estrogens/metabolism , Hirudins/administration & dosage , Hirudins/genetics , Liver/pathology , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/genetics , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Quail , Rats , Recombinant Fusion Proteins/genetics , Treatment Outcome
6.
World J Gastroenterol ; 23(4): 603-613, 2017 Jan 28.
Article En | MEDLINE | ID: mdl-28216966

AIM: To evaluate the impact of recombinant Bacteroides fragilis enterotoxin-2 (BFT-2, or Fragilysin) on colorectal tumorigenesis in mice induced by azoxymethane/dextran sulfate sodium (AOM/DSS). METHODS: Recombinant proBFT-2 was expressed in Escherichia coli strain Rosetta (DE3) and BFT-2 was obtained and tested for its biological activity via colorectal adenocarcinoma cell strains SW-480. Seventy C57BL/6J mice were randomly divided into a blank (BC; n = 10), model (AD; n = 20), model + low-dose toxin (ADLT; n = 20, 10 µg), and a model + high-dose toxin (ADHT; n = 20, 20 µg) group. Mice weight, tumor formation and pathology were analyzed. Immunohistochemistry determined Ki-67 and Caspase-3 expression in normal and tumor tissues of colorectal mucosa. RESULTS: Recombinant BFT-2 was successfully obtained, along with its biological activity. The most obvious weight loss occurred in the AD group compared with the ADLT group (21.82 ± 0.68 vs 23.23 ± 0.91, P < 0.05) and the ADHT group (21.82 ± 0.68 vs 23.57 ± 1.06, P < 0.05). More tumors were found in the AD group than in the ADLT and ADHT groups (19.75 ± 3.30 vs 6.50 ± 1.73, P < 0.05; 19.75 ± 3.30 vs 6.00 ± 2.16, P < 0.05). Pathology showed that 12 mice had adenocarcinoma and 6 cases had adenoma in the AD group. Five mice had adenocarcinoma and 15 had adenoma in the ADLT group. Four mice had adenocarcinoma and 16 had adenoma in the ADHT group. The incidence of colorectal adenocarcinoma in both the ADHT group and the ADHT group was reduced compared to that in the AD group (P < 0.05, P < 0.05). The positive rate of Ki-67 in the ADLT group and the ADHT group was 50% and 40%, respectively, both of which were lower than that found in the AD group (94.44%, P < 0.05, P < 0.05). Caspase-3 expression in the ADLT group and the ADHT group was 45% and 55%, both of which were higher than that found in the BC group (16.67%, P < 0.05, P < 0.05). CONCLUSION: Oral administration with lower-dose biologically active recombinant BFT-2 inhibited colorectal tumorigenesis in mice.


Colorectal Neoplasms/therapy , Intestines/microbiology , Metalloendopeptidases/administration & dosage , Administration, Oral , Animals , Azoxymethane , Bacteroides fragilis , Body Weight , Caspase 3/metabolism , Cell Line, Tumor , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/microbiology , Dextran Sulfate , Humans , Immunohistochemistry , Intestines/pathology , Ki-67 Antigen/metabolism , Mice , Mice, Inbred C57BL , Recombinant Proteins/administration & dosage
7.
Front Med ; 11(1): 1-19, 2017 Mar.
Article En | MEDLINE | ID: mdl-28116631

Once thrombi have formed as part of the pathology defining myocardial infarction, ischemic stroke, peripheral arterial disease, deep venous thrombosis or other embolic disorders, the only clinically meaningful thrombolytic agents available for reversing the thrombogenic process are various plasminogen activators. These agents are enzymes that reverse fibrin polymerization underlying the coagulation process by converting endogenous plasminogen to plasmin, which cleaves the fibrin network to form increasingly smaller protein fragments, a process known as fibrinolysis. For the most part, the major clinically used thrombolytics, tissue plasminogen activator, urokinase and streptokinase, as well as the experimentally investigated agent staphylokinase, are the products of recombinant DNA technology, which permits molecular optimization of clinical efficacy. In all cases of molecular optimization and targeting, however, the primary challenge of thrombolytic therapy remains hemorrhagic side effects, which are especially devastating when they occur intracerebrally. Currently, the best strategy to ameliorate this adverse effect is nanoparticulate encapsulation or complexation, and many strategies of this sort are being actively pursued. This review summarizes the variety of targeted and untargeted thrombolytic formulations that have been investigated in preclinical studies.


Fibrinolytic Agents/administration & dosage , Thrombolytic Therapy/methods , Thrombosis/drug therapy , Tissue Plasminogen Activator/administration & dosage , Animals , Dogs , Fibrinolysis , Humans , Metalloendopeptidases/administration & dosage , Mice , Myocardial Infarction/complications , Streptokinase/administration & dosage , Stroke/complications , Thrombosis/complications , Thrombosis/diagnostic imaging , Ultrasonography , Urokinase-Type Plasminogen Activator/administration & dosage
8.
Curr Top Med Chem ; 17(32): 3425-3443, 2017.
Article En | MEDLINE | ID: mdl-29357802

Overexpressed cell-surface receptors are hallmarks of many disease states and are often used as markers for targeting diseased cells over healthy counterparts. Cell adhesion peptides, which are often derived from interacting regions of these receptor-ligand proteins, mimic surfaces of intact proteins and, thus, have been studied as targeting agents for various payloads to certain cell targets for cancers and autoimmune diseases. Because many cytotoxic agents in the free form are often harmful to healthy cells, the use of cell adhesion peptides in targeting their delivery to diseased cells has been studied to potentially reduce required effective doses and associated harmful side-effects. In this review, multiple cell adhesion peptides from extracellular matrix and ICAM proteins were used to selectively direct drug payloads, signal-inhibitor peptides, and diagnostic molecules, to diseased cells over normal counterparts. RGD constructs have been used to improve the selectivity and efficacy of diagnostic and drug-peptide conjugates against cancer cells. From this precedent, novel conjugates of antigenic and cell adhesion peptides, called Bifunctional Peptide Inhibitors (BPIs), have been designed to selectively regulate immune cells and suppress harmful inflammatory responses in autoimmune diseases. Similar peptide conjugations with imaging agents have delivered promising diagnostic methods in animal models of rheumatoid arthritis. BPIs have also been shown to generate immune tolerance and suppress autoimmune diseases in animal models of type-1 diabetes, rheumatoid arthritis, and multiple sclerosis. Collectively, these studies show the potential of cell adhesion peptides in improving the delivery of drugs and diagnostic agents to diseased cells in clinical settings.


ATPases Associated with Diverse Cellular Activities/chemistry , ATPases Associated with Diverse Cellular Activities/pharmacokinetics , Autoimmune Diseases/diagnosis , Autoimmune Diseases/drug therapy , Drug Delivery Systems/methods , Metalloendopeptidases/chemistry , Metalloendopeptidases/pharmacokinetics , Neoplasms/diagnosis , Neoplasms/drug therapy , ATPases Associated with Diverse Cellular Activities/administration & dosage , ATPases Associated with Diverse Cellular Activities/therapeutic use , Autoimmune Diseases/metabolism , Humans , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/therapeutic use , Neoplasms/metabolism
9.
PLoS One ; 11(12): e0168643, 2016.
Article En | MEDLINE | ID: mdl-27992592

The precise mechanisms by which Snake Venom Metalloproteinases (SVMPs) disrupt the microvasculature and cause haemorrhage have not been completely elucidated, and novel in vivo models are needed. In the present study, we compared the effects induced by BaP1, a PI SVMP isolated from Bothrops asper venom, and CsH1, a PIII SVMP from Crotalus simus venom, on cremaster muscle microvasculature by topical application of the toxins on isolated tissue (i.e., ex vivo model), and by intra-scrotal administration of the toxins (i.e., in vivo model). The whole tissue was fixed and immunostained to visualize the three components of blood vessels by confocal microscopy. In the ex vivo model, BaP1 was able to degrade type IV collagen and laminin from the BM of microvessels. Moreover, both SVMPs degraded type IV collagen from the BM in capillaries to a higher extent than in PCV and arterioles. CsH1 had a stronger effect on type IV collagen than BaP1. In the in vivo model, the effect of BaP1 on type IV collagen was widespread to the BM of arterioles and PCV. On the other hand, BaP1 was able to disrupt the endothelial barrier in PCV and to increase vascular permeability. Moreover, this toxin increased the size of gaps between pericytes in PCV and created new gaps between smooth muscle cells in arterioles in ex vivo conditions. These effects were not observed in the case of CsH1. In conclusion, our findings demonstrate that both SVMPs degrade type IV collagen from the BM in capillaries in vivo. Moreover, while the action of CsH1 is more directed to the BM of microvessels, the effects of BaP1 are widespread to other microvascular components. This study provides new insights in the mechanism of haemorrhage and other pathological effects induced by these toxins.


Abdominal Muscles/blood supply , Hemorrhage/chemically induced , Metalloendopeptidases/administration & dosage , Microvessels/drug effects , Snake Venoms/enzymology , Abdominal Muscles/drug effects , Administration, Topical , Animals , Capillary Permeability , Collagen Type IV/drug effects , Collagen Type IV/ultrastructure , Disease Models, Animal , Male , Metalloendopeptidases/pharmacology , Mice , Microscopy, Confocal , Microvessels/ultrastructure
10.
J Biotechnol ; 234: 105-115, 2016 Sep 20.
Article En | MEDLINE | ID: mdl-27496565

The goal of this study was to produce milligram quantities of pure, catalytically active, endotoxin-free recombinant neurolysin (rNln) in standard laboratory conditions for use as a research tool. To this end, we transformed E. coli cells with a plasmid construct for polyhistidine-tagged rNln, selected a high-expressing clone and determined the optimal time-point for translation of rNln. rNln was purified to homogeneity from the soluble pool of the cell lysate using Ni-NTA affinity and size-exclusion chromatography, followed by removal of endotoxins. Using this protocol ∼3mg pure, catalytically active and nearly endotoxin-free (≈0.003EU/µg protein) rNln was reproducibly obtained from 1l of culture. Lack of cytotoxicity of rNln preparation was documented in cultured mouse cells, whereas stability in whole mouse blood. Intraperitonealy administered rNln in mice reached the systemic circulation in intact and enzymatically active form with Tmax of 1h and T1/2 of ∼30min. Administration of rNln (2 and 10mg/kg) did not alter arterial blood pressure, heart rate, body temperature and blood glucose levels in mice. These studies demonstrate that the rNln preparation is suitable for cell culture and in vivo studies and can serve as a research tool to investigate the (patho)physiological function of this peptidase.


Metalloendopeptidases/biosynthesis , Metalloendopeptidases/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Animals , Astrocytes , Brain , Chromatography, Affinity , Chromatography, Gel , Endotoxins/chemistry , Escherichia coli/genetics , Female , Histidine/chemistry , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/isolation & purification , Mice , Neurons , Protein Stability , Rats , Recombinant Proteins/administration & dosage , Recombinant Proteins/isolation & purification , Tandem Mass Spectrometry
11.
Sci Rep ; 6: 27206, 2016 06 02.
Article En | MEDLINE | ID: mdl-27251373

The most effective strategy for protection against intracellular infections such as Leishmania is vaccination with live parasites. Use of recombinant proteins avoids the risks associated with live vaccines. However, due to low immunogenicity, they fail to trigger T cell responses particularly of CD8(+) cells requisite for persistent immunity. Previously we showed the importance of protein entrapment in cationic liposomes and MPL as adjuvant for elicitation of CD4(+) and CD8(+) T cell responses for long-term protection. In this study we investigated the role of cationic liposomes on maturation and antigen presentation capacity of dendritic cells (DCs). We observed that cationic liposomes were taken up very efficiently by DCs and transported to different cellular sites. DCs activated with liposomal rgp63 led to efficient presentation of antigen to specific CD4(+) and CD8(+) T cells. Furthermore, lymphoid CD8(+) T cells from liposomal rgp63 immunized mice demonstrated better proliferative ability when co-cultured ex vivo with stimulated DCs. Addition of MPL to vaccine enhanced the antigen presentation by DCs and induced more efficient antigen specific CD8(+) T cell responses when compared to free and liposomal antigen. These liposomal formulations presented to CD8(+) T cells through TAP-dependent MHC-I pathway offer new possibilities for a safe subunit vaccine.


CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/drug effects , Histocompatibility Antigens Class I/metabolism , Liposomes/administration & dosage , Recombinant Proteins/administration & dosage , Adjuvants, Immunologic , Animals , Antigen Presentation , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Immunization , Liposomes/pharmacology , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/immunology , Mice , Recombinant Proteins/immunology
12.
J Control Release ; 227: 45-57, 2016 Apr 10.
Article En | MEDLINE | ID: mdl-26876783

Several plasminogen activators (PAs) have been found effective in treating different thromboembolic diseases. However, administration of conventional thrombolytic therapy is limited by a low efficacy of present formulations of PAs. Conventional treatments using these therapeutic proteins are associated with several limitations including rapid inactivation and clearance, short half-life, bleeding complications or non-specific tissue targeting. Liposome-based formulations of PAs such as streptokinase, tissue-plasminogen activator and urokinase have been developed to improve the therapeutic efficacy of these proteins. Resulting liposomal formulations were found to preserve the original activity of PAs, promote their selective delivery and improve thrombus targeting. Therapeutic potential of these liposome-based PAs has been demonstrated successfully in various pre-clinical models in vivo. Reductions in unwanted side effects (e.g., hemorrhage or immunogenicity) as well as enhancements of efficacy and safety were achieved in comparison to currently existing treatment options based on conventional formulations of PAs. This review summarizes present achievements in: (i) preparation of liposome-based formulations of various PAs, (ii) development of PEGylated and targeted liposomal PAs, (iii) physico-chemical characterization of these developed systems, and (iv) testing of their thrombolytic efficacy. We also look to the future and the imminent arrival of theranostic liposomal formulations to move this field forward.


Fibrinolysin/administration & dosage , Fibrinolytic Agents/administration & dosage , Liposomes/chemistry , Metalloendopeptidases/administration & dosage , Streptokinase/administration & dosage , Tissue Plasminogen Activator/administration & dosage , Urokinase-Type Plasminogen Activator/administration & dosage , Animals , Fibrinolysin/therapeutic use , Fibrinolytic Agents/therapeutic use , Humans , Liposomes/ultrastructure , Metalloendopeptidases/therapeutic use , Nanostructures/chemistry , Nanostructures/ultrastructure , Streptokinase/therapeutic use , Thromboembolism/drug therapy , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/therapeutic use , Urokinase-Type Plasminogen Activator/therapeutic use
13.
Mol Cell Proteomics ; 15(2): 542-57, 2016 Feb.
Article En | MEDLINE | ID: mdl-26685126

Neurons require efficient transport mechanisms such as fast axonal transport to ensure neuronal homeostasis and survival. Neurotrophins and their receptors are conveyed via fast axonal retrograde transport of signaling endosomes to the soma, where they elicit transcriptional responses. Despite the essential roles of signaling endosomes in neuronal differentiation and survival, little is known about their molecular identity, dynamics, and regulation. Gaining a better mechanistic understanding of these organelles and their kinetics is crucial, given the growing evidence linking vesicular trafficking deficits to neurodegeneration. Here, we exploited an affinity purification strategy using the binding fragment of tetanus neurotoxin (HCT) conjugated to monocrystalline iron oxide nanoparticles (MIONs), which in motor neurons, is transported in the same carriers as neurotrophins and their receptors. To quantitatively assess the molecular composition of HCT-containing signaling endosomes, we have developed a protocol for triple Stable Isotope Labeling with Amino acids in Cell culture (SILAC) in embryonic stem cell-derived motor neurons. After HCT internalization, retrograde carriers were magnetically isolated at different time points and subjected to mass-spectrometry and Gene Ontology analyses. This purification strategy is highly specific, as confirmed by the presence of essential regulators of fast axonal transport in the make-up of these organelles. Our results indicate that signaling endosomes undergo a rapid maturation with the acquisition of late endosome markers following a specific time-dependent kinetics. Strikingly, signaling endosomes are specifically enriched in proteins known to be involved in neurodegenerative diseases and neuroinfection. Moreover, we highlighted the presence of novel components, whose precise temporal recruitment on signaling endosomes might be essential for proper sorting and/or transport of these organelles. This study provides the first quantitative proteomic analysis of signaling endosomes isolated from motor neurons and allows the assembly of a functional map of these axonal carriers involved in long-range neuronal signaling.


Axons/metabolism , Motor Neurons/metabolism , Nerve Growth Factors/metabolism , Neurodegenerative Diseases/genetics , Proteomics , Animals , Axonal Transport/drug effects , Axonal Transport/genetics , Axons/drug effects , Axons/pathology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endocytosis/genetics , Endosomes/genetics , Endosomes/metabolism , Endosomes/pathology , Ferric Compounds/administration & dosage , Ferric Compounds/chemistry , Humans , Isotope Labeling , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/chemistry , Mice , Motor Neurons/drug effects , Motor Neurons/pathology , Nerve Growth Factors/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Signal Transduction , Tetanus Toxin/administration & dosage , Tetanus Toxin/chemistry
14.
Int J Hyperthermia ; 31(1): 67-73, 2015 Feb.
Article En | MEDLINE | ID: mdl-25766387

BACKGROUND: Clinical efficacy of thrombolytic drugs is limited by lack of specific delivery and requires large therapeutic doses which increase toxicity. Encapsulating these drugs in temperature-sensitive liposomes and applying hyperthermia to deliver thrombolytic agents locally to thrombus might theoretically favourably alter the therapeutic window. The objectives of this study were to formulate liposomes encapsulating thrombolytics and assess thrombolytic activity following hyperthermia. METHODS: Three liposome formulations were investigated: temperature-sensitive liposome (TSL, DPPC:DSPE-PEG2000 (mol% 95:5)), low temperature-sensitive liposome (LTSL, DPPC:MSPC:DSPE-PEG2000 (mol% 85.3:9.7:5)), and traditional temperature-sensitive liposome (TTSL, DPPC:HSPC:Chol:DSPE-PEG2000 (mol% 55:25:15:5)). To characterise temperature-dependent release of high molecular weight cargo from each formulation, fluorescein-conjugated dextrans (70 kDa) were loaded and release was quantified via spectrophotometry. Staphylokinase (SAK), urokinase, and tissue-type plasminogen activator were also loaded individually into each liposome formulation. Leakage at 37 °C and release at 38-44 °C were quantified via chromogenic enzymatic activity assay. Clot lysis was evaluated by measuring mass of blood clots before and after thrombolytic liposome treatment. RESULTS: The LTSL formulation had optimal release characteristics with maximum release at 41.3 °C. Release of dextrans from LTSLs was observed to be 11.5 ± 1.5%, 79.7 ± 1.6%, and 93.6 ± 3.7% after 15 min in plasma at 37°, 39°, and 41.3 °C, respectively. The SAK LTSL had the highest release/leakage ratio and demonstrated greater clot lysis. CONCLUSIONS: The SAK LTSL achieves significant clot lysis in vitro. When combined with local hyperthermia, the SAK LTSL potentially produces sufficient thrombolysis while minimising systemic side effects.


Blood Coagulation/drug effects , Fibrinolytic Agents/administration & dosage , Metalloendopeptidases/administration & dosage , Tissue Plasminogen Activator/administration & dosage , Urokinase-Type Plasminogen Activator/administration & dosage , Humans , Hyperthermia, Induced , Lipids/chemistry , Liposomes , Male , Metalloendopeptidases/chemistry , Polyethylene Glycols/chemistry , Temperature , Tissue Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/chemistry
15.
Biosci Biotechnol Biochem ; 78(9): 1514-21, 2014.
Article En | MEDLINE | ID: mdl-25209498

It is an usual clinical phenomenon that cancer patients are prone to thrombosis. Until now, there have been no efficient methods or appropriate drugs to prevent and cure tumor thrombus. Therefore, the construction of a bifunctional chimeric protein for the treatment of cancer, complicated with thrombosis, is of great significance. Utilizing the superantigenic activity of staphylococcal enterotoxin C2 (SEC2) and the thrombolytic activity of staphylokinase (Sak), Sak-linker-SEC2 and SEC2-linker-Sak were constructed which had good anti-tumor and thrombolytic activities at the same time. Due to the intrinsic emetic activity of SEC2 and high molecular weight (MW) of chimeric proteins (44 kDa), their clinical applications will be restricted. In this study, novel chimeric proteins including ΔSEC2-ΔSak and ΔSak-ΔSEC2 were constructed through the truncation of SEC2 and Sak without 9-Ala linker and His-tag. Compared with the former, both the truncated proteins preserved nearly the same anti-tumor and thrombolytic activities. In addition, their MWs were only 29 kDa and their immunoreactivities were slightly lower than that of Sak-linker-SEC2 and SEC2-linker-Sak, respectively. Therefore, the novel chimeric proteins possessed merits and characteristics, such as low MS, low immunogenicity, and difunctionality which the former had not. It will be of great interest if the above-mentioned proteins can be used to cure Trousseau syndrome in clinic.


Enterotoxins/genetics , Metalloendopeptidases/genetics , Neoplasms/drug therapy , Recombinant Fusion Proteins/administration & dosage , Thrombosis/drug therapy , Enterotoxins/administration & dosage , Fibrinolytic Agents/administration & dosage , Humans , Metalloendopeptidases/administration & dosage , Neoplasms/complications , Neoplasms/genetics , Neoplasms/pathology , Protein Engineering , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Staphylococcus aureus/chemistry , Thrombosis/complications , Thrombosis/genetics , Thrombosis/pathology
16.
J Neurosci ; 32(6): 1962-8, 2012 Feb 08.
Article En | MEDLINE | ID: mdl-22323709

Vesicular (v)- and target (t)-SNARE proteins assemble in SNARE complex to mediate membrane fusion. Tetanus neurotoxin-insensitive vesicular-associated membrane protein (TI-VAMP/VAMP7), a vesicular SNARE expressed in several cell types including neurons, was previously shown to play a major role in exocytosis involved in neurite growth in cultured neurons. Here we generated a complete constitutive knock-out by deleting the exon 3 of Vamp7. Loss of TI-VAMP expression did not lead to any striking developmental or neurological defect. Knock-out mice displayed decreased brain weight and increased third ventricle volume. Axon growth appeared normal in cultured knock-out neurons. Behavioral characterization unraveled that TI-VAMP knock-out was associated with increased anxiety. Our results thus suggest compensatory mechanisms allowing the TI-VAMP knock-out mice to fulfill major developmental processes. The phenotypic traits unraveled here further indicate an unexpected role of TI-VAMP-mediated vesicular traffic in anxiety and suggest a role for TI-VAMP in higher brain functions.


Anxiety/genetics , Metalloendopeptidases , R-SNARE Proteins/deficiency , R-SNARE Proteins/genetics , Tetanus Toxin , Animals , Anxiety/etiology , Anxiety/psychology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Male , Metalloendopeptidases/administration & dosage , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Rabbits , Tetanus Toxin/administration & dosage
17.
Pharmacol Rep ; 63(5): 1169-79, 2011.
Article En | MEDLINE | ID: mdl-22180359

SAK-RGD-K2-Hir and SAK-RGD-K2-Hirul are recombinant proteins that are derivatives of r-SAK (recombinant staphylokinase). They are characterized by their fibrin-specific plasminogen activation properties and their antithrombin and antiplatelet activities. The difference between these proteins is the presence of the antithrombotic fragment (hirudin or hirulog) in the C-terminal portion of the r-SAK. The aim of the present study was to examine the thrombolytic potentials of SAK-RGD-K2-Hir and SAK-RGD-K2-Hirul in an electrically induced carotid artery thrombosis model in rats and to compare the potentials to that of r-SAK. We determined that a bolus injection of SAK-RGD-K2-Hirul was more effective than one of r-SAK in the improvement and maintenance of carotid patency and in arterial thrombus weight reduction; however, it had the same potency as SAK-RGD-K2-Hir. The bleeding time, prothrombin time and activated partial thromboplastin time were significantly prolonged in the animals that were treated with either dose (1.5 or 3.0 mg/kg) of SAK-RGD-K2-Hir or SAK-RGD-K2-Hirul, whereas no changes were observed in the plasma fibrinogen concentration or the α2 plasmin inhibitor level. r-SAK alone did not change the bleeding time or coagulation parameters. In conclusion, our findings demonstrate the thrombolytic activity of intravenous bolus injection of the novel thrombolytic agent SAK-RGD-K2-Hirul in rats. Although this protein compares favorably with r-SAK, we were unable to show the presence of any beneficial effects of SAK-RGD-K2-Hirul over those of SAK-RGD-K2-Hir. Furthermore, our results suggest that high doses of SAK-RGD-K2-Hirul bear the risk of bleeding.


Carotid Artery Thrombosis/drug therapy , Fibrinolytic Agents/pharmacology , Hirudins/pharmacology , Metalloendopeptidases/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Bleeding Time/methods , Blood Coagulation/drug effects , Carotid Artery Thrombosis/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/toxicity , Hirudins/administration & dosage , Hirudins/toxicity , Injections, Intravenous , Male , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/toxicity , Rats , Rats, Wistar , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/toxicity
18.
J Tradit Chin Med ; 31(2): 103-6, 2011 Jun.
Article En | MEDLINE | ID: mdl-21977808

OBJECTIVE: To investigate the effect of recombinant staphylokinase (r-Sak) and the Chinese medicine Yihuo Qingyi Decoction ((see test) Herbal decoction for severe acute pancreatitis) in the treatment of the severe acute pancreatitis (SAP) in rats, and to observe the synergistic effect of the two. METHODS: One hundred and sixty-two adult male SD rats with the body mass of 250-280 g were randomly divided into the following 5 groups: sham operation group (n = 18), control group (n = 36), Yihuo Qingyi Decoction treatment group (n = 36), r-Sak treatment group (n = 36), and Yihuo Qingyi Decoction plus r-Sak treatment group (n = 36). The SAP ratmodel was prepared by retrograde injection of 5% sodium taurocholate into the cholangiopancreatic duct. Two days before modeling, Yihuo Qingyi Decoction was intragastrically administrated, and r-Sak was intraperitoneally injected. The survival rate within 18 h after modeling was determined. The pancreatic blood flow, the weight of ascites, and the serum amylase and lipase were investigated at 6 h, 12 h, and 18kh after modeling, and the pancreatic tissue was examined under light microscopy to see its pathological change. RESULTS: The 18 h survival count of group A, B, C, D and E rats was 9, 2, 6, 7 and 8 respectively. After r-Sak and Yihuo Qingyi Decoction intervention, the serum amylase and lipase and the weight of ascites were significantly decreased, especially in group E.18 h after modeling, the level of the serum amylase and lipase and the weight of ascites in group E was 1 100 +/- 118 U x L(-1), 1 000 +/- 150 U x L(-1) and 13.40 +/- 1.80 g respectively, obviously lower than that of group B (P < 0.05). After SAP was induced, the pancreatic blood flow showed a tendency to decrease, but the decrease extent in the treatment groups was smaller than that in the control group. 18h after modeling, the pancreatic blood flow in group B and group E was 30.16 +/- 8.96 mL x 100 g(-1) x min(-1), and 129.10 +/- 42.58 mL x 100 g(-1) x min respectively, there was significant difference (P < 0.05). The pathological change of the pancreatic tissue was alleviated in the treatment groups. CONCLUSION: Both r-Sak and Yihuo Qingyi Decoction play a beneficial role in the treatment of rat SAP and there is a synergistic effect between the two.


Drugs, Chinese Herbal/administration & dosage , Metalloendopeptidases/administration & dosage , Pancreatitis/drug therapy , Acute Disease , Animals , Drug Synergism , Male , Myocardium/pathology , Pancreas/blood supply , Pancreas/pathology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage
19.
Parasite Immunol ; 33(2): 95-103, 2011 Feb.
Article En | MEDLINE | ID: mdl-21226722

The 63-kDa antigen of Leishmania donovani is a membrane-anchored matrix metalloprotease that has been shown to be involved in the infection process. We have shown that this antigen alone generates a Th1 type of protective response that is partial but when the animals are primed with the antigen along with the Hsp70, the level of protection is raised significantly, which is demonstrated by a considerable reduction in parasite load of immunized animals when compared to the infected controls. Delayed-type hypersensitivity responses to leishmanin were measured as an index of cell-mediated immune response and were found to be higher in immunized animals when compared to the infected controls, the maximum being in the animals immunized with cocktail of both the antigens. Maximum IgG2a and minimum IgG1 levels were observed in this group of animals. These animals also generated maximum levels of IFN-γ and IL-2 and minimum levels of IL-4 and IL-10 pointing towards the generation of a protective Th1 response and the suppression of the Th2 type of immune response.


HSP70 Heat-Shock Proteins/immunology , Leishmania donovani/immunology , Leishmaniasis Vaccines/immunology , Metalloendopeptidases/immunology , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/immunology , Female , HSP70 Heat-Shock Proteins/administration & dosage , Hypersensitivity, Delayed , Immunoglobulin G/blood , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-2/metabolism , Interleukin-4/metabolism , Leishmania donovani/enzymology , Leishmaniasis Vaccines/administration & dosage , Male , Metalloendopeptidases/administration & dosage , Mice , Mice, Inbred BALB C , Th1 Cells/immunology , Th2 Cells/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
20.
J Biol Chem ; 285(45): 34390-400, 2010 Nov 05.
Article En | MEDLINE | ID: mdl-20807771

Endothelin-converting enzyme-2 (ECE-2) is a membrane-bound zinc-dependent metalloprotease that shares a high degree of sequence homology with ECE-1, but displays an acidic pH optimum characteristic of maturing enzymes acting late in the secretory pathway. Although ECE-2, like ECE-1, can cleave the big endothelin intermediate to produce the vasoconstrictive endothelin peptide, its true physiological function remains to be elucidated, a task that is hampered by the lack of specific tools to study and discriminate ECE-2 from ECE-1, i.e. specific substrates and/or specific inhibitors. To fill this gap, we searched for novel ECE-specific peptide substrates. To this end, peptides derived from the big endothelin intermediate were tested using ECE-1 and ECE-2, leading to the identification of an ECE-1-specific substrate. Moreover, screening of our proprietary fluorigenic peptide Fluofast® libraries using ECE-1 and ECE-2 allowed the identification of Ac-SKG-Pya-F-W-Nop-GGK-NH(2) (PL405), as a specific and high affinity ECE-2 substrate. Indeed, ECE-2 cleaved PL405 at the Pya-F amide bond with a specificity constant (k(cat)/K(m)) of 8.1 ± 0.9 × 10(3) M(-1) s(-1). Using this novel substrate, we also characterized the first potent (K(i) = 7.7 ± 0.3 nM) and relatively selective ECE-2 inhibitor and developed a quantitative fluorigenic ECE-2 assay. The assay was used to study the ex vivo ECE-2 activity in wild type and ECE-2 knock-out tissues and was found to truly reflect ECE-2 expression patterns. The PL405 assay is thus the first tool to study ECE-2 inhibition using high throughput screening or for ex vivo ECE-2 quantification.


Aspartic Acid Endopeptidases/biosynthesis , Aspartic Acid Endopeptidases/chemistry , Endothelins/chemistry , Enzyme Assays/methods , Fluorescent Dyes/chemistry , Gene Expression Regulation, Enzymologic/physiology , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/chemistry , Peptides/chemistry , Animals , Aspartic Acid Endopeptidases/administration & dosage , Aspartic Acid Endopeptidases/genetics , Endothelin-Converting Enzymes , Endothelins/genetics , Endothelins/metabolism , Enzyme Inhibitors/chemistry , Humans , Metalloendopeptidases/administration & dosage , Metalloendopeptidases/genetics , Mice , Organ Specificity , Peptide Library , Peptides/genetics , Peptides/metabolism , Substrate Specificity
...