Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 522
Filter
1.
Mol Pharmacol ; 99(6): 435-447, 2021 06.
Article in English | MEDLINE | ID: mdl-33795352

ABSTRACT

Rearranged during transfection (RET) rearrangements occur in 1% to 2% of lung adenocarcinomas as well as other malignancies and are now established targets for tyrosine kinase inhibitors. We developed three novel RET fusion-positive (RET+) patient-derived cancer cell lines, CUTO22 [kinesin 5B (KIF5B)-RET fusion], CUTO32 (KIF5B-RET fusion), and CUTO42 (echinoderm microtubule-associated protein-like 4-RET fusion), to study RET signaling and response to therapy. We confirmed each of our cell lines expresses the RET fusion protein and assessed their sensitivity to RET inhibitors. We found that the CUTO22 and CUTO42 cell lines were sensitive to multiple RET inhibitors, whereas the CUTO32 cell line was >10-fold more resistant to three RET inhibitors. We discovered that our RET+ cell lines had differential regulation of the mitogen-activated protein kinase and phosphoinositide 3-kinase/protein kinase B (AKT) pathways. After inhibition of RET, the CUTO42 cells had robust inhibition of phosphorylated AKT (pAKT), whereas CUTO22 and CUTO32 cells had sustained AKT activation. Next, we performed a drug screen, which revealed that the CUTO32 cells were sensitive (<1 nM IC50) to inhibition of two cell cycle-regulating proteins, polo-like kinase 1 and Aurora kinase A. Finally, we show that two of these cell lines, CUTO32 and CUTO42, successfully establish xenografted tumors in nude mice. We demonstrated that the RET inhibitor BLU-667 was effective at inhibiting tumor growth in CUTO42 tumors but had a much less profound effect in CUTO32 tumors, consistent with our in vitro experiments. These data highlight the utility of new RET+ models to elucidate differences in response to tyrosine kinase inhibitors and downstream signaling regulation. Our RET+ cell lines effectively recapitulate the interpatient heterogeneity observed in response to RET inhibitors and reveal opportunities for alternative or combination therapies. SIGNIFICANCE STATEMENT: We have derived and characterized three novel rearranged during transfection (RET) fusion non-small cell lung cancer cell lines and demonstrated that they have differential responses to RET inhibition as well as regulation of downstream signaling, an area that has previously been limited by a lack of diverse cell line modes with endogenous RET fusions. These data offer important insight into regulation of response to RET tyrosine kinase inhibitors and other potential therapeutic targets.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Signal Transduction , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Female , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Proto-Oncogene Proteins c-ret/genetics , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Xenograft Model Antitumor Assays
2.
Sci Rep ; 10(1): 14181, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32843670

ABSTRACT

Glial fibrillary acidic protein expressing (GFAP+) glia modulate nociceptive neuronal activity in both the peripheral nervous system (PNS) and the central nervous system (CNS). Resident GFAP+ glia in dorsal root ganglia (DRG) known as satellite glial cells (SGCs) potentiate neuronal activity by releasing pro-inflammatory cytokines and neuroactive compounds. In this study, we tested the hypothesis that SGC Gq-coupled receptor (Gq-GPCR) signaling modulates pain sensitivity in vivo using Gfap-hM3Dq mice. Complete Freund's adjuvant (CFA) was used to induce inflammatory pain, and mechanical sensitivity and thermal sensitivity were used to assess the neuromodulatory effect of glial Gq-GPCR activation in awake mice. Pharmacogenetic activation of Gq-GPCR signaling in sensory SGCs decreased heat-induced nociceptive responses and reversed inflammation-induced mechanical allodynia via peripheral adenosine A1 receptor activation. These data reveal a previously unexplored role of sensory SGCs in decreasing afferent excitability. The identified molecular mechanism underlying the analgesic role of SGCs offers new approaches for reversing peripheral nociceptive sensitization.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Hyperalgesia/prevention & control , Inflammation/physiopathology , Neuroglia/enzymology , Nociception/physiology , Receptor, Adenosine A1/physiology , Receptor, Muscarinic M3/physiology , Animals , Benzilates/pharmacology , Clozapine/analogs & derivatives , Clozapine/pharmacology , Freund's Adjuvant/toxicity , Genes, Synthetic , Hot Temperature , Hyperalgesia/physiopathology , Inflammation/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscarinic Agonists/pharmacology , Neuroglia/physiology , Nortropanes/pharmacology , Promoter Regions, Genetic , Purinergic P1 Receptor Agonists/pharmacology , Purinergic P1 Receptor Antagonists/pharmacology , Receptor, Adenosine A1/drug effects , Receptor, Muscarinic M3/drug effects , Receptor, Muscarinic M3/genetics , Receptors, G-Protein-Coupled , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Theophylline/analogs & derivatives , Theophylline/pharmacology , Touch , Xanthines/pharmacology
3.
Zhonghua Fu Chan Ke Za Zhi ; 54(9): 601-607, 2019 Sep 25.
Article in Chinese | MEDLINE | ID: mdl-31550776

ABSTRACT

Objective: To explore the pathways of preeclampsia by investigating different effects of pravastatin (Pra) on and soluble FMS tyrosine kinase-1 (sFlt-1), placental growth factor (PlGF) and vascular endothelial growth factor (VEGF) in different preeclampsia (PE)-like mouse models. Methods: C57BL/6J mice were randomly subcutaneously injected with N-nitro-L-arginine methyl ester (L-NAME) or intraperitoneally injected with lipopolysaccharide (LPS) as PE-like mouse model, saline as normal pregnancy control (Con) respectively, daily at gestational 7-18 days. Pra was given daily at gestational 8-18 days in each model group and the mice were divided into Pra (L-NAME+Pra, LPS+Pra, Con+Pra) and saline (L-NAME+NS, LPS+NS, Con+NS) groups. Liver,placental tissue and blood of pregnant mice were collected on the 18th day of pregnancy. The levels of VEGF, PlGF and sFlt-1 in the liver, placenta and serum of mice in each group were compared by western blot, ELISA and real-time fluorescence quantitative PCR (RT-PCR). Results: (1) ELISA: Serum VEGF (205.70±3.43, 154.60±2.31) and PlGF (131.5±3.75, 101.50±4.31) levels were significantly increased in L-NAME+Pra group compared with L-NAME+NS group (all P<0.05). Serum VEGF (202.30±4.90, 144.50±6.71) and PlGF (121.50±3.86, 95.41±4.08) levels were significantly higher in LPS+Pra group than those in LPS+NS group (all P<0.05). Serum sFlt-1 level in LPS+Pra group was significantly lower than that in LPS+NS group (3.01±0.50, 776.60±80.06), serum sFlt-1 level in L-NAME+Pra group was significantly lower than that in L-NAME+NS group (2.60±0.06, 583.70±9.83; all P<0.05). (2) Western blot: the expression levels of PlGF (1.344±0.118, 0.664±0.143) and VEGF (1.34±0.12, 0.66±0.14) in the liver of mice in the L-NAME+Pra group were significantly higher than those in the L-NAME+NS group (all P<0.05), but the expression levels of PlGF and VEGF in the placenta of L-NAME+Pra group were not significantly different from those of L-NAME+NS group (all P>0.05). The expression levels of PlGF and VEGF in placenta and liver of pregnant mice in LPS+Pra group were not significantly different from those in LPS+N group (all P>0.05). (3) RT-PCR: the mRNA expression of PlGF and VEGF in placenta and liver of L-NAME+Pra group were not significantly different from those in L-NAME+NS group (all P>0.05). The mRNA expression levels of PlGF and VEGF in placenta and liver of LPS+Pra group were not significantly different from those of LPS+NS group (all P>0.05). Conclusions: Pra has different regulatory effects on vascular endothelial function in different PE-like models. It reveals that different pathogenesis and pathways exist in different PE-like changes.


Subject(s)
Placenta Growth Factor/drug effects , Pravastatin/therapeutic use , Pre-Eclampsia/drug therapy , Recombinant Fusion Proteins/drug effects , Vascular Endothelial Growth Factor A/drug effects , Animals , Anticholesteremic Agents/pharmacology , Biomarkers/blood , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice , Mice, Inbred C57BL , Placenta , Placenta Growth Factor/blood , Polymerase Chain Reaction , Pravastatin/pharmacology , Pre-Eclampsia/blood , Pregnancy , Real-Time Polymerase Chain Reaction , Recombinant Fusion Proteins/blood , Vascular Endothelial Growth Factor A/blood
4.
Blood ; 131(20): 2205-2214, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29588277

ABSTRACT

Hemophilia, or inherited genetic deficiencies in coagulation factors, results in uncontrolled bleeding requiring replacement therapy with recombinant proteins given preventively or on demand. However, a major problem with these approaches is the potential for development of immune responses to the administered proteins due to the underlying genetic deficiency of the factor(s) throughout life. As such, there is great interest in developing strategies that avoid immunogenicity and induce immune tolerance. Recently, recombinant factor VIII (rFVIII) and rFIX fused to the crystallizable fragment (Fc) domain of immunoglobulin G (IgG) have been developed as therapeutic agents for hemophilia A and B, respectively. Although it is well known that the possession of an Fc domain confers IgG's longer-lasting circulating half-life, it is not generally appreciated that the Fc domain also confers immunoregulatory properties that are associated with the induction of tolerance. Here, we review some of the latest advances in our understanding of the tolerogenic abilities of IgG Fc and the impact of Fc-fusion proteins of rFVIII on the treatment of hemophilia.


Subject(s)
Factor VIII/therapeutic use , Hemophilia A/drug therapy , Hemophilia A/immunology , Immune Tolerance , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Recombinant Fusion Proteins/drug effects , Animals , Disease Management , Factor IX/adverse effects , Factor IX/therapeutic use , Factor VIII/adverse effects , Factor VIII/genetics , Hemophilia A/blood , Hemophilia B/blood , Hemophilia B/drug therapy , Hemophilia B/immunology , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Immunomodulation , Isoantibodies/immunology , Receptors, Fc/metabolism , Treatment Outcome
5.
J Clin Lab Anal ; 32(1)2018 Jan.
Article in English | MEDLINE | ID: mdl-28303602

ABSTRACT

BACKGROUND: Although clinical management of drug resistance is routinely based on genotypic methods, phenotypic assays remain necessary for the characterization of novel HIV-1 inhibitors, particularly against common drug-resistant variants. We describe the development and assessment of the performance of a recombinant virus assay for measuring HIV-1 susceptibility to protease (PR), reverse transcriptase (RT), and integrase (IN) inhibitors. METHODS: The system is based on the creation of replication-competent chimeric viruses through homologous recombination between patient or laboratory virus-derived PCR fragments and the corresponding NL4-3 vector where the whole Gag-PR, RT-RNaseH or IN coding regions has been deleted through inverse PCR. The susceptibility to nucleoside (NRTIs) and non-nucleoside (NNRTIs) RT inhibitors and to IN inhibitors (INIs) is calculated through a single-round infection assay in TZM-bl cells, while protease inhibitor (PI) activity is determined through a first round of infection in MT-2 cells followed by infection of TZM-bl cells with MT-2 supernatants. RESULTS: The assay showed excellent reproducibility and accuracy when testing PI, NRTI, NNRTI, and INI susceptibility of drug-resistant clones previously characterized through the reference pseudoparticle-based Phenosense assay. The coefficient of interassay variation in fold change (FC) resistance was 12.0%-24.3% when assaying seven drug/clones pairs in three runs. FC values calculated by the Phenosense and in-house for 20 drug/clones pairs were in good agreement, with mean±SD ratio of 1.14±0.33 and no cases differing by more than twofold. CONCLUSIONS: The described phenotypic assay can be adopted to evaluate the antiviral activity of licensed and investigational HIV-1 drugs targeting any of the three HIV-1 enzymes.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Cell Line , HIV Infections/virology , HIV-1/classification , Humans , Phenotype , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Reproducibility of Results
6.
FEBS J ; 284(24): 4278-4297, 2017 12.
Article in English | MEDLINE | ID: mdl-29076638

ABSTRACT

Otolin-1 is a collagen-like protein expressed in the inner ear of vertebrates. It provides an organic scaffold for otoliths in fish and otoconia in land vertebrates. In this study, the expression and purification procedure of C1q-like domain of otolin-1 from human and zebrafish was developed. The structure and stability of the proteins were investigated. The results of sedimentation velocity analytical ultracentrifugation and small-angle X-ray scattering indicated that the C1q-like domain of otolin-1 forms stable trimers in solution in the presence of calcium ions. It was also observed that calcium ions influenced the secondary structure of the proteins. C1q-like domains were stabilized by the calcium ions. The human variant was especially affected by the calcium ions. The results indicate the importance of the C1q-like domain for the assembly of the organic matrix of otoliths and otoconia.


Subject(s)
Calcium/pharmacology , Extracellular Matrix Proteins/chemistry , Zebrafish Proteins/chemistry , Amino Acid Sequence , Animals , Calcium/physiology , Chromatography, Gel , Crystallography, X-Ray , Extracellular Matrix Proteins/drug effects , Extracellular Matrix Proteins/isolation & purification , Humans , Models, Molecular , Otolithic Membrane/metabolism , Protein Conformation , Protein Denaturation , Protein Domains , Protein Stability/drug effects , Protein Structure, Secondary/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/drug effects , Scattering, Radiation , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Structure-Activity Relationship , Ultracentrifugation , Zebrafish Proteins/drug effects , Zebrafish Proteins/isolation & purification
7.
PLoS One ; 12(1): e0169882, 2017.
Article in English | MEDLINE | ID: mdl-28118359

ABSTRACT

Voltage-gated sodium channels (VGSC) regulate neuronal excitability by governing action potential (AP) generation and propagation. Recent studies have revealed that AMP-activated protein kinase (AMPK) activators decrease sensory neuron excitability, potentially by preventing sodium (Na+) channel phosphorylation by kinases such as ERK or via modulation of translation regulation pathways. The direct positive allosteric modulator A769662 displays substantially greater efficacy than other AMPK activators in decreasing sensory neuron excitability suggesting additional mechanisms of action. Here, we show that A769662 acutely inhibits AP firing stimulated by ramp current injection in rat trigeminal ganglion (TG) neurons. PT1, a structurally dissimilar AMPK activator that reduces nerve growth factor (NGF) -induced hyperexcitability, has no influence on AP firing in TG neurons upon acute application. In voltage-clamp recordings, application of A769662 reduces VGSC current amplitudes. These findings, based on acute A769662 application, suggest a direct channel blocking effect. Indeed, A769662 dose-dependently blocks VGSC in rat TG neurons and in Nav1.7-transfected cells with an IC50 of ~ 10 µM. A769662 neither displayed use-dependent inhibition nor interacted with the local anesthetic (LA) binding site. Popliteal fossa administration of A769662 decreased noxious thermal responses with a peak effect at 5 mins demonstrating an analgesic effect. These data indicate that in addition to AMPK activation, A769662 acts as a direct blocker/modulator of VGSCs, a potential mechanism enhancing the analgesic property of this compound.


Subject(s)
AMP-Activated Protein Kinases/drug effects , Analgesics/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/drug effects , Pyrones/pharmacology , Sensory Receptor Cells/drug effects , Sodium Channel Blockers/pharmacology , Thiophenes/pharmacology , Anesthetics, Local/metabolism , Animals , Binding Sites/genetics , Biphenyl Compounds , Drug Evaluation, Preclinical , HEK293 Cells , Hot Temperature/adverse effects , Humans , Male , Metformin/pharmacology , NAV1.7 Voltage-Gated Sodium Channel/genetics , Neural Conduction/drug effects , Pain/drug therapy , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Resveratrol , Sensory Receptor Cells/enzymology , Stilbenes/pharmacology , Thiazoles/pharmacology , Trigeminal Ganglion/drug effects , meta-Aminobenzoates/pharmacology
8.
J Pharmacol Exp Ther ; 360(2): 249-259, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27856937

ABSTRACT

Rheumatoid arthritis (RA), an inflammatory autoimmune disorder, is characterized by synovial hyperplasia and bony destruction. The pathogenesis of RA includes redox dysregulation, concomitant with increased levels of proinflammatory mediators. As the ability of allylpyrocatechol (APC), a phytoconstituent of Piper betle leaves, to alleviate oxidative stress has been demonstrated in patients with RA, its antiarthritic activity was evaluated in an animal model of arthritis, and the underlying mechanism(s) of action clarified. The animal model was established by immunizing rats with bovine collagen type II (CII) followed by lipopolysaccharide, along with a booster dose of CII on day 15. Rats were treated with APC or methotrexate (MTX) from days 11 to 27, when paw edema, radiography, histopathology, and markers of inflammation were evaluated. The pro/antiinflammatory signaling pathways were studied in a RAW264.7 macrophage cell line. Allylpyrocatechol (APC) prevented the progression of arthritis as was evident from the reduction in paw edema, and attenuation of damage to bones and cartilage shown by radiography and histopathology. Additionally, there was reduction in the levels of proinflammatory cytokines [tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6)] and restoration of the redox balance. Importantly, MTX ameliorated the features of arthritis but not the associated oxidative stress. In RAW264.7, APC inhibited generation of nitric oxide and proinflammatory cytokines (TNF-α, IL-6, and IL-12p40), and modulated the phosphorylation of proinflammatory (extracellular signal-regulated kinase 1/2, stress-activated protein kinase/c-Jun N-terminal protein kinase, and Janus kinase/signal transducers and activators of transcription) and cytoprotective (nuclear factor erythroid 2-related factor 2, heme oxygenase-1) signaling pathways. Taken together, APC controlled the development of arthritis, possibly via modulation of signaling pathways, and deserves further consideration as a therapy for RA.


Subject(s)
Arthritis, Experimental/chemically induced , Arthritis, Experimental/drug therapy , Catechols/pharmacology , Collagen/adverse effects , MAP Kinase Signaling System/drug effects , Oxidative Stress/drug effects , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/chemically induced , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Catechols/therapeutic use , Cattle , Disease Progression , Female , Heme Oxygenase-1/metabolism , Inflammation Mediators/metabolism , Janus Kinases/metabolism , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Membrane Proteins/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction/drug effects , RAW 264.7 Cells , Rats , Recombinant Fusion Proteins/drug effects , STAT Transcription Factors/metabolism
9.
Br J Cancer ; 115(8): 940-948, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27575850

ABSTRACT

BACKGROUND: Only a small proportion of patients respond to anti-VEGF therapy, pressing the need for a reliable biomarker that can identify patients who will benefit. We studied the biological activity of anti-VEGF antibodies in patients' blood during anti-VEGF therapy by using the Ba/F3-VEGFR2 cell line, which is dependent on VEGF for its growth. METHODS: Serum samples from 22 patients with cancer before and during treatment with bevacizumab were tested for their effect on proliferation of Ba/F3-VEGFR2 cells. Vascular endothelial growth factor as well as bevacizumab concentrations in serum samples from these patients were determined by enzyme linked immunosorbent assay (ELISA). RESULTS: The hVEGF-driven cell proliferation was effectively blocked by bevacizumab (IC50 3.7 µg ml-1; 95% CI 1.7-8.3 µg ml-1). Cell proliferation was significantly reduced when patients' serum during treatment with bevacizumab was added (22-103% inhibition compared with pre-treatment). Although bevacizumab levels were not related, on-treatment serum VEGF levels were correlated with Ba/F3-VEGFR2 cell proliferation. CONCLUSIONS: We found that the neutralising effect of anti-VEGF antibody therapy on the biological activity of circulating VEGF can be accurately determined with a Ba/F3-VEGFR2 bioassay. The value of this bioassay to predict clinical benefit of anti-VEGF antibody therapy needs further clinical evaluation in a larger randomised cohort.


Subject(s)
Angiogenesis Inhibitors/blood , B-Lymphocytes/drug effects , Bevacizumab/blood , Biological Assay , Enzyme-Linked Immunosorbent Assay , Neoplasms/blood , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Cell Division , Cell Line , Interleukin-3/pharmacology , Mice , Neoplasms/drug therapy , Receptors, Erythropoietin/genetics , Receptors, Interleukin-3/physiology , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Reproducibility of Results , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/immunology
10.
Sci Rep ; 6: 29523, 2016 07 08.
Article in English | MEDLINE | ID: mdl-27389831

ABSTRACT

Abnormal α-synuclein (α-syn) accumulation in the CNS may underlie neuronal cell and synaptic dysfunction leading to motor and cognitive deficits in synucleinopathies including Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB). Multiple groups demonstrated α-syn accumulation in CNS accessory structures, including the eyes and olfactory terminals, as well as in peripheral organs of Parkinsonian patients. Retinal imaging studies of mice overexpressing fused α-syn::GFP were conducted to evaluate the presence and progression of retinal pathology in a PD/DLB transgenic mouse model. Bright-field image retinal maps and fluorescent images were acquired at 1-month intervals for 3 months. Retinal imaging revealed the accumulation of GFP-tagged α-syn in retinal ganglion cell layer and in the edges of arterial blood vessels in the transgenic mice. Double labeling studies confirmed that the α-syn::GFP-positive cells were retinal ganglion cells containing α-syn. Accumulation of α-syn persisted in the same cells and increased with age. Accumulation of α-syn::GFP was reduced by immunization with single chain antibodies against α-syn. In conclusion, longitudinal live imaging of the retina in the PDGF-α-syn::GFP mice might represent a useful, non-invasive tool to monitor the fate of α-syn accumulation in the CNS and to evaluate the therapeutic effects of compounds targeting α-syn.


Subject(s)
Green Fluorescent Proteins/metabolism , Lewy Body Disease/diagnostic imaging , Parkinson Disease/diagnostic imaging , Retina/diagnostic imaging , Single-Chain Antibodies/administration & dosage , alpha-Synuclein/metabolism , Animals , Disease Models, Animal , Disease Progression , Green Fluorescent Proteins/genetics , Humans , Immunotherapy , Lewy Body Disease/metabolism , Lewy Body Disease/therapy , Male , Mice , Mice, Transgenic , Optical Imaging/instrumentation , Parkinson Disease/metabolism , Parkinson Disease/therapy , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Retina/metabolism , Retinal Ganglion Cells/metabolism , Single-Chain Antibodies/pharmacology , alpha-Synuclein/drug effects , alpha-Synuclein/genetics
11.
Clin Pharmacokinet ; 55(9): 1045-58, 2016 09.
Article in English | MEDLINE | ID: mdl-27056734

ABSTRACT

Romiplostim is a novel thrombopoiesis-stimulating peptibody consisting of a carrier Fc domain and a peptide domain that binds to the thrombopoietin receptor (TPOR) on platelets and platelet precursors. Similar to endogenous thrombopoietin, romiplostim activates the TPOR to stimulate the growth and maturation of megakaryocytes, resulting in increased production of platelets in the circulation. Binding of romiplostim to TPOR on the platelets and megakaryocytes presumably triggers subsequent internalization and degradation. Therefore, increased platelet counts following romiplostim treatment results in increased elimination of the drug. The TPOR target-mediated process is saturable, resulting in nonlinear volume of distribution and clearance of romiplostim. Therefore, target-mediated disposition plays a decreasing role in drug elimination with increasing romiplostim serum concentration. Conversely, nonspecific elimination processes such as renal clearance play an increasing role with increasing romiplostim serum concentration. Limited pharmacokinetics data demonstrated that the exposure to romiplostim was lower after multiple dose administrations than after the first dose, although large inter-subject variability was observed. Large inter- and intra-subject variability in the platelet response was also observed at a given dose. These findings suggest considerable heterogeneity of disease in patients with primary immune thrombocytopenia and support the need for individual dose adjustments based on platelet counts.


Subject(s)
Blood Platelets/drug effects , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Receptors, Fc/therapeutic use , Receptors, Thrombopoietin/drug effects , Recombinant Fusion Proteins/pharmacokinetics , Thrombopoietin/pharmacokinetics , Thrombopoietin/therapeutic use , Animals , Blood Platelets/cytology , Dose-Response Relationship, Drug , Humans , Metabolic Clearance Rate , Mice , Mice, Knockout , Models, Biological , Rats , Receptors, Fc/administration & dosage , Receptors, Fc/blood , Receptors, Fc/drug effects , Receptors, Fc/metabolism , Receptors, Thrombopoietin/metabolism , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/blood , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Thrombopoiesis/drug effects , Thrombopoietin/administration & dosage , Thrombopoietin/blood
12.
Proc Natl Acad Sci U S A ; 113(11): 3060-5, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26929355

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting motor neurons that leads to progressive paralysis of skeletal muscle. Studies of ALS have revealed defects in expression of acetylcholine receptors (AChRs) in skeletal muscle that occur even in the absence of motor neuron anomalies. The endocannabinoid palmitoylethanolamide (PEA) modified the clinical conditions in one ALS patient, improving muscle force and respiratory efficacy. By microtransplanting muscle membranes from selected ALS patients into Xenopus oocytes, we show that PEA reduces the desensitization of acetylcholine-evoked currents after repetitive neurotransmitter application (i.e., rundown). The same effect was observed using muscle samples from denervated (non-ALS) control patients. The expression of human recombinant α1ß1γδ (γ-AChRs) and α1ß1εδ AChRs (ε-AChRs) in Xenopus oocytes revealed that PEA selectively affected the rundown of ACh currents in ε-AChRs. A clear up-regulation of the α1 subunit in muscle from ALS patients compared with that from non-ALS patients was found by quantitative PCR, but no differential expression was found for other subunits. Clinically, ALS patients treated with PEA showed a lower decrease in their forced vital capacity (FVC) over time as compared with untreated ALS patients, suggesting that PEA can enhance pulmonary function in ALS. In the present work, data were collected from a cohort of 76 ALS patients and 17 denervated patients. Our results strengthen the evidence for the role of skeletal muscle in ALS pathogenesis and pave the way for the development of new drugs to hamper the clinical effects of the disease.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Ethanolamines/therapeutic use , Molecular Targeted Therapy , Muscle, Skeletal/drug effects , Palmitic Acids/therapeutic use , Receptors, Nicotinic/drug effects , Action Potentials/drug effects , Adult , Aged , Aged, 80 and over , Amides , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Cell Membrane/drug effects , Cell Membrane/transplantation , Ethanolamines/pharmacology , Female , Humans , Male , Microinjections , Middle Aged , Muscle Denervation , Muscle, Skeletal/ultrastructure , Neuromuscular Junction/physiopathology , Oocytes , Palmitic Acids/pharmacology , Receptors, Nicotinic/physiology , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Single-Blind Method , Xenopus laevis
13.
J Clin Invest ; 125(10): 3904-14, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26348896

ABSTRACT

Valproic acid (VPA) has been widely used for decades to treat epilepsy; however, its mechanism of action remains poorly understood. Here, we report that the anticonvulsant effects of nonacute VPA treatment involve preservation of the M-current, a low-threshold noninactivating potassium current, during seizures. In a wide variety of neurons, activation of Gq-coupled receptors, such as the m1 muscarinic acetylcholine receptor, suppresses the M-current and induces hyperexcitability. We demonstrated that VPA treatment disrupts muscarinic suppression of the M-current and prevents resultant agonist-induced neuronal hyperexcitability. We also determined that VPA treatment interferes with M-channel signaling by inhibiting palmitoylation of a signaling scaffold protein, AKAP79/150, in cultured neurons. In a kainate-induced murine seizure model, administration of a dose of an M-channel inhibitor that did not affect kainate-induced seizure transiently eliminated the anticonvulsant effects of VPA. Retigabine, an M-channel opener that does not open receptor-suppressed M-channels, provided anticonvulsant effects only when administered prior to seizure induction in control animals. In contrast, treatment of VPA-treated mice with retigabine induced anticonvulsant effects even when administered after seizure induction. Together, these results suggest that receptor-induced M-current suppression plays a role in the pathophysiology of seizures and that preservation of the M-current during seizures has potential as an effective therapeutic strategy.


Subject(s)
Anticonvulsants/pharmacology , KCNQ2 Potassium Channel/physiology , Valproic Acid/pharmacology , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , A Kinase Anchor Proteins/physiology , Action Potentials/drug effects , Animals , Anthracenes/pharmacology , Anticonvulsants/therapeutic use , Carbamates/pharmacology , Cells, Cultured , Drug Interactions , Female , Hippocampus/cytology , Humans , KCNQ2 Potassium Channel/drug effects , Kainic Acid/toxicity , Lipoylation/drug effects , Male , Mice , Mice, Inbred C57BL , Muscarinic Agonists/pharmacology , Muscarinic Antagonists/pharmacology , Neurons/drug effects , Neurons/metabolism , Phenylenediamines/pharmacology , Phosphorylation/drug effects , Potassium Channel Blockers/pharmacology , Protein Processing, Post-Translational/drug effects , Rats , Receptor, Muscarinic M1/drug effects , Receptor, Muscarinic M1/physiology , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Seizures/chemically induced , Seizures/drug therapy , Seizures/physiopathology , Signal Transduction/drug effects , Superior Cervical Ganglion/cytology , Valproic Acid/therapeutic use
14.
J Mol Biol ; 427(1): 176-89, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-24862283

ABSTRACT

The N-methyl-d-aspartate receptors (NMDARs) constitute an important class of ligand-gated cation channels that are involved in the majority of excitatory neurotransmission in the human brain. Compounds that bind in the NMDAR ion channel and act as blockers are use- and voltage-dependent inhibitors of NMDAR activity and have therapeutic potential for treatment of a variety of brain diseases or as pharmacological tools for studies of the neurobiological role of NMDARs. We have performed a kinetic analysis of the blocking mechanism of the prototypical polyamine toxin NMDAR ion channel blocker argiotoxin-636 (ArgTX-636) at recombinant GluN1/2A receptors to provide detailed information on the mechanism of block. The predicted binding site of ArgTX-636 is in the pore region of the NMDAR ion channel formed by residues in the transmembrane M3 and the M2 pore-loop segments of the GluN1 and GluN2A subunits. To assess the predicted binding mode in further detail, we performed an alanine- and glycine-scanning mutational analysis of this pore-loop segment to systematically probe the role of pore-lining M2 residues in GluN1 and GluN2A in the channel block by ArgTX-636. Comparison of M2 positions in GluN1 and GluN2A where mutation influences ArgTX-636 potency suggests differential contribution of the M2-loops of GluN1 and GluN2A to binding of ArgTX-636. The results of the mutational analysis are highly relevant for the future structure-based development of argiotoxin-derived NMDAR channel blockers.


Subject(s)
Glutamic Acid/metabolism , Indoleacetic Acids/pharmacology , Polyamines/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Recombinant Fusion Proteins/metabolism , Spider Venoms/pharmacology , Animals , Binding Sites , Electrophysiology , HEK293 Cells , Humans , Ion Channels/drug effects , Ion Channels/metabolism , Kinetics , Models, Molecular , Mutagenesis , Mutation/genetics , Patch-Clamp Techniques , Protein Conformation , Protein Subunits , Rats , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Fusion Proteins/drug effects
15.
Enzyme Microb Technol ; 55: 26-30, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24411442

ABSTRACT

Aquaporin Z (AqpZ) is a water channel protein from Escherichia coli and has attracted many attentions to develop the biomimetic water filtration technology. Cell-free protein synthesis (CFPS) system, one of the most complex multi-enzymatic systems, has the ability of producing the integral membrane protein in vitro. To enhance the synthesis of AqpZ in E. coli cell-free system, several natural leader peptides were respectively fused at the N-terminus and were verified to enhance the expression level significantly. Moreover, the supplementation of detergents or liposome could activate leader peptidase from the cell-free extract and provide hydrophobic environment for proper folding of AqpZ. Thus, the release of mature AqpZ via the in situ removal of leader peptide was achieved, with a specific water transport activity of (2.1 ± 0.1) × 10⁻¹4 cm³ s⁻¹ monomer⁻¹. Using this in situ removable leader peptide strategy, the transcription-translation, leader sequence cleavage and membrane protein folding were integrated into a simple process in the cell-free system, providing a convenient approach to enhance the expression of target proteins, especially those membrane proteins difficult to achieve.


Subject(s)
Aquaporins/biosynthesis , Escherichia coli Proteins/biosynthesis , Escherichia coli/enzymology , Protein Sorting Signals/genetics , Amino Acid Sequence , Aquaporins/genetics , Aquaporins/isolation & purification , Cell-Free System , Detergents/pharmacology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Gene Expression Regulation, Enzymologic , Genes, Synthetic , Genetic Vectors , Liposomes , Membrane Proteins/metabolism , Molecular Sequence Data , Protein Biosynthesis , Protein Sorting Signals/physiology , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/isolation & purification , Serine Endopeptidases/metabolism , Transcription, Genetic , Water/metabolism
16.
Metab Brain Dis ; 29(4): 983-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24136581

ABSTRACT

At high concentrations, the glutamine synthetase inhibitor L-methionine-S,R-sulfoximine (MSO) is a convulsant, especially in dogs. Nevertheless, sub-convulsive doses of MSO are neuroprotective in rodent models of hyperammonemia, acute liver disease, and amyotrophic lateral sclerosis and suggest MSO may be clinically useful. Previous work has also shown that much lower doses of MSO are required to produce convulsions in dogs than in primates. Evidence from the mid-20th century suggests that humans are also less sensitive. In the present work, the inhibition of recombinant human glutamine synthetase by MSO is shown to be biphasic-an initial reversible competitive inhibition (K i 1.19 mM) is followed by rapid irreversible inactivation. This K i value for the human enzyme accounts, in part, for relative insensitivity of primates to MSO and suggests that this inhibitor could be used to safely inhibit glutamine synthetase activity in humans.


Subject(s)
Convulsants/pharmacology , Enzyme Inhibitors/pharmacology , Glutamate-Ammonia Ligase/antagonists & inhibitors , Methionine Sulfoximine/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nervous System Diseases/drug therapy , Adenosine Triphosphate/metabolism , Amyotrophic Lateral Sclerosis/drug therapy , Animals , Bacterial Proteins/antagonists & inhibitors , Dogs/metabolism , Humans , Kinetics , Nervous System Diseases/enzymology , Plant Proteins/antagonists & inhibitors , Protein Binding , Recombinant Fusion Proteins/drug effects , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Sheep/metabolism , Species Specificity
18.
Protein Expr Purif ; 92(2): 156-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24084007

ABSTRACT

Dengue virus (DENV), a member of the flavivirus genus, affects 50-100 million people in tropical and sub-tropical regions. The DENV protease domain is located at the N-terminus of the NS3 protease and requires for its enzymatic activity a hydrophilic segment of the NS2B that acts as a cofactor. The protease is an important antiviral drug target because it plays a crucial role in virus replication by cleaving the genome-coded polypeptide into mature functional proteins. Currently, there are no drugs to inhibit DENV protease activity. Most structural and functional studies have been conducted using protein constructs containing the NS3 protease domain connected to a soluble segment of the NS2B membrane protein via a nine-residue linker. For in vitro structural and functional studies, it would be useful to produce a natural form of the DENV protease containing the NS3 protease domain and the full-length NS2B protein. Herein, we describe the expression and purification of a natural form of DENV protease (NS2BFL-NS3pro) containing the full-length NS2B protein and the protease domain of NS3 (NS3pro). The protease was expressed and purified in detergent micelles necessary for its folding. Our results show that this purified protein was active in detergent micelles such as lyso-myristoyl phosphatidylcholine (LMPC). These findings should facilitate further structural and functional studies of the protease and will facilitate drug discovery targeting DENV.


Subject(s)
Lysophosphatidylcholines/pharmacology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/drug effects , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/drug effects , Amino Acid Sequence , Detergents/chemistry , Enzyme Stability , Lysophosphatidylcholines/chemistry , Micelles , Molecular Sequence Data , Protein Structure, Tertiary , RNA Helicases/chemistry , RNA Helicases/drug effects , RNA Helicases/genetics , RNA Helicases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/drug effects , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
19.
Neurotoxicology ; 35: 121-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23313730

ABSTRACT

Chronic exposure to Mn results in the development of a neurological disorder known as manganism characterized by neurological deficits resembling that seen in Parkinsonism. Although dopaminergic neurons within the nigrostriatal pathway appear intact, Mn-induced irregularities in DA transmission have been observed including decreased amphetamine-induced DA release and loss of the dopamine transporter (DAT). Results of studies to evaluate the effect of Mn and DA on cell viability in control and DAT-transfected HEK cells reveal that Mn is equally toxic to both cell lines whereas DA was only toxic to cells containing DAT. DA toxicity was saturable suggesting that transport may be rate limiting. When Mn and DA were added simultaneously to the media, cell toxicity was similar to that produced by Mn alone suggesting that Mn may suppress DA uptake in the DAT containing cells. Preincubation of DA prior to the addition of Mn resulted in cell death which was essentially additive with that produced independently by the two agents. Mn was also shown to decrease DA uptake and amphetamine-induced DA efflux in DAT containing cells. Time-lapsed confocal microscopy indicates that Mn can promote trafficking of cell surface DAT into intracellular compartments which may account for the decrease in DA uptake and DA efflux in these cells. Mn-induced internalization of DAT may provide an explanation for disruption in DA transmission previously reported in the striatum.


Subject(s)
Chlorides/toxicity , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine/toxicity , Manganese Poisoning/etiology , Amphetamine/pharmacology , Cell Survival/drug effects , Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Manganese Compounds , Manganese Poisoning/genetics , Manganese Poisoning/metabolism , Manganese Poisoning/pathology , Membrane Potentials , Microscopy, Confocal , Protein Transport , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Time Factors , Time-Lapse Imaging , Transfection
20.
Proc Natl Acad Sci U S A ; 109(9): 3552-7, 2012 Feb 28.
Article in English | MEDLINE | ID: mdl-22331907

ABSTRACT

Large-conductance voltage- and Ca(2+)-activated K(+) (Slo1 BK) channels serve numerous cellular functions, and their dysregulation is implicated in various diseases. Drugs activating BK channels therefore bear substantial therapeutic potential, but their deployment has been hindered in part because the mode of action remains obscure. Here we provide mechanistic insight into how the dehydroabietic acid derivative Cym04 activates BK channels. As a representative of NS1619-like BK openers, Cym04 reversibly left-shifts the half-activation voltage of Slo1 BK channels. Using an established allosteric BK gating model, the Cym04 effect can be simulated by a shift of the voltage sensor and the ion conduction gate equilibria toward the activated and open state, respectively. BK activation by Cym04 occurs in a splice variant-specific manner; it does not occur in such Slo1 BK channels using an alternative neuronal exon 9, which codes for the linker connecting the transmembrane segment S6 and the cytosolic RCK1 domain--the S6/RCK linker. In addition, Cym04 does not affect Slo1 BK channels with a two-residue deletion within this linker. Mutagenesis and model-based gating analysis revealed that BK openers, such as Cym04 and NS1619 but not mallotoxin, activate BK channels by functionally interacting with the S6/RCK linker, mimicking site-specific shortening of this purported passive spring, which transmits force from the cytosolic gating ring structure to open the channel's gate.


Subject(s)
Abietanes/pharmacology , Ion Channel Gating/drug effects , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/drug effects , Potassium/metabolism , Allosteric Regulation , Amino Acid Sequence , HEK293 Cells , Humans , Ion Channel Gating/physiology , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/chemistry , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/genetics , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Membrane Potentials , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Patch-Clamp Techniques , Protein Conformation , Protein Isoforms/chemistry , Protein Isoforms/drug effects , Protein Isoforms/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...