Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Drug Discov Today ; 28(10): 103732, 2023 10.
Article in English | MEDLINE | ID: mdl-37541423

ABSTRACT

External innovation initiatives in the pharmaceutical industry have become an integral part of research and development. Collaborations have been built to enhance innovation, mitigate risk, and share cost, especially for neurodegenerative diseases, a therapeutic area that has suffered from high attrition rates. This article outlines the Eisai-University College London (UCL) Drug Discovery and Development Collaboration as a case study of how to implement a productive industry-academic partnership. In the first 10 years, seven projects have been established and the first project, a novel anti-tau antibody for Alzheimer's disease, has entered clinical trials, providing early validation of this collaboration model.


Subject(s)
Alzheimer Disease , Drug Discovery , Humans , Universities , London , Alzheimer Disease/drug therapy , Drug Industry
2.
Bioorg Med Chem Lett ; 81: 129130, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36640928

ABSTRACT

Glucocerebrosidase (GCase) is a lysosomal enzyme encoded by the GBA1 gene, loss of function variants of which cause an autosomal recessive lysosomal storage disorder, Gaucher disease (GD). Heterozygous variants of GBA1 are also known as the strongest common genetic risk factor for Parkinson's disease (PD). Restoration of GCase enzymatic function using a pharmacological chaperone strategy is considered a promising therapeutic approach for PD and GD. We identified compound 4 as a GCase pharmacological chaperone with sub-micromolar activity from a high-throughput screening (HTS) campaign. Compound 4 was further optimised to ER-001230194 (compound 25). ER-001230194 shows improved ADME and physicochemical properties and therefore represents a novel pharmacological chaperone with which to investigate GCase pharmacology further.


Subject(s)
Gaucher Disease , Parkinson Disease , Humans , Glucosylceramidase/genetics , Mutation , Parkinson Disease/drug therapy , Gaucher Disease/drug therapy , Lysosomes
3.
SLAS Discov ; 28(3): 73-87, 2023 04.
Article in English | MEDLINE | ID: mdl-36608804

ABSTRACT

Mitochondrial dysfunction and aberrant mitochondrial homeostasis are key aspects of Parkinson's disease (PD) pathophysiology. Mutations in PINK1 and Parkin proteins lead to autosomal recessive PD, suggesting that defective mitochondrial clearance via mitophagy is key in PD etiology. Accelerating the identification and/or removal of dysfunctional mitochondria could therefore provide a disease-modifying approach to treatment. To that end, we performed a high-content phenotypic screen (HCS) of ∼125,000 small molecules to identify compounds that positively modulate mitochondrial accumulation of the PINK1-Parkin-dependent mitophagy initiation marker p-Ser65-Ub in Parkin haploinsufficiency (Parkin +/R275W) human fibroblasts. Following confirmatory counter-screening and orthogonal assays, we selected compounds of interest that enhance mitophagy-related biochemical and functional endpoints in patient-derived fibroblasts. Identification of inhibitors of the ubiquitin-specific peptidase and negative regulator of mitophagy USP30 within our hits further validated our approach. The compounds identified in this work provide a novel starting point for further investigation and optimization.


Subject(s)
Mitophagy , Parkinson Disease , Humans , Mitophagy/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Ubiquitination/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Mutation , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
4.
Biochem J ; 478(23): 4099-4118, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34704599

ABSTRACT

Mitochondrial dysfunction is implicated in Parkinson disease (PD). Mutations in Parkin, an E3 ubiquitin ligase, can cause juvenile-onset Parkinsonism, probably through impairment of mitophagy. Inhibition of the de-ubiquitinating enzyme USP30 may counter this effect to enhance mitophagy. Using different tools and cellular approaches, we wanted to independently confirm this claimed role for USP30. Pharmacological characterisation of additional tool compounds that selectively inhibit USP30 are reported. The consequence of USP30 inhibition by these compounds, siRNA knockdown and overexpression of dominant-negative USP30 on the mitophagy pathway in different disease-relevant cellular models was explored. Knockdown and inhibition of USP30 showed increased p-Ser65-ubiquitin levels and mitophagy in neuronal cell models. Furthermore, patient-derived fibroblasts carrying pathogenic mutations in Parkin showed reduced p-Ser65-ubiquitin levels compared with wild-type cells, levels that could be restored using either USP30 inhibitor or dominant-negative USP30 expression. Our data provide additional support for USP30 inhibition as a regulator of the mitophagy pathway.


Subject(s)
Mitochondrial Proteins/metabolism , Mitophagy , Parkinson Disease/metabolism , Protein Kinases/metabolism , Thiolester Hydrolases/metabolism , Ubiquitin-Protein Ligases/metabolism , Cell Line , Fibroblasts , Humans
5.
Sci Rep ; 11(1): 13316, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172778

ABSTRACT

The R47H variant of the microglial membrane receptor TREM2 is linked to increased risk of late onset Alzheimer's disease. Human induced pluripotent stem cell derived microglia (iPS-Mg) from patient iPSC lines expressing the AD-linked R47Hhet TREM2 variant, common variant (Cv) or an R47Hhom CRISPR edited line and its isogeneic control, demonstrated that R47H-expressing iPS-Mg expressed a deficit in signal transduction in response to the TREM2 endogenous ligand phosphatidylserine with reduced pSYK-pERK1/2 signalling and a reduced NLRP3 inflammasome response, (including ASC speck formation, Caspase-1 activation and IL-1beta secretion). Apoptotic cell phagocytosis and soluble TREM2 shedding were unaltered, suggesting a disjoint between these pathways and the signalling cascades downstream of TREM2 in R47H-expressing iPS-Mg, whilst metabolic deficits in glycolytic capacity and maximum respiration were reversed when R47H expressing iPS-Mg were exposed to PS+ expressing cells. These findings suggest that R47H-expressing microglia are unable to respond fully to cell damage signals such as phosphatidylserine, which may contribute to the progression of neurodegeneration in late-onset AD.


Subject(s)
Alzheimer Disease/metabolism , Inflammasomes/metabolism , Membrane Glycoproteins/metabolism , Microglia/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/physiology , Brain/metabolism , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Phagocytosis/physiology
6.
Acta Neuropathol Commun ; 8(1): 13, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32019610

ABSTRACT

Tau deposition in the brain is a pathological hallmark of many neurodegenerative disorders, including Alzheimer's disease (AD). During the course of these tauopathies, tau spreads throughout the brain via synaptically-connected pathways. Such propagation of pathology is thought to be mediated by tau species ("seeds") containing the microtubule binding region (MTBR) composed of either three repeat (3R) or four repeat (4R) isoforms. The tau MTBR also forms the core of the neuropathological filaments identified in AD brain and other tauopathies. Multiple approaches are being taken to limit tau pathology, including immunotherapy with anti-tau antibodies. Given its key structural role within fibrils, specifically targetting the MTBR with a therapeutic antibody to inhibit tau seeding and aggregation may be a promising strategy to provide disease-modifying treatment for AD and other tauopathies. Therefore, a monoclonal antibody generating campaign was initiated with focus on the MTBR. Herein we describe the pre-clinical generation and characterisation of E2814, a humanised, high affinity, IgG1 antibody recognising the tau MTBR. E2814 and its murine precursor, 7G6, as revealed by epitope mapping, are antibodies bi-epitopic for 4R and mono-epitopic for 3R tau isoforms because they bind to sequence motif HVPGG. Functionally, both antibodies inhibited tau aggregation in vitro. They also immunodepleted a variety of MTBR-containing tau protein species. In an in vivo model of tau seeding and transmission, attenuation of deposition of sarkosyl-insoluble tau in brain could also be observed in response to antibody treatment. In AD brain, E2814 bound different types of tau filaments as shown by immunogold labelling and recognised pathological tau structures by immunohistochemical staining. Tau fragments containing HVPGG epitopes were also found to be elevated in AD brain compared to PSP or control. Taken together, the data reported here have led to E2814 being proposed for clinical development.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/therapy , Antibodies, Monoclonal/immunology , Immunization, Passive/methods , tau Proteins/genetics , tau Proteins/immunology , Alzheimer Disease/pathology , Animals , Antibodies, Monoclonal/pharmacology , Frontal Lobe/metabolism , Frontal Lobe/pathology , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Male , Mice, Transgenic , Protein Aggregation, Pathological/immunology , Protein Isoforms/immunology , Protein Isoforms/pharmacology
8.
Sci Rep ; 7(1): 10492, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874733

ABSTRACT

Mitochondrial Ca2+ uptake has a key role in cellular Ca2+ homeostasis. Excessive matrix Ca2+ concentrations, especially when coincident with oxidative stress, precipitate opening of an inner mitochondrial membrane, high-conductance channel: the mitochondrial permeability transition pore (mPTP). mPTP opening has been implicated as a final cell death pathway in numerous diseases and therefore understanding conditions dictating mPTP opening is crucial for developing targeted therapies. Here, we have investigated the impact of mitochondrial metabolic state on the probability and consequences of mPTP opening. Isolated mitochondria were energised using NADH- or FADH2-linked substrates. The functional consequences of Ca2+-induced mPTP opening were assessed by Ca2+ retention capacity, using fluorescence-based analysis, and simultaneous measurements of mitochondrial Ca2+ handling, membrane potential, respiratory rate and production of reactive oxygen species (ROS). Succinate-induced, membrane potential-dependent reverse electron transfer sensitised mitochondria to mPTP opening. mPTP-induced depolarisation under succinate subsequently inhibited reverse electron transfer. Complex I-driven respiration was reduced after mPTP opening but sustained in the presence of complex II-linked substrates, consistent with inhibition of complex I-supported respiration by leakage of matrix NADH. Additionally, ROS generated at complex III did not sensitise mitochondria to mPTP opening. Thus, cellular metabolic fluxes and metabolic environment dictate mitochondrial functional response to Ca2+ overload.


Subject(s)
Ion Channel Gating , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/physiology , Animals , Calcium/metabolism , Cell Respiration , Electron Transport Complex I/metabolism , Energy Metabolism , Female , Hydrogen Peroxide/metabolism , Membrane Potential, Mitochondrial , Mitochondrial Permeability Transition Pore , NAD/metabolism , Oxygen Consumption , Rats , Reactive Oxygen Species/metabolism , Succinic Acid/metabolism
9.
Sci Rep ; 6: 37798, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27886240

ABSTRACT

Growing evidence suggests persistent mitochondrial permeability transition pore (mPTP) opening is a key pathophysiological event in cell death underlying a variety of diseases. While it has long been clear the mPTP is a druggable target, current agents are limited by off-target effects and low therapeutic efficacy. Therefore identification and development of novel inhibitors is necessary. To rapidly screen large compound libraries for novel mPTP modulators, a method was exploited to cryopreserve large batches of functionally active mitochondria from cells and tissues. The cryopreserved mitochondria maintained respiratory coupling and ATP synthesis, Ca2+ uptake and transmembrane potential. A high-throughput screen (HTS), using an assay of Ca2+-induced mitochondrial swelling in the cryopreserved mitochondria identified ER-000444793, a potent inhibitor of mPTP opening. Further evaluation using assays of Ca2+-induced membrane depolarisation and Ca2+ retention capacity also indicated that ER-000444793 acted as an inhibitor of the mPTP. ER-000444793 neither affected cyclophilin D (CypD) enzymatic activity, nor displaced of CsA from CypD protein, suggesting a mechanism independent of CypD inhibition. Here we identified a novel, CypD-independent inhibitor of the mPTP. The screening approach and compound described provides a workflow and additional tool to aid the search for novel mPTP modulators and to help understand its molecular nature.


Subject(s)
Cryopreservation , Cyclophilins/physiology , High-Throughput Screening Assays/methods , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Quinolines/pharmacology , Adenosine Triphosphate/biosynthesis , Animals , Peptidyl-Prolyl Isomerase F , Energy Metabolism , Female , HeLa Cells , Humans , Mitochondria/metabolism , Mitochondrial Permeability Transition Pore , Rats , Rats, Sprague-Dawley
10.
Hum Mol Genet ; 21(19): 4201-13, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22736029

ABSTRACT

The G2019S leucine rich repeat kinase 2 (LRRK2) mutation is the most common genetic cause of Parkinson's disease (PD), clinically and pathologically indistinguishable from idiopathic PD. Mitochondrial abnormalities are a common feature in PD pathogenesis and we have investigated the impact of G2019S mutant LRRK2 expression on mitochondrial bioenergetics. LRRK2 protein expression was detected in fibroblasts and lymphoblasts at levels higher than those observed in the mouse brain. The presence of G2019S LRRK2 mutation did not influence LRRK2 expression in fibroblasts. However, the expression of the G2019S LRRK2 mutation in both fibroblast and neuroblastoma cells was associated with mitochondrial uncoupling. This was characterized by decreased mitochondrial membrane potential and increased oxygen utilization under basal and oligomycin-inhibited conditions. This resulted in a decrease in cellular ATP levels consistent with compromised cellular function. This uncoupling of mitochondrial oxidative phosphorylation was associated with a cell-specific increase in uncoupling protein (UCP) 2 and 4 expression. Restoration of mitochondrial membrane potential by the UCP inhibitor genipin confirmed the role of UCPs in this mechanism. The G2019S LRRK2-induced mitochondrial uncoupling and UCP4 mRNA up-regulation were LRRK2 kinase-dependent, whereas endogenous LRRK2 levels were required for constitutive UCP expression. We propose that normal mitochondrial function was deregulated by the expression of G2019S LRRK2 in a kinase-dependent mechanism that is a modification of the normal LRRK2 function, and this leads to the vulnerability of selected neuronal populations in PD.


Subject(s)
Membrane Transport Proteins/metabolism , Mitochondria/enzymology , Mutation, Missense , Parkinson Disease/enzymology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism
11.
Neurosci Lett ; 444(3): 245-9, 2008 Oct 31.
Article in English | MEDLINE | ID: mdl-18761055

ABSTRACT

A method to evaluate kinase inhibitor action was reported [L. Morgan, S.J. Neame, H. Child, R. Chung, B. Shah, L. Barden, J.M. Staddon, T.R. Patel, Development of a pentylenetetrazole-induced seizure model to evaluate kinase inhibitor efficacy in the central nervous system, Neurosci. Lett. 395 (2006) 143-148]. In this, acute administration of the GABA antagonist pentylenetetrazole triggers seizures through glutamate-dependent pathways. Under such conditions, activation of the c-Jun N-terminal kinase (JNK) pathway was detected in hippocampal extracts. Phosphorylation of the upstream JNK kinase MKK4 was also revealed through use of a phospho-MKK4-specific antibody. Here, this antibody is shown to also react with a protein of approximately 125 kDa which underwent increased phosphorylation in response to pentylenetetrazole treatment. The present study aimed to identify the approximately 125 kDa protein as it may provide novel insight into signalling, neuronal activity and seizures. Using chromatographic methods and mass spectrometry, the protein was identified as amphiphysin I. This was confirmed by 2D gel analysis and immunoblot with amphiphysin I-specific antibodies. Although the phospho-MKK4 antibody was raised against an MKK4-specific peptide, partial sequence homology between this sequence and a region of amphiphysin was discerned. New antibodies raised against the phospho-threonine 260-amphiphysin-specific sequence detected increased phosphorylation in response to pentylenetetrazole treatment. This particular phosphorylation site does not seem to have been described before, possibly reflecting a novel regulatory aspect of amphiphysin biology. As amphiphysin is involved in the regulation of endocytosis, phosphorylation at this site may play a role in the regulated re-uptake of synaptic vesicles after neurotransmitter release.


Subject(s)
GABA Antagonists , Nerve Tissue Proteins/metabolism , Pentylenetetrazole , Seizures/metabolism , Threonine/metabolism , Animals , Hippocampus/metabolism , MAP Kinase Kinase 4/metabolism , Mice , Phosphorylation , Seizures/chemically induced
12.
Neurosci Lett ; 395(2): 143-8, 2006 Mar 06.
Article in English | MEDLINE | ID: mdl-16300886

ABSTRACT

c-Jun N-terminal kinases (JNKs) are implicated in cell death in neurodegenerative disorders. Therefore, JNK inhibitors could act as neuroprotective agents. To evaluate potential candidates, reproducible and quantitative CNS in vivo models are required. To that end, a pentylenetetrazole-induced seizure model was explored. c-Jun phosphorylation was detected in hippocampal extracts by blotting c-Jun immunoprecipitates with phosphorylation-specific antibodies. Pentylenetetrazole administration induced rapid and reproducible increases in c-Jun phosphorylation. However, special attention had to be paid to the composition of the extraction buffer to ensure stabilization of protein phosphorylation, as demonstrated using internal standards of phosphorylated recombinant c-Jun. As JNK and its upstream activator MKK4 are activated by phosphorylation, these events were also evaluated. In principle, kinase inhibitors could act at the level of JNK or upstream kinases to inhibit c-Jun phosphorylation. MKK4 phosphorylation was dramatically increased in response to pentylenetetrazole but, again, only when appropriate phosphatase inhibitors were in the extraction buffer. In contrast, JNK was found to be constitutively phosphorylated and unaltered upon pentylenetetrazole treatment. The JNK inhibitor SP600125 was shown to inhibit c-Jun phosphorylation without affecting MKK4 phosphorylation. Our procedures enable analysis of JNK pathway signalling in a CNS model and, also, should be applicable to that of other protein phosphorylation events in vivo.


Subject(s)
Convulsants/toxicity , Disease Models, Animal , Hippocampus/metabolism , MAP Kinase Kinase 4/metabolism , Pentylenetetrazole/toxicity , Seizures/chemically induced , Animals , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Enzyme Activation/physiology , Immunoblotting , Immunohistochemistry , MAP Kinase Kinase 4/drug effects , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-jun/drug effects , Proto-Oncogene Proteins c-jun/metabolism
13.
Bioorg Med Chem Lett ; 15(21): 4666-70, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16153829

ABSTRACT

Imidazole-based structures of p38 inhibitors served as a starting point for the design of JNK3 inhibitors. Construction of a 6,7-dihydro-5H-pyrrolo[1,2-a]imidazole scaffold led to the synthesis of the (S)-enantiomers, which exhibited p38/JNK3 IC50 ratio of up to 10 and were up to 20 times more potent inhibitors of JNK3 than the relevant (R)-enantiomers. The JNK3 inhibitory potency correlated well with inhibition of c-Jun phosphorylation and neuroprotective properties of the compounds in low K+-induced cell death of rat cerebellar granule neurones.


Subject(s)
Mitogen-Activated Protein Kinase 10/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Animals , Cell Death/drug effects , Cerebellum/cytology , Imidazoles , Neurons/cytology , Neurons/drug effects , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Rats , Stereoisomerism , Structure-Activity Relationship , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
14.
J Cell Sci ; 116(Pt 23): 4777-90, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14600263

ABSTRACT

Death-associated protein kinase (DAP kinase) is a proapoptotic, calcium/calmodulin-dependent serine/threonine kinase. Here, we report that DAP kinase phosphorylates the regulatory light chain of myosin II (MLC) both in vitro and in vivo, and that this phosphorylation occurs preferentially at residue Ser19. In quiescent fibroblasts, DAP kinase stabilizes stress fibers through phosphorylation of MLC, but it is dispensable for the formation of peripheral microfilament bundles. This cytoskeletal effect of DAP kinase occurs before the onset of apoptosis and does not require an intact death domain. In addition, DAP kinase is required for serum-induced stress-fiber formation, which is associated with the upregulation of its catalytic activity. Despite being both sufficient and necessary for the assembly or maintenance of stress fibers, DAP kinase is incapable of stimulating the formation of focal adhesions in quiescent cells. Moreover, it promotes the disassembly of focal adhesions but not stress fibers in cells receiving serum factors. Together, our results identify a novel and unique function of DAP kinase in the uncoupling of stress fibers and focal adhesions. Such uncoupling would lead to a perturbation of the balance between contractile and adhesion forces and subsequent cell detachment, which might contribute to its pro-apoptotic activity.


Subject(s)
Actin Cytoskeleton/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Focal Adhesions/metabolism , Myosin Light Chains/metabolism , Myosin Type II/metabolism , Stress Fibers/metabolism , 3T3 Cells , Actins/metabolism , Animals , Apoptosis Regulatory Proteins , Baculoviridae , Calmodulin , Cells, Cultured , Cloning, Molecular , Death-Associated Protein Kinases , Humans , Mice , Microscopy, Fluorescence , Microscopy, Interference , Phosphorylation , Protein Binding , Protein Structure, Tertiary
15.
Anal Biochem ; 322(2): 170-8, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14596824

ABSTRACT

Members of the Bcl-2 family are critical regulators of apoptosis. Antiapoptotic family proteins such as Bcl-2 and Bcl-x(L) function, at least in part, by binding proapoptotic members such as Bax and Bak and thereby preventing release of apoptotic proteins, including cytochrome c, from the mitochondria. "BH3-only" members of the family disrupt this interaction by binding, via their BH3 domain, to a hydrophobic pocket on the surface of the antiapoptotic members. Disruption of heterodimerizations by small-molecule inhibitors could be used to modulate cell death in both cancer (to increase apoptosis) and degenerative disorders (to decrease apoptosis), and assays are necessary to screen compound libraries. Fluorescence polarization and enzyme-linked immunosorbent assay-based methods to detect Bcl-2 protein interactions have been described. Here, two further methods that are rapid, "mix and read," homogeneous reactions, insensitive to compound autofluorescence, and amenable to high-throughput screening, are described: a scintillation proximity assay and a time-resolved fluorescence resonance energy transfer assay (HTRF). The assays are designed using tags such that different Bcl-2 family members or BH3 domain peptides can be readily applied to either format, as exemplified by the use here of histidine-tagged Bcl-x(L) and biotinylated BH3 peptides.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/metabolism , Scintillation Counting/methods , Amino Acid Sequence , Binding, Competitive , Biotinylation , Chelating Agents/chemistry , Copper/chemistry , DNA-Binding Proteins/metabolism , Dimerization , Histidine/genetics , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Mapping , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT3 Transcription Factor , Trans-Activators/metabolism , bcl-X Protein
16.
Infect Immun ; 71(9): 5188-93, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12933863

ABSTRACT

Cytotoxic necrotizing factor 1 (CNF-1) is an exotoxin of Escherichia coli that constitutively activates the GTPases Rho, Rac, and CDC42. Stimulation of Rho was shown to enhance myosin light-chain (MLC) phosphorylation via Rho kinase-mediated inhibition of MLC phosphatase in endothelial cells. Here we report that 3 h after CNF stimulation of endothelial cells, RhoA was activated and MLC phosphorylation was increased in a Rho/Rho-kinase-dependent manner, but no decrease in MLC phosphatase activity could be detected. Despite continuous RhoA activation, MLC phosphatase activity was doubled after 24 h of CNF stimulation, and this coincided with decreased MLC phosphorylation and cell spreading. Rac was also activated at 3 to 24 h but did not contribute to MLC phosphorylation, and its amount gradually decreased in the CNF-stimulated cells. CDC42Hs was not activated above control values by CNF. These results suggest that CNF can induce specific decoupling (Rho kinase from MLC phosphatase) and deactivation events in Rho GTPase signaling, potentially reflecting cellular protection mechanisms against permanently active Rho GTPases.


Subject(s)
Bacterial Toxins/toxicity , Cytotoxins/toxicity , Escherichia coli Proteins , Escherichia coli/pathogenicity , Myosin Light Chains/metabolism , Actomyosin/metabolism , Bacterial Toxins/genetics , Base Sequence , Cells, Cultured , Cytotoxins/genetics , DNA, Bacterial/genetics , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Escherichia coli/genetics , Humans , Intracellular Signaling Peptides and Proteins , Kinetics , Myosin-Light-Chain Phosphatase , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/toxicity , cdc42 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases
17.
FEBS Lett ; 545(2-3): 161-6, 2003 Jun 19.
Article in English | MEDLINE | ID: mdl-12804768

ABSTRACT

In epithelial and endothelial cells, tight junctions limit paracellular flux of ions, proteins and other macromolecules. However, mechanisms regulating tight junction function are not clear. Occludin, a tight junction protein, undergoes phosphorylation changes in several situations but little is known about occludin kinases. A recombinant C-terminal fragment of occludin is a substrate for a kinase in crude extracts of brain. This activity was purified about 10000-fold and identified as CK2 (casein kinase 2) by peptide mass fingerprinting, immunoblotting and mutation of CK2 sites within the occludin sequence. CK2 is therefore a candidate kinase for regulation of occludin phosphorylation in vivo.


Subject(s)
Brain/enzymology , Cell Extracts/chemistry , Membrane Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Casein Kinase II , Cell Fractionation , Mutagenesis, Site-Directed , Occludin , Phosphorylation , Point Mutation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/isolation & purification , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Swine
18.
Am J Physiol Lung Cell Mol Physiol ; 283(3): L596-603, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12169579

ABSTRACT

Reactive oxygen species (ROS) increase the contractile response of airway smooth muscle (ASM). Heme oxygenase (HO) catabolizes heme to the powerful antioxidant bilirubin. Because HO is expressed in the airways, we investigated its effects on ASM contractility and ROS production in guinea pig trachea. HO expression was higher in the epithelium than in tracheal smooth muscle. Incubation of tracheal rings (TR) with the HO inhibitor tin protoporphyrin (SnPP IX) or the HO substrate hemin increased and decreased, respectively, ASM contractile response to carbamylcholine. The effect of hemin was reversed by SnPP and mimicked by the antioxidants superoxide dismutase (SOD) and catalase. Hemin significantly reduced the effect of carbamylcholine in rings treated with the guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo-[4,3-a]quinoxalin-1-one (ODQ), compared with ODQ-treated rings without hemin incubation, suggesting that the CO-guanosine 3',5'-cyclic monophosphate pathway was not involved in the control of tracheal reactivity. SnPP and hemin increased and decreased ROS production by TR by 18 and 38%, respectively. Bilirubin (100 pM) significantly decreased TR contractility and ROS production. Hemin, bilirubin, and SOD/catalase decreased phosphorylation of the contractile protein myosin light chain, whereas SnPP significantly augmented it. These data suggest that modulation of the redox status by HO and, moreover, by bilirubin modulates ASM contractility by modulating levels of phosphorylated myosin light chain.


Subject(s)
Bilirubin/physiology , Heme Oxygenase (Decyclizing)/physiology , Muscle Contraction/physiology , Muscle, Smooth/physiology , Oxidants/metabolism , Trachea/physiology , Animals , Carbon Monoxide/metabolism , Cyclic GMP/metabolism , Epithelium/metabolism , Guinea Pigs , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , In Vitro Techniques , Male , Myosin Light Chains/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...