Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
2.
PLOS Digit Health ; 1(8): e0000083, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36812621

ABSTRACT

OBJECTIVE: Cognitive-behavioral therapy (CBT) has growing evidence of efficacy for Attention-Deficit/Hyperactivity Disorder (ADHD) in adults. Mobile health apps are promising tools for delivering scalable CBT. In a 7-week open study of Inflow, a CBT-based mobile app, we assessed usability and feasibility to prepare for a randomized controlled trial (RCT). METHOD: 240 adults recruited online completed baseline and usability assessments at 2 (n = 114), 4 (n = 97) and after 7 weeks (n = 95) of Inflow use. 93 participants self-reported ADHD symptoms and impairment at baseline and 7 weeks. RESULTS: Participants rated Inflow's usability favorably, used the app a median of 3.86 times per week, and a majority of those using the app for 7 weeks self-reported decreases in ADHD symptoms and impairment. CONCLUSION: Inflow demonstrated usability and feasibility among users. An RCT will determine whether Inflow is associated with improvement among more rigorously assessed users and beyond non-specific factors.

3.
Clin Geriatr Med ; 37(2): 289-300, 2021 05.
Article in English | MEDLINE | ID: mdl-33858611

ABSTRACT

Patients with cancer may experience neuropathy at any stage of malignancy, ranging from symptoms that are the earliest signs of cancer to side effects of treatment. Peripheral nerves are affected most commonly in a symmetric, stocking-glove pattern. Sensory neuronopathies, plexopathies, and radiculopathies may also be seen. The most common type of neuropathy in patients with cancer is related to chemotherapy, and recently peripheral nerve complications have been described as an effect of immune checkpoint inhibitors too. Other causes include paraneoplastic syndromes, direct tumor infiltration, and radiation. Treatment focuses on addressing the underlying cancer and management of neuropathic pain.


Subject(s)
Brachial Plexus Neuropathies/etiology , Neoplasms/complications , Paraneoplastic Syndromes , Peripheral Nervous System Diseases/chemically induced , Radiation Injuries/complications , Antineoplastic Agents/adverse effects , Brachial Plexus Neuropathies/diagnosis , Drug-Related Side Effects and Adverse Reactions , Humans , Neoplasms/drug therapy , Paraneoplastic Syndromes/chemically induced , Paraneoplastic Syndromes/diagnosis , Peripheral Nervous System Diseases/complications , Radiation Injuries/diagnosis
4.
Sci Adv ; 5(3): eaav8423, 2019 03.
Article in English | MEDLINE | ID: mdl-30906870

ABSTRACT

The urea channel of Helicobacter pylori (HpUreI) is an ideal drug target for preventing gastric cancer but incomplete understanding of its gating mechanism has hampered development of inhibitors for the eradication of H. pylori. Here, we present the cryo-EM structures of HpUreI in closed and open conformations, both at a resolution of 2.7 Å. Our hexameric structures of this small membrane protein (~21 kDa/protomer) resolve its periplasmic loops and carboxyl terminus that close and open the channel, and define a gating mechanism that is pH dependent and requires cooperativity between protomers in the hexamer. Gating is further associated with well-resolved changes in the channel-lining residues that modify the shape and length of the urea pore. Site-specific mutations in the periplasmic domain and urea pore identified key residues important for channel function. Drugs blocking the urea pore based on our structures should lead to a new strategy for H. pylori eradication.


Subject(s)
Bacterial Proteins/ultrastructure , Helicobacter Infections/microbiology , Helicobacter pylori/ultrastructure , Membrane Transport Proteins/ultrastructure , Bacterial Proteins/chemistry , Cryoelectron Microscopy , Helicobacter Infections/genetics , Helicobacter pylori/chemistry , Helicobacter pylori/genetics , Humans , Hydrogen-Ion Concentration , Membrane Transport Proteins/chemistry , Protein Conformation , Stomach Neoplasms/genetics , Stomach Neoplasms/microbiology
5.
J Bacteriol ; 200(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29735759

ABSTRACT

Helicobacter pylori is an organism known to colonize the normal human stomach. Previous studies have shown that the bacterium does this by elevating its periplasmic pH via the hydrolysis of urea. However, the value of the periplasmic pH was calculated indirectly from the proton motive force equation. To measure the periplasmic pH directly in H. pylori, we fused enhanced green fluorescent protein (EGFP) to the predicted twin-arginine signal peptides of HydA and KapA from H. pylori and TorA from Escherichia coli The fusion proteins were expressed in the H. pylori genome under the control of the cagA promoter. Confocal microscopic and cell fractionation/immunoblotting analyses detected TorA-EGFP in the periplasm and KapA-EGFP in both the periplasm and cytoplasm, while the mature form of HydA-EGFP was seen at low levels in the periplasm, with major cytoplasmic retention of the precursor form. With H. pylori expressing TorA-EGFP, we established a system to directly measure periplasmic pH based on the pH-sensitive fluorimetry of EGFP. These measurements demonstrated that the addition of 5 mM urea has little effect on the periplasmic pH at a medium pH higher than pH 6.5 but rapidly increases the periplasmic pH to pH 6.1 at an acidic medium pH (pH 5.0), corresponding to the opening of the proton-gated channel, UreI, and confirming the basis of gastric colonization. Measurements of the periplasmic pH in an HP0244 (FlgS)-deficient mutant of H. pylori expressing TorA-EGFP revealed a significant loss of the urea-dependent increase in the periplasmic pH at an acidic medium pH, providing additional evidence that FlgS is responsible for recruitment of urease to the inner membrane in association with UreI.IMPORTANCEHelicobacter pylori has been identified as the major cause of chronic superficial gastritis and peptic ulcer disease. In addition, persistent infection with H. pylori, which, if untreated, lasts for the lifetime of an infected individual, predisposes one to gastric malignancies, such as adenocarcinoma and mucosa-associated lymphoid tissue (MALT) lymphoma. A unique feature of the neutralophilic bacterium H. pylori is its ability to survive in the extremely acidic environment of the stomach through its acid acclimation mechanism. The presented results on measurements of periplasmic pH in H. pylori based on fluorimetry of fully active green fluorescent protein fusion proteins exported with the twin-arginine translocase system provide a reliable and rapid tool for the investigation of acid acclimation in H. pylori.


Subject(s)
Fluorometry/methods , Green Fluorescent Proteins/metabolism , Helicobacter pylori/metabolism , Antigens, Bacterial , Bacterial Proteins , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/physiology , Helicobacter pylori/genetics , Hydrogen-Ion Concentration , Mutation , Promoter Regions, Genetic , Urea/metabolism , Urea/pharmacology
6.
Helicobacter ; 23(3): e12490, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29696729

ABSTRACT

BACKGROUND: The pathogen Helicobacter pylori encounters many stressors as it transits to and infects the gastric epithelium. Gastric acidity is the predominate stressor encountered by the bacterium during initial infection and establishment of persistent infection. H. pylori initiates a rapid response to acid to maintain intracellular pH and proton motive force appropriate for a neutralophile. However, acid sensing by H. pylori may also serve as a transcriptional trigger to increase the levels of other pathogenic factors needed to subvert host defenses such as acid acclimation, antioxidants, flagellar synthesis and assembly, and CagA secretion. MATERIALS AND METHODS: Helicobacter pylori were acid challenged at pH 3.0, 4.5, 6.0 vs nonacidic pH for 4 hours in the presence of urea, followed by RNA-seq analysis and qPCR. Cytoplasmic pH was monitored under the same conditions. RESULTS: About 250 genes were induced, and an equal number were repressed at acidic pHs. Genes encoding for antioxidant proteins, flagellar structural proteins, particularly class 2 genes, T4SS/Cag-PAI, Fo F1 -ATPase, and proteins involved in acid acclimation were highly expressed at acidic pH. Cytoplasmic pH decreased from 7.8 at pHout of 8.0 to 6.0 at pHout of 3.0. CONCLUSIONS: These results suggest that increasing extracellular or intracellular acidity or both are detected by the bacterium and serve as a signal to initiate increased production of protective and pathogenic factors needed to counter host defenses for persistent infection. These changes are dependent on degree of acidity and time of acid exposure, triggering a coordinated response to the environment required for colonization.


Subject(s)
Acids/metabolism , Gene Expression Regulation, Bacterial , Helicobacter pylori/genetics , Stomach/microbiology , Transcriptome/genetics , Adaptation, Physiological , Bacterial Proteins/genetics , Culture Media , Helicobacter pylori/physiology , Hydrogen-Ion Concentration , Multigene Family , Proteome/genetics , RNA, Bacterial/genetics , Urease/genetics , Virulence Factors/genetics
7.
Sci Rep ; 7(1): 6632, 2017 07 26.
Article in English | MEDLINE | ID: mdl-28747707

ABSTRACT

The gastric proton pump H+,K+-ATPase acidifies the gastric lumen, and thus its inhibitors, including the imidazo[1,2-a]pyridine class of K+-competitive acid blockers (P-CABs), have potential application as acid-suppressing drugs. We determined the electron crystallographic structure of H+,K+-ATPase at 6.5 Å resolution in the E2P state with bound BYK99, a potent P-CAB with a restricted ring structure. The BYK99 bound structure has an almost identical profile to that of a previously determined structure with bound SCH28080, the original P-CAB prototype, but is significantly different from the previously reported P-CAB-free form, illustrating a common conformational change is required for P-CAB binding. The shared conformational changes include a distinct movement of transmembrane helix 2 (M2), from its position in the previously reported P-CAB-free form, to a location proximal to the P-CAB binding site in the present BYK99-bound structure. Site-specific mutagenesis within M2 revealed that D137 and N138, which face the P-CAB binding site in our model, significantly affect the inhibition constant (K i) of P-CABs. We also found that A335 is likely to be near the bridging nitrogen at the restricted ring structure of the BYK99 inhibitor. These provide clues to elucidate the binding site parameters and mechanism of P-CAB inhibition of gastric acid secretion.


Subject(s)
Cryoelectron Microscopy , H(+)-K(+)-Exchanging ATPase/chemistry , Proton Pump Inhibitors/chemistry , Pyridines/chemistry , Animals , Protein Binding , Protein Conformation , Proton Pump Inhibitors/metabolism , Pyridines/metabolism , Swine
8.
R I Med J (2013) ; 99(12): 32-35, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27902997

ABSTRACT

Chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is an acquired immune-mediated disorder characterized by weakness and sensory deficits that can lead to significant neurological disability. The diagnosis is based on a combination of clinical examination findings, electrodiagnostic studies, and other supportive evidence. Recognizing CIDP and distinguishing it from other chronic polyneuropathies is important because many patients with CIDP are highly responsive to treatment with immunosuppressive or immunomodulatory therapies. This review summarizes the clinical features, diagnosis, and current treatment strategies for CIDP. [Full article available at http://rimed.org/rimedicaljournal-2016-12.asp].


Subject(s)
Immunomodulation , Immunosuppression Therapy , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/diagnosis , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/therapy , Humans
9.
J Biol Chem ; 291(44): 23159-23174, 2016 10 28.
Article in English | MEDLINE | ID: mdl-27624940

ABSTRACT

The Na,K-ATPase α2 subunit plays a key role in cardiac muscle contraction by regulating intracellular Ca2+, whereas α1 has a more conventional role of maintaining ion homeostasis. The ß subunit differentially regulates maturation, trafficking, and activity of α-ß heterodimers. It is not known whether the distinct role of α2 in the heart is related to selective assembly with a particular one of the three ß isoforms. We show here by immunofluorescence and co-immunoprecipitation that α2 is preferentially expressed with ß2 in T-tubules of cardiac myocytes, forming α2ß2 heterodimers. We have expressed human α1ß1, α2ß1, α2ß2, and α2ß3 in Pichia pastoris, purified the complexes, and compared their functional properties. α2ß2 and α2ß3 differ significantly from both α2ß1 and α1ß1 in having a higher K0.5K+ and lower K0.5Na+ for activating Na,K-ATPase. These features are the result of a large reduction in binding affinity for extracellular K+ and shift of the E1P-E2P conformational equilibrium toward E1P. A screen of perhydro-1,4-oxazepine derivatives of digoxin identified several derivatives (e.g. cyclobutyl) with strongly increased selectivity for inhibition of α2ß2 and α2ß3 over α1ß1 (range 22-33-fold). Molecular modeling suggests a possible basis for isoform selectivity. The preferential assembly, specific T-tubular localization, and low K+ affinity of α2ß2 could allow an acute response to raised ambient K+ concentrations in physiological conditions and explain the importance of α2ß2 for cardiac muscle contractility. The high sensitivity of α2ß2 to digoxin derivatives explains beneficial effects of cardiac glycosides for treatment of heart failure and potential of α2ß2-selective digoxin derivatives for reducing cardiotoxicity.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Enzyme Inhibitors/chemistry , Myocardium/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/chemistry , Animals , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/chemistry , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/chemistry , Dimerization , Enzyme Inhibitors/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Mice , Myocardium/chemistry , Potassium/chemistry , Potassium/metabolism , Sodium/chemistry , Sodium/metabolism , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics
10.
Open Cardiovasc Med J ; 10: 82-8, 2016.
Article in English | MEDLINE | ID: mdl-27347224

ABSTRACT

Muscular dystrophy and myasthenia gravis are two neuromuscular disorders that can involve significant cardiovascular complications. The frequency and severity of cardiac pathology varies widely among the muscular dystrophies. In some, it is nearly inevitable and requires regular evaluation. In others, assessment of cardiac function can be more symptom-driven. On-ly a minority of myasthenic patients manifest disease-related cardiovascular complications; however, their presentation can be rapidly progressive and life-threatening..

11.
J Cell Sci ; 129(12): 2394-406, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27142834

ABSTRACT

FXYD5 (also known as dysadherin), a regulatory subunit of the Na,K-ATPase, impairs intercellular adhesion by a poorly understood mechanism. Here, we determined whether FXYD5 disrupts the trans-dimerization of Na,K-ATPase molecules located in neighboring cells. Mutagenesis of the Na,K-ATPase ß1 subunit identified four conserved residues, including Y199, that are crucial for the intercellular Na,K-ATPase trans-dimerization and adhesion. Modulation of expression of FXYD5 or of the ß1 subunit with intact or mutated ß1-ß1 binding sites demonstrated that the anti-adhesive effect of FXYD5 depends on the presence of Y199 in the ß1 subunit. Immunodetection of the plasma membrane FXYD5 was prevented by the presence of O-glycans. Partial FXYD5 deglycosylation enabled antibody binding and showed that the protein level and the degree of O-glycosylation were greater in cancer than in normal cells. FXYD5-induced impairment of adhesion was abolished by both genetic and pharmacological inhibition of FXYD5 O-glycosylation. Therefore, the extracellular O-glycosylated domain of FXYD5 impairs adhesion by interfering with intercellular ß1-ß1 interactions, suggesting that the ratio between FXYD5 and α1-ß1 heterodimer determines whether the Na,K-ATPase acts as a positive or negative regulator of intercellular adhesion.


Subject(s)
Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Protein Multimerization , Protein Subunits/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , A549 Cells , Amino Acids/metabolism , Animals , Antibody Specificity , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Dogs , Epithelial Cells/metabolism , Gene Knockdown Techniques , Glycosylation , HEK293 Cells , Humans , Ion Channels , Madin Darby Canine Kidney Cells , Mice , Microfilament Proteins , Protein Binding , Protein Subunits/chemistry , Rats , Sodium-Potassium-Exchanging ATPase/chemistry
12.
Curr Gastroenterol Rep ; 18(7): 33, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27177639

ABSTRACT

Helicobacter pylori infects about 50 % of the world's population, causing at a minimum chronic gastritis. A subset of infected patients will ultimately develop gastric or duodenal ulcer disease, gastric adenocarcinoma, or MALT (mucosa-associated lymphoid tissue) lymphoma. Eradication of H. pylori requires complex regimens that include acid suppression and multiple antibiotics. The efficacy of treatment using what were once considered standard regimens have declined in recent years, mainly due to widespread development of antibiotic resistance. Addition of bismuth to standard triple therapy regimens, use of alternate antibiotics, or development of alternative regimens using known therapies in novel combinations have improved treatment efficacy in specific populations, but overall success of eradication remains less than ideal. Novel regimens under investigation either in vivo or in vitro, involving increased acid suppression ideally with fewer antibiotics or development of non-antibiotic treatment targets, show promise for future therapy.


Subject(s)
Gastritis/drug therapy , Gastritis/microbiology , Helicobacter Infections/drug therapy , Helicobacter pylori , Anti-Bacterial Agents/therapeutic use , Bismuth/therapeutic use , Chronic Disease , Drug Resistance, Bacterial , Drug Therapy, Combination , Humans , Proton Pump Inhibitors/therapeutic use
13.
Biochem J ; 473(12): 1703-18, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27048593

ABSTRACT

Septins are a family of cytoskeletal GTP-binding proteins that assemble into membrane-associated hetero-oligomers and organize scaffolds for recruitment of cytosolic proteins or stabilization of membrane proteins. Septins have been implicated in a diverse range of cancers, including gastric cancer, but the underlying mechanisms remain unclear. The hypothesis tested here is that septins contribute to cancer by stabilizing the receptor tyrosine kinase ErbB2, an important target for cancer treatment. Septins and ErbB2 were highly over-expressed in gastric cancer cells. Immunoprecipitation followed by MS analysis identified ErbB2 as a septin-interacting protein. Knockdown of septin-2 or cell exposure to forchlorfenuron (FCF), a well-established inhibitor of septin oligomerization, decreased surface and total levels of ErbB2. These treatments had no effect on epidermal growth factor receptor (EGFR), emphasizing the specificity and functionality of the septin-ErbB2 interaction. The level of ubiquitylated ErbB2 at the plasma membrane was elevated in cells treated with FCF, which was accompanied by a decrease in co-localization of ErbB2 with septins at the membrane. Cathepsin B inhibitor, but not bafilomycin or lactacystin, prevented FCF-induced decrease in total ErbB2 by increasing accumulation of ubiquitylated ErbB2 in lysosomes. Therefore, septins protect ErbB2 from ubiquitylation, endocytosis and lysosomal degradation. The FCF-induced degradation pathway is distinct from and additive with the degradation induced by inhibiting ErbB2 chaperone Hsp90. These results identify septins as novel regulators of ErbB2 expression that contribute to the remarkable stabilization of the receptor at the plasma membrane of cancer cells and may provide a basis for the development of new ErbB2-targeting anti-cancer therapies.


Subject(s)
Receptor, ErbB-2/metabolism , Septins/metabolism , Stomach Neoplasms/metabolism , Blotting, Western , Cell Line, Tumor , Chromatography, Liquid , Cytoskeleton/metabolism , Humans , Immunoprecipitation , Phenylurea Compounds/pharmacology , Protein Binding/drug effects , Pyridines/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Receptor, ErbB-2/genetics , Septins/antagonists & inhibitors , Septins/genetics , Signal Transduction/physiology , Tandem Mass Spectrometry , Ubiquitination/drug effects
14.
Dig Dis Sci ; 61(7): 1783-4, 2016 07.
Article in English | MEDLINE | ID: mdl-27074922
15.
Helicobacter ; 21(1): 69-81, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25997502

ABSTRACT

BACKGROUND: The pH-sensitive Helicobacter pylori ArsRS two-component system (TCS) aids survival of this neutralophile in the gastric environment by directly sensing and responding to environmental acidity. ArsS is required for acid-induced trafficking of urease and its accessory proteins to the inner membrane, allowing rapid, urea-dependent cytoplasmic and periplasmic buffering. Expression of ArsR, but not its phosphorylation, is essential for bacterial viability. The aim of this study was to characterize the roles of ArsS and ArsR in the response of H. pylori to acid. MATERIALS AND METHODS: Wild-type H. pylori and an arsR(D52N) phosphorylation-deficient strain were incubated at acidic or neutral pH. Gene and protein expression, survival, membrane trafficking of urease proteins, urease activity, and internal pH were studied. RESULTS: Phosphorylation of ArsR is not required for acid survival. ArsS-driven trafficking of urease proteins to the membrane in acid, required for recovery of internal pH, is independent of ArsR phosphorylation. ArsR phosphorylation increases expression of the urease gene cluster, and the loss of negative feedback in a phosphorylation-deficient mutant leads to an increase in total urease activity. CONCLUSIONS: ArsRS has a dual function in acid acclimation: regulation of urease trafficking to UreI at the cytoplasmic membrane, driven by ArsS, and regulation of urease gene cluster expression, driven by phosphorylation of ArsR. ArsS and ArsR work through phosphorylation-dependent and phosphorylation-independent regulatory mechanisms to impact acid acclimation and allow gastric colonization. Furthering understanding of the intricacies of acid acclimation will impact the future development of targeted, nonantibiotic treatment regimens.


Subject(s)
Acids/metabolism , Bacterial Proteins/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/metabolism , Bacterial Proteins/genetics , Cell Membrane/genetics , Cell Membrane/metabolism , Gene Expression Regulation, Bacterial , Helicobacter pylori/genetics , Humans , Phosphorylation , Protein Transport , Urease/genetics , Urease/metabolism
16.
F1000Res ; 52016.
Article in English | MEDLINE | ID: mdl-30023042

ABSTRACT

Infection of the stomach by the gastric pathogen Helicobacter pylori results in chronic active gastritis and leads to the development of gastric and duodenal ulcer disease and gastric adenocarcinoma. Eradication of H. pylori infection improves or resolves the associated pathology. Current treatments of H. pylori infection rely on acid suppression in combination with at least two antibiotics. The role of acid suppression in eradication therapy has been variously attributed to antibacterial activity of proton pump inhibitors directly or through inhibition of urease activity or increased stability and activity of antibiotics. Here we discuss the effect of acid suppression on enhanced replicative capacity of H. pylori to permit the bactericidal activity of growth-dependent antibiotics. The future of eradication therapy will rely on improvement of acid inhibition along with current antibiotics or the development of novel compounds targeting the organism's ability to survive in acid.

17.
J Biol Chem ; 290(9): 5280-97, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25575596

ABSTRACT

Septins are a family of 14 cytoskeletal proteins that dynamically form hetero-oligomers and organize membrane microdomains for protein complexes. The previously reported interactions with SNARE proteins suggested the involvement of septins in exocytosis. However, the contradictory results of up- or down-regulation of septin-5 in various cells and mouse models or septin-4 in mice suggested either an inhibitory or a stimulatory role for these septins in exocytosis. The involvement of the ubiquitously expressed septin-2 or general septin polymerization in exocytosis has not been explored to date. Here, by nano-LC with tandem MS and immunoblot analyses of the septin-2 interactome in mouse brain, we identified not only SNARE proteins but also Munc-18-1 (stabilizes assembled SNARE complexes), N-ethylmaleimide-sensitive factor (NSF) (disassembles SNARE complexes after each membrane fusion event), and the chaperones Hsc70 and synucleins (maintain functional conformation of SNARE proteins after complex disassembly). Importantly, α-soluble NSF attachment protein (SNAP), the adaptor protein that mediates NSF binding to the SNARE complex, did not interact with septin-2, indicating that septins undergo reorganization during each exocytosis cycle. Partial depletion of septin-2 by siRNA or impairment of septin dynamics by forchlorfenuron inhibited constitutive and stimulated exocytosis of secreted and transmembrane proteins in various cell types. Forchlorfenuron impaired the interaction between SNAP-25 and its chaperone Hsc70, decreasing SNAP-25 levels in cultured neuroendocrine cells, and inhibited both spontaneous and stimulated acetylcholine secretion in mouse motor neurons. The results demonstrate a stimulatory role of septin-2 and the dynamic reorganization of septin oligomers in exocytosis.


Subject(s)
Brain/metabolism , Exocytosis , Proteome/metabolism , Septins/metabolism , Animals , Blotting, Western , Brain/drug effects , Cell Line , Cell Line, Tumor , Dogs , Female , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Male , Mice, Inbred BALB C , Microscopy, Confocal , PC12 Cells , Phenylurea Compounds/pharmacology , Protein Binding/drug effects , Protein Multimerization , Proteomics , Pyridines/pharmacology , RNA Interference , Rats , Septins/chemistry , Septins/genetics , Synaptosomal-Associated Protein 25/metabolism
18.
J Cell Sci ; 127(Pt 15): 3294-308, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24928902

ABSTRACT

Proteolytic cleavage of synaptosomal-associated protein 25 by the light chain of botulinum neurotoxin type A (LCA) results in a blockade of neurotransmitter release that persists for several months in motor neurons. The L428A/L429A mutation in LCA is known to significantly shorten both the proteolytic and neuroparalytic effects of the neurotoxin in mice. To elucidate the cellular mechanism for LCA longevity, we studied the effects of L428A/L429A mutation on the interactome, localization and stability of LCA expressed in cultured neuronal cells. Mass spectrometry analysis of the LCA interactome showed that the mutation prevented the interaction of LCA with septins. The wild-type LCA was concentrated in plasma-membrane-associated clusters, colocalizing with septins-2 and septin-7, which accumulated in these clusters only in the presence of LCA. The L428A/L429A mutation decreased co-clustering of LCA and septins and accelerated proteasomal and non-proteasomal degradation of LCA. Similarly, the impairment of septin oligomerization by forchlorfenuron or silencing of septin-2 prevented LCA interaction and clustering with septins and increased LCA degradation. Therefore, the dileucine-mediated LCA-septin co-clustering is crucial for the long-lasting stabilization of LCA-related proteolytic and presumably neuroparalytic activity.


Subject(s)
Botulinum Toxins, Type A/metabolism , Cell Membrane/metabolism , Neurons/physiology , Neurotoxicity Syndromes/metabolism , Septins/metabolism , Animals , Botulinum Toxins, Type A/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Humans , Mice , Mutation/genetics , Neurons/microbiology , Neurotoxicity Syndromes/microbiology , Phenylurea Compounds/pharmacology , Protein Binding/drug effects , Protein Binding/genetics , Protein Multimerization/drug effects , Protein Stability/drug effects , Protein Transport/drug effects , Protein Transport/genetics , Pyridines/pharmacology , RNA, Small Interfering/genetics , Septins/genetics
20.
Am J Physiol Gastrointest Liver Physiol ; 305(10): G731-9, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23989011

ABSTRACT

Gastric infection by Helicobacter pylori is the most common cause of ulcer disease and gastric cancer. The mechanism of progression from gastritis and inflammation to ulcers and cancer in a fraction of those infected is not definitively known. Significant acidity is unique to the gastric environment and is required for ulcer development. The interplay between gastric acidity and H. pylori pathogenesis is important in progression to advanced disease. The aim of this study was to characterize the impact of acid on gastric epithelial integrity and cytokine release and how H. pylori infection alters these responses. Human gastric epithelial (HGE-20) cells were grown on porous inserts, and survival, barrier function, and cytokine release were studied at various apical pH levels in the presence and absence of H. pylori. With apical acidity, gastric epithelial cells demonstrate increased barrier function, as evidenced by increased transepithelial electrical resistance (TEER) and decreased paracellular permeability. This effect is reduced in the presence of wild-type, but not urease knockout, H. pylori. The epithelial inflammatory response is also modulated by acidity and H. pylori infection. Without H. pylori, epithelial IL-8 release decreases in acid, while IL-6 release increases. In the presence of H. pylori, acidic pH diminishes the magnitude of the previously reported increase in IL-8 and IL-6 release. H. pylori interferes with the gastric epithelial response to acid, contributing to altered barrier function and inflammatory response. H. pylori diminishes acid-induced tightening of cell junctions in a urease-dependent manner, suggesting that local pH elevation promotes barrier compromise and progression to mucosal damage.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/microbiology , Helicobacter pylori/metabolism , Cell Line, Tumor , Culture Media/chemistry , Electric Impedance , Electrophysiological Phenomena , Gene Expression Regulation, Bacterial , Helicobacter pylori/genetics , Humans , Hydrogen-Ion Concentration
SELECTION OF CITATIONS
SEARCH DETAIL