Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Elife ; 92020 10 19.
Article in English | MEDLINE | ID: mdl-33074099

ABSTRACT

Ubiquitination regulates many different cellular processes, including protein quality control, membrane trafficking, and stress responses. The diversity of ubiquitin functions in the cell is partly due to its ability to form chains with distinct linkages that can alter the fate of substrate proteins in unique ways. The complexity of the ubiquitin code is further enhanced by post-translational modifications on ubiquitin itself, the biological functions of which are not well understood. Here, we present genetic and biochemical evidence that serine 57 (Ser57) phosphorylation of ubiquitin functions in stress responses in Saccharomyces cerevisiae, including the oxidative stress response. We also identify and characterize the first known Ser57 ubiquitin kinases in yeast and human cells, and we report that two Ser57 ubiquitin kinases regulate the oxidative stress response in yeast. These studies implicate ubiquitin phosphorylation at the Ser57 position as an important modifier of ubiquitin function, particularly in response to proteotoxic stress.


Subject(s)
Oxidative Stress , Saccharomyces cerevisiae/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Phosphorylation , Saccharomyces cerevisiae/enzymology , Serine , Ubiquitin-Protein Ligases/physiology
2.
PLoS Genet ; 16(3): e1008677, 2020 03.
Article in English | MEDLINE | ID: mdl-32191698

ABSTRACT

Endocytosis is regulated in response to changing environmental conditions to adjust plasma membrane (PM) protein composition for optimal cell growth. Protein networks involved in cargo capture and sorting, membrane sculpting and deformation, and vesicle scission have been well-characterized, but less is known about the networks that sense extracellular cues and relay signals to trigger endocytosis of specific cargo. Hal4 and Hal5 are yeast Snf1-related kinases that were previously reported to regulate nutrient transporter stability by an unknown mechanism. Here we demonstrate that loss of Hal4 and Hal5 activates endocytosis of many different kinds of PM proteins, including Art1-mediated and Art1-independent endocytic events. Acute inhibition of Hal5 in the absence of Hal4 triggers rapid endocytosis, suggesting that Hal kinases function in a nutrient-sensing relay upstream of the endocytic response. Interestingly, Hal5 localizes to the PM, but shifts away from the cell surface in response to stimulation with specific nutrients. We propose that Hal5 functions as a nutrient-responsive regulator of PM protein stability, antagonizing endocytosis and promoting stability of endocytic cargos at the PM in nutrient-limiting conditions.


Subject(s)
Endocytosis/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Biological Transport , Cell Membrane/metabolism , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Protein Interaction Maps , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein Transport , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics
3.
J Cell Biol ; 218(3): 977-992, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30610170

ABSTRACT

Regulation of plasma membrane (PM) protein abundance by selective endocytosis is critical for cellular adaptation to stress or changing nutrient availability. One example involves rapid endocytic turnover of Mup1, a yeast methionine transporter, in response to increased methionine availability. Here, we report that methionine triggers rapid translocation of the ubiquitin ligase adaptor Art1 to the PM and dephosphorylation of Art1 at specific threonine residues. This methionine-induced dephosphorylation of Art1 is mediated by Ppz phosphatases, and analysis of phosphomimetic and phosphorylation-defective variants of Art1 indicates that these events toggle Art1 recognition of Mup1 at the PM. Importantly, we find that Ppz phosphatases are dispensable for Art1 PM translocation, but are required for Art1 interaction with Mup1. Based on our findings, we propose that methionine influx triggers Art1 translocation to the PM, followed by Ppz-mediated dephosphorylation which promotes cargo recognition at the PM.


Subject(s)
Cell Membrane/metabolism , DNA-Binding Proteins/metabolism , Endocytosis/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Transcription Factors/metabolism , Cell Membrane/genetics , DNA-Binding Proteins/genetics , Phosphorylation/physiology , Proteins/genetics , Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
4.
Neuropharmacology ; 141: 192-200, 2018 10.
Article in English | MEDLINE | ID: mdl-30170085

ABSTRACT

Numerous studies have attributed the psychopathology of anxiety and stress disorders to maladaptive behavioral responses such as an inability to extinguish fear. Therefore, understanding neural substrates of fear extinction is imperative for developing more effective therapies for anxiety and stress disorders. Although several studies indicated a role for cholinergic transmission and nicotinic acetylcholine receptors (nAChRs) in anxiety and stress disorder symptomatology, very little is known about the specific contribution of nAChRs in the fear extinction process. In the present study, we first examined the involvement of several brain regions essential for fear extinction (i.e., dorsal and ventral hippocampus, dHPC and vHPC; infralimbic, IL, and prelimbic, PL of the medial prefrontal cortex, mPFC; basolateral nucleus of the amygdala, BLA) in the impairing effects of a nAChR agonist, nicotine, on contextual fear extinction in mice. Our results showed that systemic administration of nicotine during contextual fear extinction increased c-fos expression in the vHPC and BLA while not affecting dHPC, IL or PL. In line with these results, local nicotine infusions into the vHPC, but not dHPC, resulted in impaired contextual fear extinction. Interestingly, we found that local nicotine infusions into the PL also resulted in impairment of contextual fear extinction. Second, we measured the protein levels of the GABA synthesizing enzymes GAD65 and GAD67 in the dHPC and vHPC during contextual fear extinction. Our results showed that in the group that received acute nicotine, both GAD65 and GAD67 protein levels were downregulated in the vHPC, but not in dHPC. This effect was negatively correlated with the level of freezing response during fear extinction suggesting that the downregulated GAD65/67 levels were associated with disrupted fear extinction. Finally, using c-fos/GAD65/67 double immunofluorescence, we showed that nicotine mainly increased c-fos expression in non-GABAergic ventral hippocampal cells, indicating that acute nicotine increases vHPC excitability. Overall, our results suggest that acute nicotine's impairing effects on fear extinction are associated with ventral hippocampal disinhibition. Therefore, these results further our understanding of the interaction between nicotine addiction and anxiety and stress disorders by describing novel neural mechanisms mediating fear extinction.


Subject(s)
Extinction, Psychological/drug effects , Fear/drug effects , Hippocampus/metabolism , Nicotine/pharmacology , Amygdala/drug effects , Amygdala/physiology , Animals , Down-Regulation/drug effects , Extinction, Psychological/physiology , Fear/physiology , Glutamate Decarboxylase/metabolism , Hippocampus/drug effects , Hippocampus/physiology , Immobility Response, Tonic/drug effects , Male , Mice , Microinjections , Nicotine/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Proto-Oncogene Proteins c-fos/metabolism
5.
Neurosci Lett ; 673: 142-149, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29518543

ABSTRACT

Stress and anxiety disorders such as posttraumatic stress disorder (PTSD) are characterized by disrupted safety learning. Tobacco smoking has been strongly implicated in stress and anxiety disorder symptomatology, both as a contributing factor and as a vulnerability factor. Rodent studies from our lab have recently shown that acute and chronic nicotine exposure disrupts safety learning. However, it is unknown if these effects of nicotine translate to humans. The present studies addressed this gap by administering a translational differential cued fear conditioning paradigm to both mice and humans. In mice, we found that chronic nicotine exposure reduced discrimination between a conditioned stimulus (CS) that signals for danger (CS+) and another CS that signals for safety (CS-) during both acquisition and testing. We then employed a similar differential cued fear conditioning paradigm in human smokers and non-smokers undergoing functional magnetic resonance imaging (fMRI). Smokers showed reduced CS+/CS- discrimination during fear conditioning compared to non-smokers. Furthermore, using fMRI, we found that subgenual and dorsal anterior cingulate cortex activations were lower in smokers than in non-smokers during differential cued fear conditioning. These results suggest a potential biological mechanism underlying a dysregulated ability to discriminate between danger and safety cues. Our results indicate a clear parallel between the effects of nicotine exposure on safety learning in mice and humans and therefore suggest that smoking might represent a risk factor for inability to process information related to danger and safety related cues.


Subject(s)
Cues , Discrimination, Psychological/drug effects , Fear , Gyrus Cinguli/drug effects , Gyrus Cinguli/physiology , Nicotine/administration & dosage , Animals , Brain Mapping , Cigarette Smoking , Conditioning, Classical/drug effects , Humans , Magnetic Resonance Imaging , Male , Mice, Inbred C57BL , Middle Aged
6.
Psychopharmacology (Berl) ; 235(4): 1211-1219, 2018 04.
Article in English | MEDLINE | ID: mdl-29383396

ABSTRACT

RATIONALE: Numerous studies have attributed the psychopathology of post-traumatic stress disorder (PTSD) to maladaptive behavioral responses such as an inability to extinguish fear. While exposure therapies are mostly effective in treating these disorders by enhancing extinction learning, relapse of PTSD symptoms is common. Although several studies indicated a role for cholinergic transmission and nicotinic acetylcholine receptors (nAChRs) in anxiety and stress disorder symptomatology, very little is known about the specific contribution of nAChRs to fear extinction OBJECTIVES: In the present study, we examined the effects of inhibition and desensitization of α4ß2 nAChRs via a full antagonist (Dihydro-beta-erythroidine (DhßE)) and two α4ß2 nAChR partial-agonists (varenicline and sazetidine-A) on contextual fear extinction, locomotor activity, and spontaneous recovery of contextual fear in mice. METHODS: We trained and tested the subjects in a contextual fear extinction as well as an open field paradigm and spontaneous recovery following injections of DhßE, varenicline, and sazetidine-A. RESULTS: Our results demonstrated that lower doses of DhßE (1 mg/kg) and sazetidine-A (0.01 mg/kg) enhanced contextual fear extinction whereas higher doses of varenicline (0.1 mg/kg) and sazetidine-A (0.1 mg/kg) resulted in impaired contextual fear extinction. However, the higher dose of sazetidine-A (0.1 mg/kg) decreased locomotor activity, which may contribute to increased freezing response observed during fear extinction. Finally, we found that the low dose of DhßE, but not sazetidine-A, also decreased spontaneous recovery of contextual fear following fear extinction. CONCLUSIONS: Overall, these results suggest that inhibition and desensitization of α4ß2 nAChRs enhance extinction of contextual fear memories. This suggests that modulation of α4ß2 nAChRs may be employed as an alternative pharmacological strategy to aid exposure therapies associated with PTSD by augmenting contextual fear extinction processes.


Subject(s)
Azetidines/pharmacology , Dihydro-beta-Erythroidine/pharmacology , Extinction, Psychological/drug effects , Fear/drug effects , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Pyridines/pharmacology , Receptors, Nicotinic/physiology , Varenicline/pharmacology , Animals , Anxiety/drug therapy , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Receptors, Nicotinic/drug effects , Stress Disorders, Post-Traumatic/drug therapy
7.
Behav Brain Res ; 341: 176-180, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29307664

ABSTRACT

Post-traumatic stress disorder (PTSD) is a devastating disorder with symptoms such as flashbacks, hyperarousal, and avoidance of reminders of the traumatic event. Exposure therapy, which attempts to extinguish fear responses, is a commonly used treatment for PTSD but relapse following successful exposure therapy is a frequent problem. In rodents, spontaneous recovery (SR), where extinguished fear responses resurface following extinction treatment, is used as a model of fear relapse. Previous studies from our lab showed that chronic nicotine impaired fear extinction and acute nicotine enhanced SR of contextual fear in adult male mice. In addition, we showed that acute nicotine's effects were specific to SR as acute nicotine did not affect recall of contextual fear conditioning in the absence of extinction. However, effects of chronic nicotine administration on SR are not known. Therefore, in the present study, we investigated if chronic nicotine administration altered SR or recall of contextual fear in adult male and female C57BL/6J mice. Our results showed that chronic nicotine significantly enhanced SR in female mice and significantly decreased SR in males. Chronic nicotine had no effect on recall of contextual fear in males or females. Female sham mice also had significantly less baseline SR than male sham mice. Overall, these results demonstrate sex differences in SR of fear memories and that chronic nicotine modulates these effects on SR but nicotine does not alter recall of contextual fear.


Subject(s)
Extinction, Psychological/drug effects , Fear/drug effects , Mental Recall/drug effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Sex Characteristics , Analysis of Variance , Animals , Conditioning, Psychological/drug effects , Electroshock , Female , Infusion Pumps, Implantable , Mice , Mice, Inbred C57BL
8.
Brain Res Bull ; 138: 50-55, 2018 04.
Article in English | MEDLINE | ID: mdl-28624583

ABSTRACT

Adolescence is a period of high risk for the initiation of nicotine product usage and exposure to traumatic events. In parallel, nicotine exposure has been found to age-dependently modulate acquisition of contextual fear memories; however, it is unknown if adolescent nicotine exposure alters extinction of fear related memories. Age-related differences in sensitivity to the effects of nicotine on fear extinction could increase or decrease susceptibility to anxiety disorders. In this study, we examined the effects of acute nicotine administration on extinction and spontaneous recovery of contextual fear memories in pre-adolescent (PND 23), late adolescent (PND 38), and adult (PND 53) C57B6/J mice. Mice were first trained in a background contextual fear conditioning paradigm and given an intraperitoneal injection of one of four doses of nicotine (0.045, 0.09, 0.18, or 0.36mg/kg, freebase) prior to subsequent extinction or spontaneous recovery sessions. Results indicated that all acute nicotine doses impaired extinction of contextual fear in adult mice. Late adolescent mice exhibited impaired extinction of contextual fear only following higher doses of acute nicotine, and extinction of contextual fear was unaffected by acute nicotine exposure in pre-adolescent mice. Finally, acute nicotine exposure enhanced spontaneous recovery of fear memory, but only in adult mice. Overall, our results suggest that younger mice were less sensitive to nicotine's impairing effects on extinction of contextual fear and to nicotine's enhancing effects on spontaneous recovery of contextual fear memory.


Subject(s)
Age Factors , Conditioning, Classical/drug effects , Extinction, Psychological/drug effects , Learning Disabilities/chemically induced , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Animals , Animals, Newborn , Disease Models, Animal , Dose-Response Relationship, Drug , Electroshock/adverse effects , Fear/drug effects , Fear/psychology , Male , Mice , Mice, Inbred C57BL
9.
Elife ; 62017 11 13.
Article in English | MEDLINE | ID: mdl-29130884

ABSTRACT

Despite its central role in protein degradation little is known about the molecular mechanisms that sense, maintain, and regulate steady state concentration of ubiquitin in the cell. Here, we describe a novel mechanism for regulation of ubiquitin homeostasis that is mediated by phosphorylation of ubiquitin at the Ser57 position. We find that loss of Ppz phosphatase activity leads to defects in ubiquitin homeostasis that are at least partially attributable to elevated levels of Ser57 phosphorylated ubiquitin. Phosphomimetic mutation at the Ser57 position of ubiquitin conferred increased rates of endocytic trafficking and ubiquitin turnover. These phenotypes are associated with bypass of recognition by endosome-localized deubiquitylases - including Doa4 which is critical for regulation of ubiquitin recycling. Thus, ubiquitin homeostasis is significantly impacted by the rate of ubiquitin flux through the endocytic pathway and by signaling pathways that converge on ubiquitin itself to determine whether it is recycled or degraded in the vacuole.


Subject(s)
Endocytosis , Protein Processing, Post-Translational , Ubiquitin/metabolism , Yeasts/metabolism , Homeostasis , Phosphoprotein Phosphatases/deficiency , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Saccharomyces cerevisiae Proteins/metabolism
10.
Neurobiol Learn Mem ; 145: 143-150, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29017931

ABSTRACT

Previous research has shown that acute nicotine, an agonist of nAChRs, impaired fear extinction. However, the effects of acute nicotine on consolidation of contextual fear extinction memories and associated cell signaling cascades are unknown. Therefore, we examined the effects of acute nicotine injections before (pre-extinction) and after (post-extinction) contextual fear extinction on behavior and the phosphorylation of dorsal and ventral hippocampal ERK1/2 and JNK1 and protein levels on the 1st and 3rd day of extinction. Our results showed that acute nicotine administered prior to extinction sessions downregulated the phosphorylated forms of ERK1/2 in the ventral hippocampus, but not dorsal hippocampus, and JNK1 in both dorsal and ventral hippocampus on the 3rd extinction day. These effects were absent on the 1st day of extinction. We also showed that acute nicotine administered immediately and 30 min, but not 6 h, following extinction impaired contextual fear extinction suggesting that acute nicotine disrupts consolidation of contextual fear extinction memories. Finally, acute nicotine injections immediately after extinction sessions upregulated the phosphorylated forms of ERK1/2 in the ventral hippocampus, but did not affect JNK1. These results show that acute nicotine impairs contextual fear extinction potentially by altering molecular processes responsible for the consolidation of extinction memories.


Subject(s)
Extinction, Psychological/drug effects , Hippocampus/drug effects , Memory Consolidation/drug effects , Memory, Long-Term/drug effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Animals , Conditioning, Classical/drug effects , Fear , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Phosphorylation
11.
Learn Mem ; 23(8): 405-14, 2016 08.
Article in English | MEDLINE | ID: mdl-27421892

ABSTRACT

Exposure therapy, which focuses on extinguishing fear-triggering cues and contexts, is widely used to treat post-traumatic stress disorder (PTSD). Yet, PTSD patients who received successful exposure therapy are vulnerable to relapse of fear response after a period of time, a phenomenon known as spontaneous recovery (SR). Increasing evidence suggests ventral hippocampus, basolateral amygdala, and infralimbic cortex may be involved in SR. PTSD patients also show high rates of comorbidity with nicotine dependence. While the comorbidity between smoking and PTSD might suggest nicotine may alter SR, the effects of nicotine on SR of contextual fear are unknown. In the present study, we tested the effects of acute nicotine administration on SR of extinguished contextual fear memories and c-fos immediate early gene immunohistochemistry in mice. Our results demonstrated that acute nicotine enhanced SR of extinguished fear whereas acute nicotine did not affect retrieval of unextinguished contextual memories. This suggests that the effect of acute nicotine on SR is specific for memories that have undergone extinction treatment. C-fos immunoreactive (IR) cells in the ventral hippocampus and basolateral amygdala were increased in the nicotine-treated mice following testing for SR, whereas the number of IR cells in the infralimbic cortex was decreased in the same group. Overall, this study suggests that nicotine may adversely affect context-specific relapse of fear memories and this effect is potentially mediated by the suppression of cortical regions and increased activity in the ventral hippocampus and amygdala.


Subject(s)
Brain/drug effects , Brain/metabolism , Extinction, Psychological/drug effects , Fear/drug effects , Nicotine/administration & dosage , Amygdala/drug effects , Amygdala/metabolism , Animals , Conditioning, Classical , Cues , Gene Expression , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis
12.
Brain Res ; 1642: 445-451, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27084582

ABSTRACT

Adolescence is a critical developmental period associated with both increased vulnerability to substance abuse and maturation of certain brain regions important for learning and memory such as the hippocampus. In this study, we employed a hippocampus-dependent learning context pre-exposure facilitation effect (CPFE) paradigm in order to test the effects of acute nicotine on contextual processing during adolescence (post-natal day (PND) 38) and adulthood (PND 53). In Experiment 1, adolescent or adult C57BL6/J mice received either saline or one of three nicotine doses (0.09, 0.18, and 0.36mg/kg) prior to contextual pre-exposure and testing. Our results demonstrated that both adolescent and adult mice showed CPFE in the saline groups. However, adolescent mice only showed acute nicotine enhancement of CPFE with the highest nicotine dose whereas adult mice showed the enhancing effects of acute nicotine with all three doses. In Experiment 2, to determine if the lack of nicotine's effects on CPFE shown by adolescent mice is specific to the age when they are tested, mice were either given contextual pre-exposure during adolescence or adulthood and received immediate shock and testing during adulthood after a 15day delay. We found that both adolescent and adult mice showed CPFE in the saline groups when tested during adulthood. However, like Experiment 1, mice that received contextual pre-exposure during adolescence did not show acute nicotine enhancement except at the highest dose (0.36mg/kg) whereas both low (0.09mg/kg) and high (0.36mg/kg) doses enhanced CPFE in adult mice. Finally, we showed that the enhanced freezing response found with 0.36mg/kg nicotine in the 15-day experiment may be a result of decreased locomotor activity as mice that received this dose of nicotine traveled shorter distances in an open field paradigm. Overall, our results indicate that while adolescent mice showed normal contextual processing when tested both during adolescence and adulthood, they are less sensitive to the enhancing effects of nicotine on contextual processing.


Subject(s)
Aging/drug effects , Conditioning, Psychological/drug effects , Fear/drug effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Animals , Dose-Response Relationship, Drug , Electroshock , Exploratory Behavior/drug effects , Freezing Reaction, Cataleptic/drug effects , Mice, Inbred C57BL , Models, Animal , Motor Activity/drug effects , Psychological Tests
13.
PLoS One ; 9(8): e105830, 2014.
Article in English | MEDLINE | ID: mdl-25144372

ABSTRACT

Acid sphingomyelinase (aSMase) is a human enzyme that catalyzes the hydrolysis of sphingomyelin to generate the bioactive lipid ceramide and phosphocholine. ASMase deficiency is the underlying cause of the genetic diseases Niemann-Pick Type A and B and has been implicated in the onset and progression of a number of other human diseases including cancer, depression, liver, and cardiovascular disease. ASMase is the founding member of the aSMase protein superfamily, which is a subset of the metallophosphatase (MPP) superfamily. To date, MPPs that share sequence homology with aSMase, termed aSMase-like proteins, have been annotated and presumed to function as aSMases. However, none of these aSMase-like proteins have been biochemically characterized to verify this. Here we identify RsASML, previously annotated as RSp1609: acid sphingomyelinase-like phosphodiesterase, as the first bacterial aSMase-like protein from the deadly plant pathogen Ralstonia solanacearum based on sequence homology with the catalytic and C-terminal domains of human aSMase. A biochemical characterization of RsASML does not support a role in sphingomyelin hydrolysis but rather finds RsASML capable of acting as an ATP diphosphohydrolase, catalyzing the hydrolysis of ATP and ADP to AMP. In addition, RsASML displays a neutral, not acidic, pH optimum and prefers Ni2+ or Mn2+, not Zn2+, for catalysis. This alters the expectation that all aSMase-like proteins function as acid SMases and expands the substrate possibilities of this protein superfamily to include nucleotides. Overall, we conclude that sequence homology with human aSMase is not sufficient to predict substrate specificity, pH optimum for catalysis, or metal dependence. This may have implications to the biochemically uncharacterized human aSMase paralogs, aSMase-like 3a (aSML3a) and aSML3b, which have been implicated in cancer and kidney disease, respectively, and assumed to function as aSMases.


Subject(s)
Adenosine Monophosphate/chemistry , Adenosine Triphosphate/chemistry , Bacterial Proteins/chemistry , Ralstonia solanacearum/enzymology , Sphingomyelin Phosphodiesterase/chemistry , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Bacterial Proteins/metabolism , Humans , Hydrolysis , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/chemistry , Sphingomyelins/metabolism , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...