Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
Transl Psychiatry ; 10(1): 343, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037178

ABSTRACT

The transcription factor TCF4 was confirmed in several large genome-wide association studies as one of the most significant schizophrenia (SZ) susceptibility genes. Transgenic mice moderately overexpressing Tcf4 in forebrain (Tcf4tg) display deficits in fear memory and sensorimotor gating. As second hit, we exposed Tcf4tg animals to isolation rearing (IR), chronic social defeat (SD), enriched environment (EE), or handling control (HC) conditions and examined mice with heterozygous deletion of the exon 4 (Tcf4Ex4δ+/-) to unravel gene-dosage effects. We applied multivariate statistics for behavioral profiling and demonstrate that IR and SD cause strong cognitive deficits of Tcf4tg mice, whereas EE masked the genetic vulnerability. We observed enhanced long-term depression in Tcf4tg mice and enhanced long-term potentiation in Tcf4Ex4δ+/- mice indicating specific gene-dosage effects. Tcf4tg mice showed higher density of immature spines during development as assessed by STED nanoscopy and proteomic analyses of synaptosomes revealed concurrently increased levels of proteins involved in synaptic function and metabolic pathways. We conclude that environmental stress and Tcf4 misexpression precipitate cognitive deficits in 2-hit mouse models of relevance for schizophrenia.


Subject(s)
Schizophrenia , Animals , Cognition , Disease Models, Animal , Genome-Wide Association Study , Mice , Mice, Transgenic , Neuronal Plasticity/genetics , Proteomics , Schizophrenia/genetics
2.
Transl Psychiatry ; 7(4): e1078, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28375208

ABSTRACT

The aim of this study was to identify molecular pathways related to antidepressant response. We administered paroxetine to the DBA/2J mice for 28 days. Following the treatment, the mice were grouped into responders or non-responders depending on the time they spent immobile in the forced swim test. Hippocampal metabolomics and proteomics analyses revealed that chronic paroxetine treatment affects glutamate-related metabolite and protein levels differentially in the two groups. We found significant differences in the expression of N-methyl-d-aspartate receptor and neuronal nitric oxide synthase proteins between the two groups, without any significant alterations in the respective transcript levels. In addition, we found that chronic paroxetine treatment altered the levels of proteins associated with the ubiquitin-proteasome system (UPS). The soluble guanylate cyclase-ß1, proteasome subunit α type-2 and ubiquitination levels were also affected in peripheral blood mononuclear cells from antidepressant responder and non-responder patients suffering from major depressive disorder. We submit that the glutamatergic system and UPS have a crucial role in the antidepressant treatment response in both mice and humans.


Subject(s)
Antidepressive Agents/metabolism , Depressive Disorder, Major/metabolism , Glutamic Acid/metabolism , Nitric Oxide Synthase Type I/drug effects , Paroxetine/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Hippocampus/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Male , Metabolomics , Mice , Mice, Inbred DBA , Nitric Oxide Synthase Type I/metabolism , Paroxetine/administration & dosage , Paroxetine/pharmacology , Proteomics , Receptors, N-Methyl-D-Aspartate/metabolism , Swimming/physiology , Ubiquitin
3.
J Proteomics ; 151: 97-105, 2017 01 16.
Article in English | MEDLINE | ID: mdl-27321914

ABSTRACT

Schizophrenia is a complex disorder hypothesized to develop from a combination of genetic, neurodevelopmental, and environmental factors. Molecules that are directly involved in the pathogenesis of schizophrenia and may serve as biomarker candidates can be identified with "omics" approaches such as proteomics and peptidomics. In this context, we performed a peptidomic study in schizophrenia postmortem brains, to our knowledge the first such study in schizophrenia patients. We investigated the anterior temporal lobe (ATL) and corpus callosum (CC) by liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) and a label-free ion quantification technique based on data-dependent acquisition (DDA). Results indicated alterations in a specific intracellular neurogranin peptide in both the ATL and CC and a decrease of PepH, a fragment of histone H2B type 1-H intracellular peptide, in the ATL. PepH was tested in serum-deprived Neuro2A cells and showed a protective effect against cell death. Cells were also challenged with lipopolysaccharide (LPS), and PepH was able to prevent the endotoxic effects of LPS. Our data suggest that specific intracellular peptides are altered in schizophrenia patients. The potential biological activity of PepH supports intracellular peptides as novel targets in the study not only of schizophrenia but also of other neuropsychiatric diseases. BIOLOGICAL SIGNIFICANCE: Psychiatric disorders are considerably more difficult to diagnose in their early stages. Usually, by the time the diagnosis is clear and clinical treatment can be started, the disorder is already established and thus of greater severity. Consequently, the scientific community has been searching for biomarker candidates that can aid the early detection of such disorders and for novel therapeutics to improve treatment or at least delay disease progression. Moreover, key molecules involved in the establishment of psychiatric diseases may help the understanding of their pathogenesis and thus drive the development of more effective treatments. The present work screened peptides that might be possible novel targets to control cell machinery in schizophrenia and identified an intracellular peptide with potential cytoprotective activity. To our knowledge, this is the first peptidomic study in schizophrenia patients.


Subject(s)
Corpus Callosum/chemistry , Peptides/analysis , Schizophrenia/pathology , Temporal Lobe/chemistry , Biomarkers/analysis , Cell Death/drug effects , Cell Line , Chromatography, Liquid , Corpus Callosum/pathology , Histones/analysis , Humans , Neurogranin/analysis , Proteomics/methods , Tandem Mass Spectrometry , Temporal Lobe/pathology
4.
J. Proteomics ; 151: 97-105, 2017.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: but-ib13584

ABSTRACT

Schizophrenia is a complex disorder hypothesized to develop from a combination of genetic, neurodevelopmental, and environmental factors. Molecules that are directly involved in the pathogenesis of schizophrenia and may serve as biomarker candidates can be identified with "omics" approaches such as proteomics and peptidomics. In this context, we performed a peptidomic study in schizophrenia postmortem brains, to our knowledge the first such study in schizophrenia patients. We investigated the anterior temporal lobe (ATL) and corpus callosum (CC) by liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) and a label-free ion quantification technique based on data-dependent acquisition (DDA). Results indicated alterations in a specific intracellular neurogranin peptide in both the ATL and CC and a decrease of PepH, a fragment of his tone H2B type 1-H intracellular peptide, in the ATL. PepH was tested in serum-deprived Neuro2A cells and showed a protective effect against cell death. Cells were also challenged with lipopolysaccharide (LPS), and PepH was able to prevent the endotoxic effects of LPS. Our data suggest that specific intracellular peptides are altered in schizophrenia patients. The potential biological activity of PepH supports intracellular peptides as novel targets in the study not only of schizophrenia but also of other neuropsychiatric diseases. Biological significance: Psychiatric disorders are considerably more difficult to diagnose in their early stages. Usually, by the time the diagnosis is clear and clinical treatment can be started, the disorder is already established and thus of greater severity. Consequently, the scientific community has been searching for biomarker candidates that can aid the early detection of such disorders and for novel therapeutics to improve treatment or at least delay disease progression. Moreover, key molecules involved in the establishment of psychiatric diseases may help the understanding of their pathogenesis and thus drive the development of more effective treatments. The present work screened peptides that might be possible novel targets to control cell machinery in schizophrenia and identified an intracellular peptide with potential cytoprotective activity. To our knowledge, this is the first peptidomic study in schizophrenia patients.

5.
Mol Psychiatry ; 21(2): 277-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-25849320

ABSTRACT

Psychotropic medications target glycogen synthase kinase 3ß (GSK3ß), but the functional integration with other factors relevant for drug efficacy is poorly understood. We discovered that the suggested psychiatric risk factor FK506 binding protein 51 (FKBP51) increases phosphorylation of GSK3ß at serine 9 (pGSK3ß(S9)). FKBP51 associates with GSK3ß mainly through its FK1 domain; furthermore, it also changes GSK3ß's heterocomplex assembly by associating with the phosphatase PP2A and the kinase cyclin-dependent kinase 5. FKBP51 acts through GSK3ß on the downstream targets Tau, ß-catenin and T-cell factor/lymphoid enhancing factor (TCF/LEF). Lithium and the antidepressant (AD) paroxetine (PAR) functionally synergize with FKBP51, as revealed by reporter gene and protein association analyses. Deletion of FKBP51 blunted the PAR- or lithium-induced increase in pGSK3ß(S9) in cells and mice and attenuated the behavioral effects of lithium treatment. Clinical improvement in depressive patients was predicted by baseline GSK3ß pathway activity and by pGSK3ß(S9) reactivity to ex vivo treatment of peripheral blood mononuclear lymphocytes with lithium or PAR. In sum, FKBP51-directed GSK3ß activity contributes to the action of psychotropic medications. Components of the FKBP51-GSK3ß pathway may be useful as biomarkers predicting AD response and as targets for the development of novel ADs.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Tacrolimus Binding Proteins/genetics , Adult , Animals , Antidepressive Agents/pharmacology , Biomarkers/blood , Cell Culture Techniques , Cell Line , Cyclin-Dependent Kinase 5 , Female , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Leukocytes, Mononuclear/metabolism , Lithium , Male , Mice , Middle Aged , Phosphorylation/drug effects , Psychotropic Drugs/pharmacology , Signal Transduction/drug effects , Tacrolimus Binding Proteins/metabolism , beta Catenin/metabolism
6.
Mol Psychiatry ; 20(9): 1108-19, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25266124

ABSTRACT

It has been postulated that schizophrenia (SZ) is related to a lower expression of brain-derived neurotrophic factor (BDNF). In the past few years, an increasing number of divergent clinical studies assessing BDNF in serum and plasma have been published. It is now possible to verify the relationship between BDNF levels and severity of symptoms in SZ as well as the effects of antipsychotic drugs on BDNF using meta-analysis. The aims of this study were to verify if peripheral BDNF is decreased in SZ, whether its levels are correlated with positive and negative symptomatology and if BDNF levels change after antipsychotic treatment. This report consists of two distinct meta-analyses of peripheral BDNF in SZ including a total of 41 studies and more than 7000 participants: (1) peripheral BDNF levels in serum and plasma were moderately reduced in SZ compared with controls. Notably, this decrease was accentuated with the disease duration. However, the extent of peripheral BDNF level decrease did not correlate with the severity of positive and negative symptoms. (2) In plasma, but not serum, peripheral BDNF levels are consistently increased after antipsychotic treatment irrespective of the patient's response to medication. In conclusion, there is compelling evidence that there are decreased levels of peripheral BDNF in SZ, in parallel to previously described reduced cerebral BDNF expression. It remains unclear whether these systemic changes are causally related to the development of SZ or if they are merely a pathologic epiphenomenon.


Subject(s)
Antipsychotic Agents/therapeutic use , Brain-Derived Neurotrophic Factor/blood , Schizophrenia/blood , Schizophrenia/drug therapy , Adult , Biomarkers/blood , Case-Control Studies , Female , Humans , Male , Middle Aged , Young Adult
7.
Transl Psychiatry ; 4: e478, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25369145

ABSTRACT

Fluoxetine is the only psychopharmacological agent approved for depression by the US Food and Drug Administration for children and is commonly used therapeutically in a variety of neurodevelopmental disorders. Therapeutic response shows high individual variability, and severe side effects have been observed. In the current study we set out to identify biomarkers of response to fluoxetine as well as biomarkers that correlate with impulsivity, a measure of reward delay behavior and potential side effect of the drug, in juvenile male rhesus monkeys. The study group was also genotyped for polymorphisms of monoamine oxidase A (MAOA), a gene that has been associated with psychiatric disorders. We used peripheral metabolite profiling of blood and cerebrospinal fluid (CSF) from animals treated daily with fluoxetine or vehicle for one year. Fluoxetine response metabolite profiles and metabolite/reward delay behavior associations were evaluated using multivariate analysis. Our analyses identified a set of plasma and CSF metabolites that distinguish fluoxetine- from vehicle-treated animals and metabolites that correlate with impulsivity. Some metabolites displayed an interaction between fluoxetine and MAOA genotype. The identified metabolite biomarkers belong to pathways that have important functions in central nervous system physiology. Biomarkers of response to fluoxetine in the normally functioning brain of juvenile nonhuman primates may aid in finding predictors of response to treatment in young psychiatric populations and in progress toward the realization of a precision medicine approach in the area of neurodevelopmental disorders.


Subject(s)
Delay Discounting/drug effects , Fluoxetine/metabolism , Impulsive Behavior/drug effects , Macaca mulatta/metabolism , Selective Serotonin Reuptake Inhibitors/metabolism , Animals , Biomarkers/metabolism , Fluoxetine/pharmacology , Individuality , Male , Monoamine Oxidase/genetics , Selective Serotonin Reuptake Inhibitors/pharmacology
8.
Transl Psychiatry ; 4: e481, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25386958

ABSTRACT

Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, has fast-acting antidepressant activities and is used for major depressive disorder (MDD) patients who show treatment resistance towards drugs of the selective serotonin reuptake inhibitor (SSRI) type. In order to better understand Ketamine's mode of action, a prerequisite for improved drug development efforts, a detailed understanding of the molecular events elicited by the drug is mandatory. In the present study we have carried out a time-dependent hippocampal metabolite profiling analysis of mice treated with Ketamine. After a single injection of Ketamine, our metabolomics data indicate time-dependent metabolite level alterations starting already after 2 h reflecting the fast antidepressant effect of the drug. In silico pathway analyses revealed that several hippocampal pathways including glycolysis/gluconeogenesis, pentose phosphate pathway and citrate cycle are affected, apparent by changes not only in metabolite levels but also connected metabolite level ratios. The results show that a single injection of Ketamine has an impact on the major energy metabolism pathways. Furthermore, seven of the identified metabolites qualify as biomarkers for the Ketamine drug response.


Subject(s)
Behavior, Animal/drug effects , Excitatory Amino Acid Antagonists/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Ketamine/metabolism , Metabolic Networks and Pathways/drug effects , Animals , Biomarkers/metabolism , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/administration & dosage , Ketamine/pharmacology , Male , Metabolomics , Mice , Mice, Inbred C57BL , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
9.
Transl Psychiatry ; 4: e352, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24473443

ABSTRACT

Major depressive disorder (MDD) is one of the leading causes of global disability. It is a risk factor for noncompliance with medical treatment, with about 40% of patients not responding to currently used antidepressant drugs. The identification and clinical implementation of biomarkers that can indicate the likelihood of treatment response are needed in order to predict which patients will benefit from an antidepressant drug. While analyzing the blood plasma proteome collected from MDD patients before the initiation of antidepressant medication, we observed different fibrinogen alpha (FGA) levels between drug responders and nonresponders. These results were replicated in a second set of patients. Our findings lend further support to a recently identified association between MDD and fibrinogen levels from a large-scale study.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder, Major/blood , Depressive Disorder, Major/drug therapy , Fibrinogen/analysis , Treatment Outcome , Adult , Aged , Biomarkers/blood , Female , Humans , Male , Middle Aged , Proteome/analysis , Young Adult
12.
Pharmacopsychiatry ; 44 Suppl 1: S2-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21544742

ABSTRACT

Understanding mental disorders and their neurobiological basis encompasses the conceptual management of "complexity" and "dynamics". For example, affective disorders exhibit several fluctuating state variables on psychological and biological levels and data collected of these systems levels suggest quasi-chaotic periodicity leading to use concepts and tools of the mathematics of nonlinear dynamic systems. Regarding this, we demonstrate that the concept of "Dynamic Diseases" could be a fruitful way for theory and empirical research in neuropsychiatry. In a first step, as an example, we focus on the analysis of dynamic cortisol regulation that is important for understanding depressive disorders. In this case, our message is that extremely complex phenomena of a disease may be explained as resulting from perplexingly simple nonlinear interactions of a very small number of variables. Additionally, we propose that and how widely used complex circuit diagrams representing the macroanatomic structures and connectivities of the brain involved in major depression or other mental disorders may be "animated" by quantification, even by using expert-based estimations (dummy variables). This method of modeling allows to develop exploratory computer-based numerical models that encompass the option to explore the system by computer simulations (in-silico experiments). Also inter- and intracellular molecular networks involved in affective disorders could be modeled by this procedure. We want to stimulate future research in this theoretical context.


Subject(s)
Depression/physiopathology , Depressive Disorder/physiopathology , Disease , Mental Disorders/physiopathology , Mood Disorders/physiopathology , Neurobiology , Systems Biology , Brain/anatomy & histology , Brain/pathology , Brain/physiopathology , Computer Simulation , Depressive Disorder/pathology , Humans , Hydrocortisone/metabolism , Mental Disorders/pathology , Models, Biological , Mood Disorders/metabolism , Mood Disorders/pathology , Neuropsychiatry , Nonlinear Dynamics , Signal Transduction
13.
Pharmacopsychiatry ; 44 Suppl 1: S54-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21544746

ABSTRACT

The etiopathogenesis of many psychiatric illnesses remains unclear and a variety of these diseases can coexist, partly mimicking each other while contributing to and distorting symptomatic expressions. To understand the processes involved, it is necessary to unravel signalling pathways, complex interaction networks and metabolic alterations involving a plethora of anatomical components. When addressing such largely obscure mechanisms, primary data mainly based on genomics and differential gene expression patterns turns out to be of limited usefulness. Numerous direct as well as very indirect processes modulate and dissociate gene expression from protein functions and physiological effects. Proteomics approaches that utilise metabolic labelling and high-throughput mass spectrometry to provide proteome dynamics data need to be utilised. However, the data thus gathered encompasses a complex assembly of numerous types of intermixed cells, representing biological processes that occur in both time and space across several scalar levels. The complexities represented are such that to analytically approach these diseases, a systems standpoint becomes necessary. This implies multiple experimental interrogations in an iterative interplay between experimentation and modelling. While this may be reasonably considered in the context of in vitro systems, it can hardly be contemplated when addressing CNS tissues from heterogeneous human origins, thereby imposing serious constraints upon the investigation of human cognitive disorders. In this article, the authors expose a paradigm that addresses and alleviates at least some of these major difficulties. Based on the reasoned utilisation of trait animal models and human material, this approach has already started to deliver novel and directly exploitable knowledge.


Subject(s)
Brain/physiopathology , Mental Disorders/physiopathology , Models, Biological , Neurotransmitter Agents/physiology , Proteomics/methods , Systems Biology/methods , Animals , Brain/pathology , Disease Models, Animal , Epigenesis, Genetic , Genomics , Humans , Mass Spectrometry , Mental Disorders/genetics , Mental Disorders/pathology , Proteins/metabolism , Proteome , Signal Transduction , Transcription, Genetic
14.
Transl Psychiatry ; 1: e58, 2011 Dec 13.
Article in English | MEDLINE | ID: mdl-22832350

ABSTRACT

Currently used antidepressants elevate monoamine levels in the synaptic cleft. There is good reason to assume that this is not the only source for antidepressant therapeutic activities and that secondary downstream effects may be relevant for alleviating symptoms of depression. We attempted to elucidate affected biochemical pathways downstream of monoamine reuptake inhibition by interrogating metabolomic profiles in DBA/2Ola mice after chronic paroxetine treatment. Metabolomic changes were investigated using gas chromatography-mass spectrometry profiling and group differences were analyzed by univariate and multivariate statistics. Pathways affected by antidepressant treatment were related to energy metabolism, amino acid metabolism and hormone signaling. The identified pathways reveal further antidepressant therapeutic action and represent targets for drug development efforts. A comparison of the central nervous system with blood plasma metabolite alterations identified GABA, galactose-6-phosphate and leucine as biomarker candidates for assessment of antidepressant treatment effects in the periphery.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Biogenic Monoamines/biosynthesis , Drug Delivery Systems/methods , Hippocampus/metabolism , Metabolome/drug effects , Paroxetine/pharmacology , Animals , Behavior, Animal/drug effects , Biogenic Monoamines/antagonists & inhibitors , Biomarkers/metabolism , Gas Chromatography-Mass Spectrometry , Hippocampus/drug effects , Mice , Mice, Inbred DBA
15.
Mol Psychiatry ; 15(7): 702-11, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19139748

ABSTRACT

In our biomarker identification efforts, we have reported earlier on a protein that differs in its electrophoretic mobility between mouse lines bred either for high or low trait anxiety. The altered electrophoretic behavior of enolase phosphatase (EP) is now identified to be caused by two single-nucleotide polymorphisms. In both cases, the genetic polymorphism introduces an amino acid change in the protein's sequence resulting in differential mobility on SDS gels. This was shown by recombinantly expressing the two EP isoforms. Functional studies indicate that the EP isoform from the high anxiety mouse line has a lower enzymatic activity than does its low anxiety mouse counterpart. EP is a member of the methionine salvage pathway that is responsible for the synthesis of S-adenosyl-L-methionine, a natural compound with potential antidepressant activities. In addition, it is linked to the polyamine pathway whose members have functions in anxiety/depression-related behaviors. In a freely-segregating F2 panel, both single-nucleotide polymorphisms were significantly associated with locomotion-independent trait anxiety, further supporting a functional role of EP for this phenotype. The study shows that proteomic analysis can reveal genotypic differences relevant for the phenotype. The identified protein alterations, in turn, can expose metabolic pathways pertinent to the behavioral phenotype.


Subject(s)
Anxiety/metabolism , Disease Models, Animal , Genotype , Isoenzymes/metabolism , Multienzyme Complexes/genetics , Phosphoric Monoester Hydrolases/genetics , Proteomics/methods , Animals , Brain/metabolism , Humans , Isoenzymes/genetics , Male , Mice , Mice, Inbred Strains , Models, Genetic , Multienzyme Complexes/metabolism , Phosphoric Monoester Hydrolases/metabolism , Polyamines/metabolism , Polymorphism, Single Nucleotide
16.
Pharmacopsychiatry ; 41 Suppl 1: S70-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18756423

ABSTRACT

The pathobiology of psychiatric disorders remains mostly obscure. Diagnosis is often imprecise and current treatments are empirical and at best symptomatic. The identification of biomarkers can help with developing improved drugs and establishing more precise disease diagnoses. Proteins are prime biomarker candidates, because of the central role they play in both, disease etiology and treatment. Today's high throughput methods are capable to detect potential biomarkers by screening complex proteome mixtures of different disease states. A meaningful interpretation of the candidates is indispensable for deeper insights into affective disorders and requires investigation on multiple levels of the cellular system. Thus a systems biology approach is critical to understand and explain the behavior of biomarker candidates and to make use of them in psychiatry.


Subject(s)
Biomarkers/metabolism , Mental Disorders , Proteomics , Systems Biology , Animals , Humans , Mental Disorders/diagnosis , Mental Disorders/metabolism , Mental Disorders/physiopathology
17.
Handb Exp Pharmacol ; (169): 547-60, 2005.
Article in English | MEDLINE | ID: mdl-16594272

ABSTRACT

Proteomics, the comprehensive analysis of the protein complement of the genome of an organism, is becoming an increasingly important discipline for the identification of disease targets. In addition, the effects of drug treatment and metabolism can now be studied on the protein level in a comprehensive manner.


Subject(s)
Pharmacogenetics , Proteome , Clinical Medicine , Electrophoresis, Gel, Two-Dimensional , Humans , Proteins/isolation & purification
18.
EMBO J ; 20(20): 5692-702, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11598012

ABSTRACT

Mutations in Bruton's tyrosine kinase (Btk) result in X-linked agammaglobulinemia (XLA) in humans and X-linked immunodeficiency (xid) in mice. While targeted disruption of the protein kinase C-beta (PKCbeta) gene in mice results in an immunodeficiency similar to xid, the overall tyrosine phosphorylation of Btk is significantly enhanced in PKCbeta-deficient B cells. We provide direct evidence that PKCbeta acts as a feedback loop inhibitor of Btk activation. Inhibition of PKCbeta results in a dramatic increase in B-cell receptor (BCR)-mediated Ca2+ signaling. We identified a highly conserved PKCbeta serine phosphorylation site in a short linker within the Tec homology domain of Btk. Mutation of this phosphorylation site led to enhanced tyrosine phosphorylation and membrane association of Btk, and augmented BCR and FcepsilonRI-mediated signaling in B and mast cells, respectively. These findings provide a novel mechanism whereby reversible translocation of Btk/Tec kinases regulates the threshold for immunoreceptor signaling and thereby modulates lymphocyte activation.


Subject(s)
Isoenzymes/physiology , Lymphocyte Activation/physiology , Membrane Proteins/metabolism , Protein Kinase C/physiology , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/physiology , 3T3 Cells , Agammaglobulinaemia Tyrosine Kinase , Agammaglobulinemia/genetics , Alleles , Amino Acid Sequence , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Calcium Signaling/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Feedback , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/deficiency , Isoenzymes/genetics , Mast Cells/enzymology , Mast Cells/immunology , Mice , Mice, Knockout , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Mapping , Phosphorylation , Phosphoserine/chemistry , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/deficiency , Protein Kinase C/genetics , Protein Kinase C beta , Protein Processing, Post-Translational , Protein Structure, Tertiary , Protein Transport , Protein-Tyrosine Kinases/chemistry , Receptors, IgE/physiology
19.
J Biol Chem ; 276(25): 22177-82, 2001 Jun 22.
Article in English | MEDLINE | ID: mdl-11301320

ABSTRACT

Promoter-bound steroid receptors activate gene expression by recruiting members of the p160 family of coactivators. Many steroid receptors, most notably the progesterone and estrogen receptors, are regulated both by cognate hormone and independently by growth factors. Here we show that epidermal growth factor regulates the activities of the p160 GRIP1 through the extracellular signal-regulated kinase (ERK) family of mitogen-activated protein kinases. ERKs phosphorylate GRIP1 at a specific site, Ser-736, the integrity of which is required for full growth factor induction of GRIP1 transcriptional activation and coactivator function. We propose that growth factors signal to nuclear receptors in part by targeting the p160 coactivators.


Subject(s)
Epidermal Growth Factor/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Steroid/metabolism , Signal Transduction , Transcription Factors/metabolism , HeLa Cells , Humans , Nuclear Receptor Coactivator 2 , Phosphorylation
20.
Mol Cell Biol ; 20(20): 7463-79, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003644

ABSTRACT

Splicing of the c-src N1 exon in neuronal cells depends in part on an intronic cluster of RNA regulatory elements called the downstream control sequence (DCS). Using site-specific cross-linking, RNA gel shift, and DCS RNA affinity chromatography assays, we characterized the binding of several proteins to specific sites along the DCS RNA. Heterogeneous nuclear ribonucleoprotein (hnRNP) H, polypyrimidine tract binding protein (PTB), and KH-type splicing-regulatory protein (KSRP) each bind to distinct elements within this sequence. We also identified a new 60-kDa tissue-specific protein that binds to the CUCUCU splicing repressor element of the DCS RNA. This protein was purified, partially sequenced, and cloned. The new protein (neurally enriched homolog of PTB [nPTB]) is highly homologous to PTB. Unlike PTB, nPTB is enriched in the brain and in some neural cell lines. Although similar in sequence, nPTB and PTB show significant differences in their properties. nPTB binds more stably to the DCS RNA than PTB does but is a weaker repressor of splicing in vitro. nPTB also greatly enhances the binding of two other proteins, hnRNP H and KSRP, to the DCS RNA. These experiments identify specific cooperative interactions between the proteins that assemble onto an intricate splicing-regulatory sequence and show how this hnRNP assembly is altered in different cell types by incorporating different but highly related proteins.


Subject(s)
Nerve Tissue Proteins/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins/metabolism , Trans-Activators , Amino Acid Sequence , Base Sequence , Cell Line , Chromatography, Affinity , Cloning, Molecular , Heterogeneous-Nuclear Ribonucleoprotein Group F-H , Heterogeneous-Nuclear Ribonucleoproteins , Humans , Models, Genetic , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/isolation & purification , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , Organ Specificity , Phylogeny , Polypyrimidine Tract-Binding Protein , Protein Binding , RNA Splicing/genetics , RNA, Heterogeneous Nuclear/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/isolation & purification , Regulatory Sequences, Nucleic Acid , Ribonucleoproteins/chemistry , Ribonucleoproteins/genetics , Ribonucleoproteins/isolation & purification , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...