Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 441
Filter
1.
Article in English | MEDLINE | ID: mdl-38906412

ABSTRACT

DARPP-32 (dopamine and cAMP-regulated phosphoprotein Mr. 32 kDa) is a phosphoprotein that is modulated by multiple receptors integrating intracellular pathways and playing roles in various physiological functions. It is regulated by dopaminergic receptors through the cAMP/protein kinase A (PKA) pathway, which modulates the phosphorylation of threonine 34 (Thr34). When phosphorylated at Thr34, DARPP-32 becomes a potent protein phosphatase-1 (PP1) inhibitor. Since dopamine is involved in the development of GABAergic neurons and DARPP-32 is expressed in the developing brain, it is possible that DARPP-32 has a role in GABAergic neuronal development. We cloned the zebrafish darpp-32 gene (ppp1r1b) gene and observed that it is evolutionarily conserved in its inhibitory domain (Thr34 and surrounding residues) and the docking motif (residues 7-11 (KKIQF)). We also characterized darpp-32 protein expression throughout the 5 days post-fertilization (dpf) zebrafish larval brain by immunofluorescence and demonstrated that darpp-32 is mainly expressed in regions that receive dopaminergic projections (pallium, subpallium, preoptic region, and hypothalamus). We demonstrated that dopamine acutely suppressed darpp-32 activity by reducing the levels of p-darpp-32 in the 5dpf zebrafish larval brain. In addition, the knockdown of darpp-32 resulted in a decrease in the number of GABAergic neurons in the subpallium of the 5dpf larval brain, with a concomitant increase in the number of DAergic neurons. Finally, we demonstrated that darpp-32 downregulation during development reduced the motor behavior of 5dpf zebrafish larvae. Thus, our observations suggest that darpp-32 is an evolutionarily conserved regulator of dopamine receptor signaling and is required for the formation of GABAergic neurons in the developing telencephalon.


Subject(s)
Dopamine and cAMP-Regulated Phosphoprotein 32 , Dopamine , GABAergic Neurons , Telencephalon , Zebrafish , Animals , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , GABAergic Neurons/metabolism , Telencephalon/metabolism , Telencephalon/embryology , Dopamine/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Animals, Genetically Modified , Gene Expression Regulation, Developmental/physiology
2.
Brain Res ; 1839: 149017, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38768935

ABSTRACT

Parkinson's disease (PD) is a complex disorder, primarily of idiopathic origin, with environmental stressors like rotenone and manganese linked to its development. This study explores their potential interaction and resulting neurotoxicity, aiming to understand how environmental factors contribute to PD. In an eight-day experiment, male Wistar rats weighing 280-300 g were subjected to rotenone, manganese, or a combination of both. Various parameters were assessed, including body weight, behavior, serum markers, tissue damage, protein levels (tyrosine hydroxylase, Dopamine- and cAMP-regulated neuronal phosphoprotein -DARPP-32-, and α-synuclein), and mitochondrial function. Manganese heightened rotenone's impact on reducing food intake without causing kidney or liver dysfunction. However, the combined exposure intensified neurotoxicity, which was evident in augmented broken nuclei and decreased tyrosine hydroxylase and DARPP-32 levels in the striatum. While overall mitochondrial function was preserved, co-administration reduced complex IV activity in the midbrain and liver. In conclusion, our findings revealed a parallel toxic effect induced by rotenone and manganese. Notably, while these substances do not target the same dopaminergic regions, a notable escalation in toxicity is evident in the striatum, the brain region where their toxic effects converge. This study highlights the need for further exploration regarding the interaction of environmental factors and their possible impact on the etiology of PD.


Subject(s)
Manganese , Rats, Wistar , Rotenone , Tyrosine 3-Monooxygenase , Animals , Rotenone/toxicity , Male , Manganese/toxicity , Rats , Tyrosine 3-Monooxygenase/metabolism , Brain/drug effects , Brain/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , alpha-Synuclein/metabolism , Neurotoxicity Syndromes/metabolism , Corpus Striatum/metabolism , Corpus Striatum/drug effects
3.
Sci Rep ; 13(1): 21163, 2023 11 30.
Article in English | MEDLINE | ID: mdl-38036593

ABSTRACT

DARPP-32 is a key regulator of protein-phosphatase-1 (PP-1) and protein kinase A (PKA), with its function dependent upon its phosphorylation state. We previously identified DKK1 and GRB7 as genes with linked expression using Artificial Neural Network (ANN) analysis; here, we determine protein expression in a large cohort of early-stage breast cancer patients. Low levels of DARPP-32 Threonine-34 phosphorylation and DKK1 expression were significantly associated with poor patient prognosis, while low levels of GRB7 expression were linked to better survival outcomes. To gain insight into mechanisms underlying these associations, we analysed the transcriptome of T47D breast cancer cells following DARPP-32 knockdown. We identified 202 differentially expressed transcripts and observed that some overlapped with genes implicated in the ANN analysis, including PTK7, TRAF5, and KLK6, amongst others. Furthermore, we found that treatment of DARPP-32 knockdown cells with 17ß-estradiol or PKA inhibitor fragment (6-22) amide led to the differential expression of 193 and 181 transcripts respectively. These results underscore the importance of DARPP-32, a central molecular switch, and its downstream targets, DKK1 and GRB7 in breast cancer. The discovery of common genes identified by a combined patient/cell line transcriptomic approach provides insights into the molecular mechanisms underlying differential breast cancer prognosis and highlights potential targets for therapeutic intervention.


Subject(s)
Breast Neoplasms , Female , Humans , Breast Neoplasms/metabolism , Cell Adhesion Molecules/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Phosphorylation , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Transcriptome
4.
J Comp Neurol ; 531(8): 888-920, 2023 06.
Article in English | MEDLINE | ID: mdl-37002560

ABSTRACT

The dorsal striatum forms a central node of the basal ganglia interconnecting the neocortex and thalamus with circuits modulating mood and movement. Striatal projection neurons (SPNs) include relatively intermixed populations expressing D1-type or D2-type dopamine receptors (dSPNs and iSPNs) that give rise to the direct (D1) and indirect (D2) output systems of the basal ganglia. Overlaid on this organization is a compartmental organization, in which a labyrinthine system of striosomes made up of sequestered SPNs is embedded within the larger striatal matrix. Striosomal SPNs also include D1-SPNs and D2-SPNs, but they can be distinguished from matrix SPNs by many neurochemical markers. In the rodent striatum the key signaling molecule, DARPP-32, is a exception to these compartmental expression patterns, thought to befit its functions through opposite actions in both D1- and D2-expressing SPNs. We demonstrate here, however, that in the dorsal human striatum, DARPP-32 is concentrated in the neuropil and SPNs of striosomes, especially in the caudate nucleus and dorsomedial putamen, relative to the matrix neuropil in these regions. The generally DARPP-32-poor matrix contains scattered DARPP-32-positive cells. DARPP-32 cell bodies in both compartments proved negative for conventional intraneuronal markers. These findings raise the potential for specialized DARPP-32 expression in the human striosomal system and in a set of DARPP-32-positive neurons in the matrix. If DARPP-32 immunohistochemical positivity predicts differential functional DARPP-32 activity, then the distributions demonstrated here could render striosomes and dispersed matrix cells susceptible to differential signaling through cAMP and other signaling systems in health and disease. DARPP-32 is highly concentrated in cells and neuropil of striosomes in post-mortem human brain tissue, particularly in the dorsal caudate nucleus. Scattered DARPP-32-positive cells are found in the human striatal matrix. Calbindin and DARPP-32 do not colocalize within every spiny projection neuron in the dorsal human caudate nucleus.


Subject(s)
Caudate Nucleus , Corpus Striatum , Humans , Corpus Striatum/metabolism , Caudate Nucleus/metabolism , Basal Ganglia , Neurons/metabolism , Receptors, Dopamine D2/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Neuropil/metabolism
5.
Neuropharmacology ; 228: 109462, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36792029

ABSTRACT

Dopamine plays a central role in the regulation of psychomotor functions in the brain. Furthermore, the dopaminergic system is involved in the ictogenesis in human patients and animal models of epilepsy. Dopamine and cAMP-regulated phosphoprotein, 32 kDa (DARPP-32) plays an important role in the regulation of interactions between dopamine and glutamate receptors in neurons. Indeed, SKF 83822 (a specific D1 receptor agonist) facilitates DARPP-32-mediated protein phosphatase 1 (PP1) inhibition leading to the increase in phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor (AMPAR), which potentiates channel activities and currents and thereby generates seizure activity. In the present study, we found that pyridoxal-5'-phosphate phosphatase/chronophin (PLPP/CIN), a selective phosphatase for serine (S) residues, attenuated seizure susceptibility in response to SKF 83822 by dephosphorylating DARPP-32 S97 site. Similarly, inhibition of DARPP-32 S97 phosphorylation by 2-[4,5,6,7-Tetrabromo-2-(dimethylamino)-1H-benzo[d]imidazole-1-yl]acetic acid (TMCB; a selective casein kinase 2 inhibitor) attenuated SKF 83822-induced seizure activity. These inhibitory effects of PLPP/CIN and TMCB were relevant to the regulations of DARPP-32-PP1-AMPAR signaling pathway. Therefore, our findings suggest that PLPP/CIN may be a modulator in dopaminergic neurotransmission as well as glutamatergic systems, and that the PLPP/CIN-mediated DARPP-32 regulation may be one of the potential therapeutic targets for medication of seizure or epilepsy induced by D1 receptor hyperactivation.


Subject(s)
Dopamine , Phosphates , Mice , Animals , Humans , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Dopamine/metabolism , Phosphates/metabolism , Synaptic Transmission , Phosphorylation , Seizures/metabolism , Receptors, Dopamine D1/metabolism , Protein Phosphatase 1/metabolism , Hippocampus
6.
Neurochem Int ; 162: 105438, 2023 01.
Article in English | MEDLINE | ID: mdl-36351540

ABSTRACT

Dopamine regulates psychomotor function by D1 receptor/PKA-dependent phosphorylation of DARPP-32. DARPP-32, phosphorylated at Thr34 by PKA, inhibits protein phosphatase 1 (PP1), and amplifies the phosphorylation of other PKA/PP1 substrates following D1 receptor activation. In addition to the D1 receptor/PKA/DARPP-32 signaling pathway, D1 receptor stimulation is known to activate Rap1/ERK signaling. Rap1 activation is mediated through the phosphorylation of Rasgrp2 (guanine nucleotide exchange factor; activation) and Rap1gap (GTPase-activating protein; inhibition) by PKA. In this study, we investigated the role of PP1 inhibition by phospho-Thr34 DARPP-32 in the D1 receptor-induced phosphorylation of Rasgrp2 and Rap1gap at PKA sites. The analyses in striatal and NAc slices from wild-type and DARPP-32 knockout mice revealed that the phosphorylation of Rasgrp2 at Ser116/Ser117 and Ser586, but not of Rasgrp2 at Ser554 or Rap1gap at Ser441 or Ser499 induced by a D1 receptor agonist, is under the control of the DARPP-32/PP1. The results were supported by pharmacological analyses using a selective PP1 inhibitor, tautomycetin. In addition, analyses using a PP1 and PP2A inhibitor, okadaic acid, revealed that all sites of Rasgrp2 and Rap1gap were regulated by PP2A. Thus, the interactive machinery of DARPP-32/PP1 may contribute to efficient D1 receptor signaling via Rasgrp2/Rap1 in the striatum.


Subject(s)
Corpus Striatum , Neostriatum , Animals , Mice , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/pharmacology , Corpus Striatum/metabolism , Neostriatum/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Signal Transduction , Phosphorylation , Receptors, Dopamine D1/metabolism
7.
Int J Mol Sci ; 23(19)2022 Oct 01.
Article in English | MEDLINE | ID: mdl-36232936

ABSTRACT

Antipsychotics share the common pharmacological feature of antagonizing the dopamine 2 receptor (D2R), which is abundant in the striatum and involved in both the therapeutic and side effects of this drug's class. The pharmacological blockade of striatal D2R, by disinhibiting the D2R-containing medium-sized spiny neurons (MSNs), leads to a plethora of molecular, cellular and behavioral adaptations, which are central in the action of antipsychotics. Here, we focused on the cell type-specific (D2R-MSNs) regulation of some striatal immediate early genes (IEGs), such as cFos, Arc and Zif268. Taking advantage of transgenic mouse models, pharmacological approaches and immunofluorescence analyses, we found that haloperidol-induced IEGs in the striatum required the synergistic activation of A2a (adenosine) and NMDA (glutamate) receptors. At the intracellular signaling level, we found that the PKA/DARPP-32 and mTOR pathways synergistically cooperate to control the induction of IEGs by haloperidol. By confirming and further expanding previous observations, our results provide novel insights into the regulatory mechanisms underlying the molecular/cellular action of antipsychotics in the striatum.


Subject(s)
Antipsychotic Agents , Haloperidol , Adenosine/metabolism , Animals , Antipsychotic Agents/metabolism , Antipsychotic Agents/pharmacology , Corpus Striatum/metabolism , Dopamine/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Genes, Immediate-Early , Glutamates/metabolism , Haloperidol/pharmacology , Mice , Mice, Transgenic , N-Methylaspartate/metabolism , Neurons/metabolism , Receptors, Dopamine D1/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
8.
Neuropharmacology ; 219: 109238, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36055413

ABSTRACT

Dopamine and cAMP-regulated phosphoprotein, 32 kDa (DARPP-32)-mediated protein phosphatase 1 (PP1) inhibition leads to the increase in phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor (AMPAR), which potentiates channel activity and current and thereby may facilitate seizure activity. In the present study, we found that pyridoxal-5'-phosphate phosphatase/chronophin (PLPP/CIN) transiently dephosphorylated DARPP-32 serine (S) 97 site in the early time window, and casein kinase 2 (CK2) subsequently phosphorylated this site in the later time points after kainic acid (KA) injection, which increased the latency of seizure onset in response to KA, but exacerbated the intensity (severity), duration and progression of seizures. TMCB (a CK2 inhibitor) delayed the seizure onset in response to KA, concomitant with the reduced DARPP-32 S97 phosphorylation. Therefore, our findings suggest that PLPP/CIN may play an important role in the latency of seizure onset via DARPP-32-PP1-AMPAR signaling pathway, and may be one of the potential therapeutic targets for medication of seizure or epilepsy.


Subject(s)
Kainic Acid , Serine , Animals , Casein Kinase II/metabolism , Dopamine/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Hippocampus/metabolism , Kainic Acid/pharmacology , Mice , Phosphates/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Phosphatase 1/metabolism , Pyridoxal , Seizures/chemically induced , Seizures/drug therapy , Seizures/metabolism , Serine/metabolism , Serine/pharmacology
9.
Proc Natl Acad Sci U S A ; 119(33): e2207489119, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35939707

ABSTRACT

The mechanistic target of rapamycin (mTOR) is assembled into signaling complexes of mTORC1 or mTORC2, and plays key roles in cell metabolism, stress response, and nutrient and growth factor sensing. Accumulating evidence from human and animal model studies has demonstrated a pathogenic role of hyperactive mTORC1 in age-related macular degeneration (AMD). The retinal pigment epithelium (RPE) is a primary injury site in AMD. In mouse models of RPE-specific deletion of Tuberous sclerosis 1 (Tsc1), which encodes an upstream suppressor of mTORC1, the hyperactivated mTORC1 metabolically reprogrammed the RPE and led to the degeneration of the outer retina and choroid (CH). In the current study, we use single-cell RNA sequencing (scRNA-seq) to identify an RPE mTORC1 downstream protein, dopamine- and cyclic AMP-regulated phosphoprotein of molecular weight 32,000 (DARPP-32). DARPP-32 was not found in healthy RPE but localized to drusen and basal linear deposits in human AMD eyes. In animal models, overexpressing DARPP-32 by adeno-associated virus (AAV) led to abnormal RPE structure and function. The data indicate that DARPP-32 is a previously unidentified signaling protein subjected to mTORC1 regulation and may contribute to RPE degeneration in AMD.


Subject(s)
Dopamine and cAMP-Regulated Phosphoprotein 32 , Macular Degeneration , Mechanistic Target of Rapamycin Complex 1 , Retinal Pigment Epithelium , Animals , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Enzyme Activation , Humans , Macular Degeneration/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Signal Transduction
10.
J Comp Neurol ; 530(5): 792-803, 2022 04.
Article in English | MEDLINE | ID: mdl-34545948

ABSTRACT

Adult male zebra finches (Taeniopygia guttata) continually incorporate adult-born neurons into HVC, a telencephalic brain region necessary for the production of learned song. These neurons express activity-dependent immediate early genes (e.g., zenk and c-fos) following song production, suggesting that these neurons are active during song production. Half of these adult-born HVC neurons (HVC NNs) can be backfilled from the robust nucleus of the arcopallium (RA) and are a part of the vocal motor pathway underlying learned song production, but the other half do not backfill from RA, and they remain to be characterized. Here, we used cell birth-dating, retrograde tract tracing, and immunofluorescence to demonstrate that half of all HVC NNs express the phosphoprotein DARPP-32, a protein associated with dopamine receptor expression. We also demonstrate that DARPP-32+ HVC NNs are contacted by tyrosine hydroxylase immunoreactive fibers, suggesting that they receive catecholaminergic input, have transiently larger nuclei than DARPP-32-neg HVC NNs, and do not backfill from RA. Taken together, these findings help characterize a group of HVC NNs that have no apparent projections to RA and so far have eluded positive identification other than HVC NN status.


Subject(s)
Brain/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , High Vocal Center/metabolism , Neurons/metabolism , Vocalization, Animal/physiology , Age Factors , Animals , Finches
11.
Oncogene ; 41(1): 83-98, 2022 01.
Article in English | MEDLINE | ID: mdl-34675407

ABSTRACT

Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI)-refractory lung adenocarcinoma (LUAD) progression is a major clinical problem. New approaches to predict and prevent acquired resistance to EGFR TKIs are urgently needed. Here, we show that dopamine and cyclic AMP-regulated phosphoprotein, Mr 32000 (DARPP-32) physically recruits ERBB3 (HER3) to EGFR to mediate switching from EGFR homodimers to EGFR:ERBB3 heterodimers to bypass EGFR TKI-mediated inhibition by potentiating ERBB3-dependent activation of oncogenic signaling. In paired LUAD patient-derived specimens before and after EGFR TKI-refractory disease progression, we reveal that DARPP-32 and kinase-activated EGFR and ERBB3 proteins are overexpressed upon acquired resistance. In mice, DARPP-32 ablation sensitizes gefitinib-resistant xenografts to EGFR TKIs, while DARPP-32 overexpression increases gefitinib-refractory LUAD progression in gefitinib-sensitive lung tumors. We introduce a DARPP-32-mediated, ERBB3-dependent mechanism the LUAD cells use to evade EGFR TKI-induced cell death, potentially paving the way for the development of therapies to better combat therapy-refractory LUAD progression.


Subject(s)
Adenocarcinoma of Lung/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Molecular Targeted Therapy/methods , Receptor, ErbB-3/metabolism , Adenocarcinoma of Lung/pathology , Animals , Cell Line, Tumor , Humans , Mice
12.
PLoS One ; 16(10): e0258486, 2021.
Article in English | MEDLINE | ID: mdl-34648564

ABSTRACT

Huntington's disease (HD) results from an expansion mutation in the polyglutamine tract in huntingtin. Although huntingtin is ubiquitously expressed in the body, the striatum suffers the most severe pathology. Rhes is a Ras-related small GTP-binding protein highly expressed in the striatum that has been reported to modulate mTOR and sumoylation of mutant huntingtin to alter HD mouse model pathogenesis. Reports have varied on whether Rhes reduction is desirable for HD. Here we characterize multiple behavioral and molecular endpoints in the Q175 HD mouse model with genetic Rhes knockout (KO). Genetic RhesKO in the Q175 female mouse resulted in both subtle attenuation of Q175 phenotypic features, and detrimental effects on other kinematic features. The Q175 females exhibited measurable pathogenic deficits, as measured by MRI, MRS and DARPP32, however, RhesKO had no effect on these readouts. Additionally, RhesKO in Q175 mixed gender mice deficits did not affect mTOR signaling, autophagy or mutant huntingtin levels. We conclude that global RhesKO does not substantially ameliorate or exacerbate HD mouse phenotypes in Q175 mice.


Subject(s)
GTP-Binding Proteins/genetics , Huntington Disease/pathology , Animals , Biomechanical Phenomena , Body Weight , Brain/physiology , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Female , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/metabolism , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
13.
Cell Prolif ; 54(8): e13082, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34152047

ABSTRACT

OBJECTIVES: To investigate whether human HLA-homozygous induced pluripotent stem cell (iPSC)-derived neural precursor cells (iPSC-NPCs) can provide functional benefits in Huntington's disease (HD), we transplanted them into the YAC128 transgenic HD mouse model. MATERIALS AND METHODS: CHAi001-A, an HLA-homozygous iPSC line (A*33:03-B*44:03-DRB1*13:02), was differentiated into neural precursor cells, and then, they were transplanted into 6 months-old YAC128 mice. Various behavioural and histological analyses were performed for five months after transplantation. RESULTS: Motor and cognitive functions were significantly improved in transplanted animals. Cells transplanted in the striatum showed multipotential differentiation. Five months after transplantation, the donor cells had differentiated into neurons, oligodendrocytes and astrocytes. Transplantation restored DARPP-32 expression, synaptophysin density, myelin basic protein expression in the corpus callosum and astrocyte function. CONCLUSION: Altogether, these results strongly suggest that iPSC-NPCs transplantation induces neuroprotection and functional recovery in a mouse model of HD and should be taken forward for clinical trials in HD patients.


Subject(s)
Cell Differentiation , Huntington Disease/pathology , Neural Stem Cells/transplantation , Animals , Astrocytes/cytology , Astrocytes/metabolism , Behavior, Animal , Cell Line , Corpus Callosum/metabolism , Disease Models, Animal , Disease Progression , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Humans , Huntington Disease/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Maze Learning , Mice , Mice, Transgenic , Myelin Basic Protein/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Phosphoric Monoester Hydrolases/metabolism
14.
Neurobiol Dis ; 157: 105423, 2021 09.
Article in English | MEDLINE | ID: mdl-34144125

ABSTRACT

BACKGROUND: Dravet syndrome is a rare, severe pediatric epileptic encephalopathy associated with intellectual and motor disabilities. Proteomic profiling in a mouse model of Dravet syndrome can provide information about the molecular consequences of the genetic deficiency and about pathophysiological mechanisms developing during the disease course. METHODS: A knock-in mouse model of Dravet syndrome with Scn1a haploinsufficiency was used for whole proteome, seizure, and behavioral analysis. Hippocampal tissue was dissected from two- (prior to epilepsy manifestation) and four- (following epilepsy manifestation) week-old male mice and analyzed using LC-MS/MS with label-free quantification. Proteomic data sets were subjected to bioinformatic analysis including pathway enrichment analysis. The differential expression of selected proteins was confirmed by immunohistochemical staining. RESULTS: The findings confirmed an increased susceptibility to hyperthermia-associated seizures, the development of spontaneous seizures, and behavioral alterations in the novel Scn1a-A1873V mouse model of Dravet syndrome. As expected, proteomic analysis demonstrated more pronounced alterations following epilepsy manifestation. In particular, proteins involved in neurotransmitter dynamics, receptor and ion channel function, synaptic plasticity, astrogliosis, neoangiogenesis, and nitric oxide signaling showed a pronounced regulation in Dravet mice. Pathway enrichment analysis identified several significantly regulated pathways at the later time point, with pathways linked to synaptic transmission and glutamatergic signaling dominating the list. CONCLUSION: In conclusion, the whole proteome analysis in a mouse model of Dravet syndrome demonstrated complex molecular alterations in the hippocampus. Some of these alterations may have an impact on excitability or may serve a compensatory function, which, however, needs to be further confirmed by future investigations. The proteomic data indicate that, due to the molecular consequences of the genetic deficiency, the pathophysiological mechanisms may become more complex during the course of the disease. As a result, the management of Dravet syndrome may need to consider further molecular and cellular alterations. Ensuing functional follow-up studies, this data set may provide valuable guidance for the future development of novel therapeutic approaches.


Subject(s)
Epilepsies, Myoclonic/metabolism , Hippocampus/metabolism , Proteomics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Animals , Behavior, Animal , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Carbon-Nitrogen Ligases/metabolism , Chromatography, Liquid , Disease Models, Animal , Disease Progression , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Elevated Plus Maze Test , Epilepsies, Myoclonic/genetics , Epilepsies, Myoclonic/physiopathology , Female , Gene Knock-In Techniques , Gliosis , Haploinsufficiency , Hyperthermia/physiopathology , Immunohistochemistry , Male , Mice , NAV1.1 Voltage-Gated Sodium Channel/genetics , Neovascularization, Physiologic , Neuronal Plasticity , Nitric Oxide , Open Field Test , Rotarod Performance Test , Signal Transduction , Social Behavior , Synaptic Transmission , Tandem Mass Spectrometry , Vascular Endothelial Growth Factor Receptor-2/metabolism , ras-GRF1/metabolism
15.
J Cell Mol Med ; 25(11): 5015-5024, 2021 06.
Article in English | MEDLINE | ID: mdl-33991172

ABSTRACT

Cyclic AMP-dependent protein kinase A (PKA) and protein phosphatase 1 (PP1) are proteins involved in numerous essential signalling pathways that modulate physiological and pathological functions. Both PP1 and PKA can be inhibited by dopamine- and cAMP-regulated phosphoprotein 32 kD (DARPP-32). Using immunohistochemistry, PKA and PP1 expression was determined in a large primary breast tumour cohort to evaluate associations between clinical outcome and clinicopathological criteria (n > 1100). In addition, mRNA expression of PKA and PP1 subunits was assessed in the METABRIC data set (n = 1980). Low protein expression of PKA was significantly associated with adverse survival of breast cancer patients; interestingly, this relationship was stronger in ER-positive breast cancer patients. PP1 protein expression was not associated with patient survival. PKA and PP1 subunit mRNA was also assessed; PPP1CA, PRKACG and PRKAR1B were associated with breast cancer-specific survival. In patients with high expression of DARPP-32, low expression of PP1 was associated with adverse survival when compared to high expression in the same group. PKA expression and PP1 expression are of significant interest in cancer as they are involved in a wide array of cellular processes, and these data indicate PKA and PP1 may play an important role in patient outcome.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Expression Regulation, Neoplastic , Protein Phosphatase 1/metabolism , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Combined Modality Therapy , Cyclic AMP-Dependent Protein Kinases/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Female , Humans , Middle Aged , Phosphorylation , Prognosis , Protein Phosphatase 1/genetics , Retrospective Studies , Survival Rate
16.
Adv Pharmacol ; 90: 67-87, 2021.
Article in English | MEDLINE | ID: mdl-33706939

ABSTRACT

DARPP-32 (dopamine- and cAMP-regulated phosphoprotein with an apparent Mr of 32,000), now also known as phosphoprotein phosphatase 1 regulatory subunit 1B (PPP1R1B), is a potent inhibitor of protein phosphatase 1 (PP1, also known as PPP1) when phosphorylated at Thr34 by cAMP-dependent protein kinase (PKA). DARPP-32 exhibits a remarkable regional distribution in brain, roughly similar to that of dopamine innervation. Its discovery was a culmination of the long-standing effort of Paul Greengard to understand the mechanisms through which neurotransmitters such as dopamine exert their effects on target neurons. DARPP-32 is particularly enriched in striatal projection neurons where it is regulated by numerous signals through which it integrates and amplifies responses to many stimuli. Molecular studies of DARPP-32 have revealed that its regulation and function are more complex than anticipated. It is phosphorylated on multiple sites by several protein kinases that modulate DARPP-32 properties. Primarily, when phosphorylated at Thr34 DARPP-32 is a potent inhibitor of PP1, whereas when phosphorylated at Thr75 by Cdk5 it inhibits PKA. Phosphorylation at serine residues by CK1 and CK2 modulates its intracellular localization and its sensitivity to kinases or phosphatases. Modeling studies provide evidence that the signaling pathways including DARPP-32 are endowed of strong robustness and bistable properties favoring switch-like responses. Thus DARPP-32 combined with a set of other distinct signaling molecules enriched in striatal projection neurons plays a key role in the characteristic properties and physiological function of these neurons.


Subject(s)
Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Animals , Basal Ganglia/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/chemistry , Humans , Nervous System Diseases/metabolism , Neurotransmitter Agents/metabolism , Phosphorylation
17.
Adv Pharmacol ; 90: 39-65, 2021.
Article in English | MEDLINE | ID: mdl-33706938

ABSTRACT

Decades of research led by Paul Greengard identified protein phosphorylation as a ubiquitous and vital post-translational modification involved in many neuronal signaling pathways. In particular, his discovery that second messenger-regulated protein phosphorylation plays a central role in the propagation and transduction of signals in the nervous system has been essential in understanding the molecular mechanisms of neuronal communication. The establishment of dopamine (DA) as an essential neurotransmitter in the central nervous system, combined with observations that DA activates G-protein-coupled receptors to control the production of cyclic adenosine monophosphate (cAMP) in postsynaptic neurons, has provided fundamental insight into the regulation of neurotransmission. Notably, DA signaling in the striatum is involved in many neurological functions such as control of locomotion, reward, addiction, and learning, among others. This review focuses on the history, characterization, and function of cAMP-mediated regulation of serine/threonine protein phosphatases and their role in DA-mediated signaling in striatal neurons. Several small, heat- and acid-stable proteins, including DARPP-32, RCS, and ARPP-16/19, were discovered by the Greengard laboratory to be regulated by DA- and cAMP signaling, and found to undergo a complex but coordinated sequence of phosphorylation and dephosphorylation events. These studies have contributed significantly to the establishment of protein phosphorylation as a ubiquitous and vital process in signal propagation in neurons, paradigm shifting discoveries at the time. Understanding DA-mediated signaling in the context of signal propagation has led to numerous insights into human conditions and the development of treatments and therapies.


Subject(s)
Corpus Striatum/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphoproteins/metabolism , Protein Kinases/metabolism , Signal Transduction , Animals , Humans
18.
Adv Pharmacol ; 90: 89-115, 2021.
Article in English | MEDLINE | ID: mdl-33706940

ABSTRACT

Paul Greengard brought to neuroscience the idea of, and evidence for, the role of second messenger systems in neuronal signaling. The fundamental nature of his contributions is evident in the far reach of his work, relevant to various subfields and topics in neuroscience. In this review, we discuss some of Greengard's work from the perspective of nicotinic acetylcholine receptors and their relevance to nicotine addiction. Specifically, we review the roles of dopamine- and cAMP-regulated phospho-protein of 32kDa (DARPP-32) and Ca2+/calmodulin-dependent kinase II (CaMKII) in nicotine-dependent behaviors. For each protein, we discuss the historical context of their discovery and initial characterization, focusing on the extensive biochemical and immunohistochemical work conducted by Greengard and colleagues. We then briefly summarize contemporary understanding of each protein in key intracellular signaling cascades and evidence for the role of each protein with respect to systems and behaviors relevant to nicotine addiction.


Subject(s)
Behavior, Addictive/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Nicotine/pharmacology , Signal Transduction , Animals , Humans , Receptors, Nicotinic/metabolism , Signal Transduction/drug effects
19.
Cells ; 10(2)2021 01 26.
Article in English | MEDLINE | ID: mdl-33530478

ABSTRACT

Relief learning is the association of environmental cues with the cessation of aversive events. While there is increasing knowledge about the neural circuitry mediating relief learning, the respective molecular pathways are not known. Therefore, the aim of the present study was to examine different putative molecular pathways underlying relief learning. To this purpose, male rats were subjected either to relief conditioning or to a pseudo conditioning procedure. Forty-five minutes or 6 h after conditioning, samples of five different brain regions, namely the prefrontal cortex, nucleus accumbens (NAC), dorsal striatum, dorsal hippocampus, and amygdala, were collected. Using quantitative Western blots, the expression level of CREB, pCREB, ERK1/2, pERK1/2, CaMKIIα, MAP2K, PKA, pPKA, Akt, pAkt, DARPP-32, pDARPP-32, 14-3-3, and neuroligin2 were studied. Our analyses revealed that relief conditioned rats had higher CREB phosphorylation in NAC 6 h after conditioning than pseudo conditioned rats. The data further revealed that this CREB phosphorylation was mainly induced by dopamine D1 receptor-mediated activation of PKA, however, other kinases, downstream of the NMDA receptor, may also contribute. Taken together, the present study suggests that CREB phosphorylation, induced by a combination of different molecular pathways downstream of dopamine D1 and NMDA receptors, is essential for the acquisition and consolidation of relief learning.


Subject(s)
Conditioning, Classical , Cyclic AMP Response Element-Binding Protein/metabolism , Nucleus Accumbens/metabolism , 14-3-3 Proteins/metabolism , Animals , Behavior, Animal , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase 1/metabolism , Male , Nerve Tissue Proteins/metabolism , Organ Specificity , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Receptors, Dopamine D1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
20.
Alcohol Clin Exp Res ; 45(3): 608-619, 2021 03.
Article in English | MEDLINE | ID: mdl-33471948

ABSTRACT

BACKGROUND: Caffeine is frequently consumed with ethanol to reduce the impairing effects induced by ethanol, including psychomotor slowing or incoordination. Both drugs modulate dopamine (DA)-related markers in accumbens (Acb), and Acb DA is involved in voluntary locomotion and locomotor sensitization. The present study determined whether caffeine can affect locomotion induced by acute and repeated ethanol administration in adult male CD-1 mice. METHODS: Acute administration of caffeine (7.5 to 30.0 mg/kg) was evaluated for its effects on acute ethanol-induced (1.5 to 3.5 g/kg) changes in open-field horizontal locomotion, supported rearing, and rearing not supported by the wall. DA receptor-dependent phosphorylation markers were assessed: extracellular signal-regulated kinase (pERK), and dopamine-and cAMP-regulated phosphoprotein Mr32kDa phosphorylated at threonine 75 site (pDARPP-32-Thr75) in Acb core and shell. Acutely administered caffeine was also evaluated in ethanol-sensitized (1.5 g/kg) mice. RESULTS: Acute ethanol decreased both types of rearing. Caffeine increased supported rearing but did not block ethanol -induced decreases in rearing. Both substances increased horizontal locomotion in a biphasic manner, and caffeine potentiated ethanol-induced locomotion. Although ethanol administered repeatedly induced sensitization of locomotion and unsupported rearing, acute administration of caffeine to ethanol-sensitized mice in an ethanol-free state resulted in blunted stimulant effects compared with those seen in ethanol-naïve mice. Ethanol increased pERK immunoreactivity in both subregions of the Acb, but coadministration with caffeine blunted this increase. There were no effects on pDARPP-32(Thr75) immunoreactivity. CONCLUSIONS: The present results demonstrated that, after the first administration, caffeine potentiated the stimulating actions of ethanol, but did not counteract its suppressant or ataxic effects. Moreover, our results show that caffeine has less activating effects in ethanol-sensitized animals.


Subject(s)
Caffeine/administration & dosage , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Ethanol/administration & dosage , Locomotion/physiology , MAP Kinase Signaling System/physiology , Nucleus Accumbens/metabolism , Animals , Dose-Response Relationship, Drug , Ethanol/antagonists & inhibitors , Locomotion/drug effects , MAP Kinase Signaling System/drug effects , Male , Nucleus Accumbens/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL