Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 933
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38906412

RESUMEN

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.


Asunto(s)
Fosfoproteína 32 Regulada por Dopamina y AMPc , Dopamina , Neuronas GABAérgicas , Telencéfalo , Pez Cebra , Animales , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Neuronas GABAérgicas/metabolismo , Telencéfalo/metabolismo , Telencéfalo/embriología , Dopamina/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Animales Modificados Genéticamente , Regulación del Desarrollo de la Expresión Génica/fisiología
2.
Brain Res ; 1839: 149017, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-38768935

RESUMEN

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.


Asunto(s)
Manganeso , Ratas Wistar , Rotenona , Tirosina 3-Monooxigenasa , Animales , Rotenona/toxicidad , Masculino , Manganeso/toxicidad , Ratas , Tirosina 3-Monooxigenasa/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , alfa-Sinucleína/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Cuerpo Estriado/metabolismo , Cuerpo Estriado/efectos de los fármacos
3.
Sci Rep ; 13(1): 21163, 2023 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-38036593

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Neoplasias de la Mama/metabolismo , Moléculas de Adhesión Celular/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Fosforilación , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Transcriptoma
4.
Neoplasia ; 45: 100937, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37769528

RESUMEN

The therapeutic scenario of Human Epidermal Growth Factor Receptor 2 positive advanced breast cancer (ABC) has been recently enriched by a number of innovative agents, which are reshaping treatment sequence. While randomized trials have documented an advantage in terms of efficacy, for the newly available agents we lack effectiveness and tolerability evidence from the real-world setting. Similarly, the identification of predictive biomarkers might improve clinical decision. We herein describe the outline of a prospective/retrospective study which aims to explore the optimal sequence of treatment in HER2+, pertuzumab pre-treated ABC patients treated in II line with anti-HER2 agents in clinical practice. As part of the pre-clinical tasks envisioned by the STEP study, in vitro cell models of resistance were exploited to investigate molecular features associated with reduced efficacy of HER2 targeting agents at the transcript level. The aggressive behavior of resistant cell populations was measured by growth assessment in mouse models. This approach led to the identification of DARPP-32 and t-DARPP proteins as possible predictive biomarkers of efficacy of anti-HER2 agents. Biomarkers validation and the clinical goals will be reached through patients' inclusion into two independent cohorts, i.e., the prospective and retrospective cohorts, whose setup is currently ongoing.


Asunto(s)
Neoplasias de la Mama , Ratones , Animales , Humanos , Femenino , Trastuzumab/uso terapéutico , Estudios Retrospectivos , Estudios Prospectivos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc , Biomarcadores , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica
5.
J Comp Neurol ; 531(8): 888-920, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37002560

RESUMEN

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.


Asunto(s)
Núcleo Caudado , Cuerpo Estriado , Humanos , Cuerpo Estriado/metabolismo , Núcleo Caudado/metabolismo , Ganglios Basales , Neuronas/metabolismo , Receptores de Dopamina D2/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Neurópilo/metabolismo
6.
Neuropharmacology ; 228: 109462, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36792029

RESUMEN

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.


Asunto(s)
Dopamina , Fosfatos , Ratones , Animales , Humanos , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Dopamina/metabolismo , Fosfatos/metabolismo , Transmisión Sináptica , Fosforilación , Convulsiones/metabolismo , Receptores de Dopamina D1/metabolismo , Proteína Fosfatasa 1/metabolismo , Hipocampo
7.
Chem Biol Interact ; 372: 110366, 2023 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-36706892

RESUMEN

Recently, phosphodiesterases (PDEs) have gained great attention due to their implication in Parkinson's disease (PD) pathogenesis. Noteworthy, the PDE4 enzyme is highly expressed in the striatum and selectively degrades cyclic adenosine monophosphate (cAMP). The cAMP was shown to play a vital role in dopamine (DA) signaling besides maintaining the plasticity of dopaminergic neurons as well as protecting them from inflammation and oxidative stress-mediated death. Thus, PDE4 inhibition could be a promising strategy for treating PD. Accordingly, the present study investigated the neuroprotective efficacy of roflumilast, a PDE4 inhibitor, in abolishing neurodegeneration in the rotenone-induced PD model. Rotenone (1.5 mg/kg, s.c) was delivered via 11 injections on matching days. Roflumilast treatment (0.5 mg/kg, p.o) was given daily after the fifth rotenone injection. Roflumilast significantly reversed rotenone's adverse effects, as it enhanced trophic factors expression and abrogated inflammation as well as oxidative stress. Thus, promoting dopaminergic neuronal plasticity and survival, as well as restoring striatal DA level and function, which resulted in enhanced motor performance. The beneficial effect of roflumilast was mediated through inhibition of striatal PDE4 with consequent activation of cAMP-dependent protein kinase A (PKA) signaling pathways, including the cAMP response element-binding protein (CREB) pathway and dopamine and cAMP-regulated phosphoprotein 32,000 (DARPP-32) pathway that is essential for maintaining dopaminergic function. Therefore, the present work sheds light on the substantial neuroprotective potential of roflumilast in treating PD through the activation of the cAMP/PKA cascade.


Asunto(s)
Enfermedad de Parkinson , Ratas , Animales , Fosfoproteína 32 Regulada por Dopamina y AMPc/farmacología , Rotenona/toxicidad , Dopamina/metabolismo , Transducción de Señal , Fosfoproteínas
8.
Neurochem Int ; 162: 105438, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36351540

RESUMEN

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.


Asunto(s)
Cuerpo Estriado , Neostriado , Animales , Ratones , Proteína Fosfatasa 1/metabolismo , Proteína Fosfatasa 1/farmacología , Cuerpo Estriado/metabolismo , Neostriado/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Transducción de Señal , Fosforilación , Receptores de Dopamina D1/metabolismo
9.
PeerJ ; 10: e14516, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36540795

RESUMEN

Dynamic modelling has considerably improved our understanding of complex molecular mechanisms. Ordinary differential equations (ODEs) are the most detailed and popular approach to modelling the dynamics of molecular systems. However, their application in signalling networks, characterised by multi-state molecular complexes, can be prohibitive. Contemporary modelling methods, such as rule- based (RB) modelling, have addressed these issues. The advantages of RB modelling over ODEs have been presented and discussed in numerous reviews. In this study, we conduct a direct comparison of the time courses of a molecular system founded on the same reaction network but encoded in the two frameworks. To make such a comparison, a set of reactions that underlie an ODE model was manually encoded in the Kappa language, one of the RB implementations. A comparison of the models was performed at the level of model specification and dynamics, acquired through model simulations. In line with previous reports, we confirm that the Kappa model recapitulates the general dynamics of its ODE counterpart with minor differences. These occur when molecules have multiple sites binding the same interactor. Furthermore, activation of these molecules in the RB model is slower than in the ODE one. As reported for other molecular systems, we find that, also for the DARPP-32 reaction network, the RB representation offers a more expressive and flexible syntax that facilitates access to fine details of the model, easing model reuse. In parallel with these analyses, we report a refactored model of the DARPP-32 interaction network that can serve as a canvas for the development of more complex dynamic models to study this important molecular system.


Asunto(s)
Transducción de Señal , Fosfoproteína 32 Regulada por Dopamina y AMPc
10.
Int J Mol Sci ; 23(19)2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36232936

RESUMEN

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.


Asunto(s)
Antipsicóticos , Haloperidol , Adenosina/metabolismo , Animales , Antipsicóticos/metabolismo , Antipsicóticos/farmacología , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Genes Inmediatos-Precoces , Glutamatos/metabolismo , Haloperidol/farmacología , Ratones , Ratones Transgénicos , N-Metilaspartato/metabolismo , Neuronas/metabolismo , Receptores de Dopamina D1/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
11.
Neuropharmacology ; 219: 109238, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36055413

RESUMEN

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.


Asunto(s)
Ácido Kaínico , Serina , Animales , Quinasa de la Caseína II/metabolismo , Dopamina/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Hipocampo/metabolismo , Ácido Kaínico/farmacología , Ratones , Fosfatos/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Proteína Fosfatasa 1/metabolismo , Piridoxal , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Serina/metabolismo , Serina/farmacología
12.
Proc Natl Acad Sci U S A ; 119(33): e2207489119, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35939707

RESUMEN

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.


Asunto(s)
Fosfoproteína 32 Regulada por Dopamina y AMPc , Degeneración Macular , Diana Mecanicista del Complejo 1 de la Rapamicina , Epitelio Pigmentado de la Retina , Animales , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Activación Enzimática , Humanos , Degeneración Macular/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal
13.
Cells ; 11(10)2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35626701

RESUMEN

Huntington's disease (HD) is a neurodegenerative inherited genetic disorder, which leads to the onset of motor, neuropsychiatric and cognitive disturbances. HD is characterized by the loss of gamma-aminobutyric acid (GABA)ergic medium spiny neurons (MSNs). To date, there is no treatment for HD. Mesenchymal stem cells (MSCs) provide a substantial therapeutic opportunity for the HD treatment. Herein, we investigated the therapeutic potential of human immature dental pulp stem cells (hIDPSC), a special type of MSC originated from the neural crest, for HD treatment. Two different doses of hIDPSC were intravenously administrated in a subacute 3-nitropropionic acid (3NP)-induced rat model. We demonstrated hIDPSC homing in the striatum, cortex and subventricular zone using specific markers for human cells. Thirty days after hIDPSC administration, the cells found in the brain are still express hallmarks of undifferentiated MSC. Immunohistochemistry quantities analysis revealed a significant increase in the number of BDNF, DARPP32 and D2R positive stained cells in the striatum and cortex in the groups that received hIDPSC. The differences were more expressive in animals that received only one administration of hIDPSC. Altogether, these data suggest that the intravenous administration of hIDPSCs can restore the BDNF, DARPP32 and D2R expression, promoting neuroprotection and neurogenesis.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Fosfoproteína 32 Regulada por Dopamina y AMPc , Enfermedad de Huntington , Trasplante de Células Madre , Células Madre , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Pulpa Dental/citología , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Infusiones Intravenosas , Ratas , Células Madre/citología
14.
Oncogene ; 41(1): 83-98, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34675407

RESUMEN

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.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Terapia Molecular Dirigida/métodos , Receptor ErbB-3/metabolismo , Adenocarcinoma del Pulmón/patología , Animales , Línea Celular Tumoral , Humanos , Ratones
15.
J Comp Neurol ; 530(5): 792-803, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34545948

RESUMEN

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.


Asunto(s)
Encéfalo/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Centro Vocal Superior/metabolismo , Neuronas/metabolismo , Vocalización Animal/fisiología , Factores de Edad , Animales , Pinzones
16.
Mov Disord ; 37(2): 365-374, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34820905

RESUMEN

BACKGROUND: The dystonias are a heterogeneous group of hyperkinetic disorders characterized by sustained or intermittent muscle contractions that cause abnormal movements and/or postures. Although more than 200 causal genes are known, many cases of primary dystonia have no clear genetic cause. OBJECTIVES: To identify the causal gene in a consanguineous family with three siblings affected by a complex persistent generalized dystonia, generalized epilepsy, and mild intellectual disability. METHODS: We performed exome sequencing in the parents and two affected siblings and characterized the expression of the identified gene by immunohistochemistry in control human and zebrafish brains. RESULTS: We identified a novel missense variant (c.142G>A (NM_032192); p.Glu48Lys) in the protein phosphatase 1 regulatory inhibitor subunit 1B gene (PPP1R1B) that was homozygous in all three siblings and heterozygous in the parents. This gene is also known as dopamine and cAMP-regulated neuronal phosphoprotein 32 (DARPP-32) and has been involved in the pathophysiology of abnormal movements. The uncovered variant is absent in public databases and modifies the conserved glutamate 48 localized close to the serine 45 phosphorylation site. The PPP1R1B protein was shown to be expressed in cells and regions involved in movement control, including projection neurons of the caudate-putamen, substantia nigra neuropil, and cerebellar Purkinje cells. The latter cells were also confirmed to be positive for PPP1R1B expression in the zebrafish brain. CONCLUSIONS: We report the association of a PPP1R1B/DARPP-32 variant with generalized dystonia in man. It might be relevant to include the sequencing of this new gene in the diagnosis of patients with otherwise unexplained movement disorders. © 2021 International Parkinson and Movement Disorder Society.


Asunto(s)
Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Distonía , Trastornos Distónicos , Animales , Trastornos Distónicos/genética , Homocigoto , Humanos , Pez Cebra
17.
PLoS One ; 16(10): e0258486, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34648564

RESUMEN

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.


Asunto(s)
Proteínas de Unión al GTP/genética , Enfermedad de Huntington/patología , Animales , Fenómenos Biomecánicos , Peso Corporal , Encéfalo/fisiología , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Femenino , Proteínas de Unión al GTP/deficiencia , Proteínas de Unión al GTP/metabolismo , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
18.
Stem Cell Res ; 55: 102486, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34385043

RESUMEN

Human induced pluripotent stem cells (hiPSCs) are powerful tools for modeling human brain development and treating neurodegenerative diseases. Here we established a robust protocol with high scalability for generating striatal medium spiny neurons (MSNs) from hiPSCs using small molecules under two- and three-dimensional culture conditions. Using this protocol, GSH2+ lateral ganglionic eminence (LGE) progenitors were generated in two-dimensional culture by Sonic hedgehog signaling activation using purmorphamine, WNT signaling inhibition using XAV939, and dual-SMAD inhibition using LDN193189 and A83-01. Quantitative PCR analysis revealed sequential expression of LGE and striatal genes during differentiation. These LGE progenitors subsequently gave rise to DARPP32+ MSNs exhibiting spontaneous and evoked monophasic spiking activity. Applying this protocol in three-dimensional culture, we generated striatal neurospheres with gene expression profiles and cell layer organization resembling that of the developing striatum, including distinct ventricular and subventricular zones and DARPP32+ neurons at the surface. This protocol provides a useful experimental model for studying striatal development and yields cells potentially applicable for regenerative medicine to treat striatum-related disorders such as Huntington's disease.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular , Cuerpo Estriado/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Proteínas Hedgehog/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo
19.
Gene ; 799: 145808, 2021 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-34224831

RESUMEN

We set out to uncover transcriptome and chromatin landscape changes that occur in HER2 + breast cancer (BC) cells upon acquiring resistance to trastuzumab. RNA-seq analysis was applied to two independently-derived BC cell lines with acquired resistance to trastuzumab (SKBr3.HerR and BT-474HerR) and their parental drug-sensitive cell lines (SKBr3 and BT-474). Chromatin landscape analysis indicated that the most significant increase in accessibility in resistant cells occurs in PPP1R1B within a segment spanning introns 1b through intron 3. Footprint analysis of this segment revealed that FoxJ3 (within intron 2) and Pou5A1/Sox2 (within inton 3) transcription factor motifs are protected in resistant cells. Overall, 344 shared genes were upregulated in both resistant cell lines relative to their parental counterparts and 453 shared genes were downregulated in both resistant cell lines relative to their parental counterparts. In resistant cells, genes associated with autophagy and mitochondria organization are upregulated and genes associated with ribosome assembly and cell cycle are downregulated relative to parental cells. The five top upregulated genes in drug-resistant breast cancer cells are APOD, AZGP1, ETV5, ALPP, and PPP1R1B. This is the first report of increased chromatin accessibility within PPP1R1B associated with its t-Darpp transcript increase, and points to a possible mechanism for its activation in trastuzumab-resistant cells.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Cromatina/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Trastuzumab/farmacología , Antineoplásicos Inmunológicos/farmacología , Autofagia/efectos de los fármacos , Autofagia/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cromatina/genética , Fosfoproteína 32 Regulada por Dopamina y AMPc/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Humanos , Receptor ErbB-2/metabolismo , Factores de Transcripción SOXB1/genética
20.
Cell Prolif ; 54(8): e13082, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34152047

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Enfermedad de Huntington/patología , Células-Madre Neurales/trasplante , Animales , Astrocitos/citología , Astrocitos/metabolismo , Conducta Animal , Línea Celular , Cuerpo Calloso/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...