Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Biol Res ; 57(1): 40, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890753

ABSTRACT

BACKGROUND: The brain cortex is responsible for many higher-level cognitive functions. Disruptions during cortical development have long-lasting consequences on brain function and are associated with the etiology of brain disorders. We previously found that the protein tyrosine phosphatase receptor delta Ptprd, which is genetically associated with several human neurodevelopmental disorders, is essential to cortical brain development. Loss of Ptprd expression induced an aberrant increase of excitatory neurons in embryonic and neonatal mice by hyper-activating the pro-neurogenic receptors TrkB and PDGFRß in neural precursor cells. However, whether these alterations have long-lasting consequences in adulthood remains unknown. RESULTS: Here, we found that in Ptprd+/- or Ptprd-/- mice, the developmental increase of excitatory neurons persists through adulthood, affecting excitatory synaptic function in the medial prefrontal cortex. Likewise, heterozygosity or homozygosity for Ptprd also induced an increase of inhibitory cortical GABAergic neurons and impaired inhibitory synaptic transmission. Lastly, Ptprd+/- or Ptprd-/- mice displayed autistic-like behaviors and no learning and memory impairments or anxiety. CONCLUSIONS: These results indicate that loss of Ptprd has long-lasting effects on cortical neuron number and synaptic function that may aberrantly impact ASD-like behaviors.


Subject(s)
Autistic Disorder , Neurons , Receptor-Like Protein Tyrosine Phosphatases, Class 2 , Animals , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Mice , Autistic Disorder/genetics , Autistic Disorder/physiopathology , Disease Models, Animal , Male , Cerebral Cortex/metabolism , Mice, Knockout , Synaptic Transmission/physiology , Mice, Inbred C57BL , Female
2.
J Hepatol ; 64(2): 361-369, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26453970

ABSTRACT

BACKGROUND & AIMS: Niemann-Pick C2 (NPC2) is a lysosomal protein involved in the egress of low-density lipoprotein-derived cholesterol from lysosomes to other intracellular compartments. NPC2 has been detected in several tissues and is also secreted from the liver into bile. We have previously shown that NPC2-deficient mice fed a lithogenic diet showed reduced biliary cholesterol secretion as well as cholesterol crystal and gallstone formation. This study aimed to investigate the consequences of NPC2 hepatic overexpression on liver cholesterol metabolism, biliary lipid secretion, gallstone formation and the effect of NPC2 on cholesterol crystallization in model bile. METHODS: We generated NPC2 transgenic mice (Npc2.Tg) and fed them either chow or lithogenic diets. We studied liver cholesterol metabolism, biliary lipid secretion, bile acid composition and gallstone formation. We performed cholesterol crystallization studies in model bile using a recombinant NPC2 protein. RESULTS: No differences were observed in biliary cholesterol content or secretion between wild-type and Npc2.Tg mice fed the chow or lithogenic diets. Interestingly, Npc2.Tg mice showed an increased susceptibility to the lithogenic diet, developing more cholesterol gallstones at early times, but did not show differences in the bile acid hydrophobicity and gallbladder cholesterol saturation indices compared to wild-type mice. Finally, recombinant NPC2 decreased nucleation time in model bile. CONCLUSIONS: These results suggest that NPC2 promotes cholesterol gallstone formation by decreasing the cholesterol nucleation time, indicating a pro-nucleating function of NPC2 in bile.


Subject(s)
Cholesterol , Gallstones/metabolism , Vesicular Transport Proteins/metabolism , Animals , Bile/metabolism , Bile Acids and Salts/metabolism , Cholesterol/chemistry , Cholesterol/metabolism , Crystallization , Disease Models, Animal , Liver/metabolism , Mice , Mice, Transgenic , Models, Chemical , Time Factors
3.
J Neurosci ; 34(2): 539-53, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24403153

ABSTRACT

The three-layered piriform cortex, an integral part of the olfactory system, processes odor information relayed by olfactory bulb mitral cells. Specifically, mitral cell axons form the lateral olfactory tract (LOT) by targeting lateral olfactory tract (lot) guidepost cells in the piriform cortex. While lot cells and other piriform cortical neurons share a pallial origin, the factors that specify their precise phenotypes are poorly understood. Here we show that in mouse, the proneural genes Neurog1 and Neurog2 are coexpressed in the ventral pallium, a progenitor pool that first gives rise to Cajal-Retzius (CR) cells, which populate layer I of all cortical domains, and later to layer II/III neurons of the piriform cortex. Using loss-of-function and gain-of-function approaches, we find that Neurog1 has a unique early role in reducing CR cell neurogenesis by tempering Neurog2's proneural activity. In addition, Neurog1 and Neurog2 have redundant functions in the ventral pallium, acting in two phases to first specify a CR cell fate and later to specify layer II/III piriform cortex neuronal identities. In the early phase, Neurog1 and Neurog2 are also required for lot cell differentiation, which we reveal are a subset of CR neurons, the loss of which prevents mitral cell axon innervation and LOT formation. Consequently, mutation of Trp73, a CR-specific cortical gene, results in lot cell and LOT axon displacement. Neurog1 and Neurog2 thus have unique and redundant functions in the piriform cortex, controlling the timing of differentiation of early-born CR/lot cells and specifying the identities of later-born layer II/III neurons.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cerebral Cortex/embryology , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurons/cytology , Animals , Cell Differentiation/physiology , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Electroporation , Embryo, Mammalian , Female , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Mutant Strains , Neural Stem Cells/metabolism
4.
J Neurosci ; 33(31): 12569-85, 2013 Jul 31.
Article in English | MEDLINE | ID: mdl-23904595

ABSTRACT

The molecular mechanisms that regulate adult neural precursor cell (NPC) survival, and thus maintain adult neurogenesis, are not well defined. Here, we investigate the role of p63, a p53 family member, in adult NPC function in mice. Conditional ablation of p63 in adult NPCs or p63 haploinsufficiency led to reduced numbers of NPCs and newborn neurons in the neurogenic zones of the hippocampus and lateral ventricles and in the olfactory bulb. These reductions were attributable to enhanced apoptosis of NPCs and newborn neurons and were rescued by inhibition of caspase activity, p53, or the p53 apoptotic effector PUMA (p53-upregulated modulator of apoptosis). Moreover, these cellular deficits were functionally important because they led to perturbations in hippocampus-dependent memory formation. These results indicate that p63 regulates the numbers of adult NPCs and adult-born neurons as well as neural stem cell-dependent cognitive functions, and that it does so, at least in part, by inhibiting p53-dependent cell death.


Subject(s)
Adult Stem Cells/physiology , Exploratory Behavior/physiology , Hippocampus/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Phosphoproteins/metabolism , Trans-Activators/metabolism , Animals , Bromodeoxyuridine/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Cerebral Ventricles/cytology , Conditioning, Psychological/physiology , Cues , Exploratory Behavior/drug effects , Fear/psychology , Intermediate Filament Proteins/genetics , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nestin , Neurogenesis/drug effects , Neurogenesis/genetics , Phosphoproteins/genetics , Proteins/genetics , RNA, Untranslated , Tamoxifen/pharmacology , Trans-Activators/genetics , Transcriptional Activation/drug effects , Tumor Suppressor Protein p53/genetics
5.
Front Cell Dev Biol ; 12: 1357862, 2024.
Article in English | MEDLINE | ID: mdl-38487272

ABSTRACT

Neurodevelopmental disorders are characterized by alterations in the development of the cerebral cortex, including aberrant changes in the number and function of neural cells. Although neurogenesis is one of the most studied cellular processes in these pathologies, little evidence is known about glial development. Genetic association studies have identified several genes associated with neurodevelopmental disorders. Indeed, variations in the PTPRD gene have been associated with numerous brain disorders, including autism spectrum disorder, restless leg syndrome, and schizophrenia. We previously demonstrated that constitutive loss of PTPRD expression induces significant alterations in cortical neurogenesis, promoting an increase in intermediate progenitors and neurons in mice. However, its role in gliogenesis has not been evaluated. To assess this, we developed a conditional knockout mouse model lacking PTPRD expression in telencephalon cells. Here, we found that the lack of PTPRD in the mouse cortex reduces glial precursors, astrocytes, and oligodendrocytes. According to our results, this decrease in gliogenesis resulted from a reduced number of radial glia cells at gliogenesis onset and a lower gliogenic potential in cortical neural precursors due to less activation of the JAK/STAT pathway and reduced expression of gliogenic genes. Our study shows PTPRD as a regulator of the glial/neuronal balance during cortical neurodevelopment and highlights the importance of studying glial development to understand the etiology of neurodevelopmental diseases.

6.
Cell Rep ; 43(5): 114144, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38656874

ABSTRACT

The molecular mechanisms underlying seizure generation remain elusive, yet they are crucial for developing effective treatments for epilepsy. The current study shows that inhibiting c-Abl tyrosine kinase prevents apoptosis, reduces dendritic spine loss, and maintains N-methyl-d-aspartate (NMDA) receptor subunit 2B (NR2B) phosphorylated in in vitro models of excitotoxicity. Pilocarpine-induced status epilepticus (SE) in mice promotes c-Abl phosphorylation, and disrupting c-Abl activity leads to fewer seizures, increases latency toward SE, and improved animal survival. Currently, clinically used c-Abl inhibitors are non-selective and have poor brain penetration. The allosteric c-Abl inhibitor, neurotinib, used here has favorable potency, selectivity, pharmacokinetics, and vastly improved brain penetration. Neurotinib-administered mice have fewer seizures and improved survival following pilocarpine-SE induction. Our findings reveal c-Abl kinase activation as a key factor in ictogenesis and highlight the impact of its inhibition in preventing the insurgence of epileptic-like seizures in rodents and humans.


Subject(s)
Pilocarpine , Proto-Oncogene Proteins c-abl , Seizures , Animals , Male , Mice , Apoptosis/drug effects , Mice, Inbred C57BL , Neurons/drug effects , Neurons/pathology , Neurons/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Seizures/chemically induced , Seizures/drug therapy , Seizures/pathology , Status Epilepticus/chemically induced , Status Epilepticus/drug therapy , Status Epilepticus/pathology
7.
Antioxidants (Basel) ; 12(11)2023 Nov 16.
Article in English | MEDLINE | ID: mdl-38001860

ABSTRACT

The endoplasmic reticulum is a subcellular organelle key in the control of synthesis, folding, and sorting of proteins. Under endoplasmic reticulum stress, an adaptative unfolded protein response is activated; however, if this activation is prolonged, cells can undergo cell death, in part due to oxidative stress and mitochondrial fragmentation. Here, we report that endoplasmic reticulum stress activates c-Abl tyrosine kinase, inducing its translocation to mitochondria. We found that endoplasmic reticulum stress-activated c-Abl interacts with and phosphorylates the mitochondrial fusion protein MFN2, resulting in mitochondrial fragmentation and apoptosis. Moreover, the pharmacological or genetic inhibition of c-Abl prevents MFN2 phosphorylation, mitochondrial fragmentation, and apoptosis in cells under endoplasmic reticulum stress. Finally, in the amyotrophic lateral sclerosis mouse model, where endoplasmic reticulum and oxidative stress has been linked to neuronal cell death, we demonstrated that the administration of c-Abl inhibitor neurotinib delays the onset of symptoms. Our results uncovered a function of c-Abl in the crosstalk between endoplasmic reticulum stress and mitochondrial dynamics via MFN2 phosphorylation.

8.
Front Aging Neurosci ; 15: 1180987, 2023.
Article in English | MEDLINE | ID: mdl-37358955

ABSTRACT

Background: Growing evidence suggests that the non-receptor tyrosine kinase, c-Abl, plays a significant role in the pathogenesis of Alzheimer's disease (AD). Here, we analyzed the effect of c-Abl on the cognitive performance decline of APPSwe/PSEN1ΔE9 (APP/PS1) mouse model for AD. Methods: We used the conditional genetic ablation of c-Abl in the brain (c-Abl-KO) and pharmacological treatment with neurotinib, a novel allosteric c-Abl inhibitor with high brain penetrance, imbued in rodent's chow. Results: We found that APP/PS1/c-Abl-KO mice and APP/PS1 neurotinib-fed mice had improved performance in hippocampus-dependent tasks. In the object location and Barnes-maze tests, they recognized the displaced object and learned the location of the escape hole faster than APP/PS1 mice. Also, APP/PS1 neurotinib-fed mice required fewer trials to reach the learning criterion in the memory flexibility test. Accordingly, c-Abl absence and inhibition caused fewer amyloid plaques, reduced astrogliosis, and preserved neurons in the hippocampus. Discussion: Our results further validate c-Abl as a target for AD, and the neurotinib, a novel c-Abl inhibitor, as a suitable preclinical candidate for AD therapies.

9.
Front Immunol ; 13: 816619, 2022.
Article in English | MEDLINE | ID: mdl-35464419

ABSTRACT

Infections during pregnancy can seriously damage fetal neurodevelopment by aberrantly activating the maternal immune system, directly impacting fetal neural cells. Increasing evidence suggests that these adverse impacts involve alterations in neural stem cell biology with long-term consequences for offspring, including neurodevelopmental disorders such as autism spectrum disorder, schizophrenia, and cognitive impairment. Here we review how maternal infection with viruses such as Influenza A, Cytomegalovirus, and Zika during pregnancy can affect the brain development of offspring by promoting the release of maternal pro-inflammatory cytokines, triggering neuroinflammation of the fetal brain, and/or directly infecting fetal neural cells. In addition, we review insights into how these infections impact human brain development from studies with animal models and brain organoids. Finally, we discuss how maternal infection with SARS-CoV-2 may have consequences for neurodevelopment of the offspring.


Subject(s)
Autism Spectrum Disorder , COVID-19 , Virus Diseases , Zika Virus Infection , Zika Virus , Animals , Autism Spectrum Disorder/etiology , Brain , Cytokines , Female , Pregnancy , SARS-CoV-2 , Virus Diseases/complications
10.
IBRO Neurosci Rep ; 13: 378-387, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36590096

ABSTRACT

Parkinson's disease is the second most common neurodegenerative disorder. Although it is clear that dopaminergic neurons degenerate, the underlying molecular mechanisms are still unknown, and thus, successful treatment is still elusive. One pro-apoptotic pathway associated with several neurodegenerative diseases is the tyrosine kinase c-Abl and its target p73. Here, we evaluated the contribution of c-Abl and p73 in the degeneration of dopaminergic neurons induced by the neurotoxin 6-hydroxydopamine as a model for Parkinson's disease. First, we found that in SH-SY5Y cells treated with 6-hydroxydopamine, c-Abl and p73 phosphorylation levels were up-regulated. Also, we found that the pro-apoptotic p73 isoform TAp73 was up-regulated. Then, to evaluate whether c-Abl tyrosine kinase activity is necessary for 6-hydroxydopamine-induced apoptosis, we co-treated SH-SY5Y cells with 6-hydroxydopamine and Imatinib, a c-Abl specific inhibitor, observing that Imatinib prevented p73 phosphorylation, TAp73 up-regulation, and protected SH-SY5Y cells treated with 6-hydroxydopamine from apoptosis. Interestingly, this observation was confirmed in the c-Abl conditional null mice, where 6-hydroxydopamine stereotaxic injections induced a lesser reduction of dopaminergic neurons than in the wild-type mice significantly. Finally, we found that the intraperitoneal administration of Imatinib prevented the death of dopaminergic neurons induced by injecting 6-hydroxydopamine stereotaxically in the mice striatum. Thus, our findings support the idea that the c-Abl/p73 pathway is involved in 6-hydroxydopamine degeneration and suggest that inhibition of its kinase activity might be used as a therapeutical drug in Parkinson's disease.

11.
Neurodegener Dis ; 8(3): 124-8, 2011.
Article in English | MEDLINE | ID: mdl-20714112

ABSTRACT

BACKGROUND: Niemann-Pick type C (NPC) disease is a fatal lysosomal storage disease related to progressive neurodegeneration secondary to abnormal intracellular accumulation of cholesterol. Signs of endoplasmic reticulum (ER) stress have been reported in other lipidoses. Adaptation to ER stress is mediated by the unfolded protein response (UPR), an integrated signal transduction pathway that attenuates stress or triggers apoptosis of irreversibly damaged cells. OBJECTIVE: To investigate the possible engagement of ER stress responses in NPC models. METHODS: We used NPC1 deficient mice and an NPC cell-based model by knocking down the expression of NPC1 to measure several UPR markers through different approaches. RESULTS: Despite expectations that the UPR will be activated in NPC, our results indicate a lack of ER stress reactions in the cerebellum of symptomatic mice. Similarly, knocking down NPC1 in Neuro2a cells leads to clear cholesterol accumulation without evidence of UPR activation. CONCLUSION: Our results suggest that cholesterol overload and neuronal dysfunction in NPC is not associated with ER stress, which contrasts with recent reports suggesting the activation of the UPR in other lysosomal storage diseases.


Subject(s)
Endoplasmic Reticulum/physiology , Niemann-Pick Disease, Type C/physiopathology , Proteins/physiology , Unfolded Protein Response/physiology , Animals , Apoptosis/physiology , Cerebellum/metabolism , Cerebellum/pathology , Cholesterol/metabolism , Disease Models, Animal , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/metabolism , Proteins/genetics , Signal Transduction/physiology
12.
Front Cell Dev Biol ; 9: 659951, 2021.
Article in English | MEDLINE | ID: mdl-34966732

ABSTRACT

Protein phosphatases are major regulators of signal transduction and they are involved in key cellular mechanisms such as proliferation, differentiation, and cell survival. Here we focus on one class of protein phosphatases, the type IIA Receptor-type Protein Tyrosine Phosphatases (RPTPs), or LAR-RPTP subfamily. In the last decade, LAR-RPTPs have been demonstrated to have great importance in neurobiology, from neurodevelopment to brain disorders. In vertebrates, the LAR-RPTP subfamily is composed of three members: PTPRF (LAR), PTPRD (PTPδ) and PTPRS (PTPσ), and all participate in several brain functions. In this review we describe the structure and proteolytic processing of the LAR-RPTP subfamily, their alternative splicing and enzymatic regulation. Also, we review the role of the LAR-RPTP subfamily in neural function such as dendrite and axon growth and guidance, synapse formation and differentiation, their participation in synaptic activity, and in brain development, discussing controversial findings and commenting on the most recent studies in the field. Finally, we discuss the clinical outcomes of LAR-RPTP mutations, which are associated with several brain disorders.

13.
Cell Rep ; 30(1): 215-228.e5, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31914388

ABSTRACT

PTPRD is a receptor protein tyrosine phosphatase that is genetically associated with neurodevelopmental disorders. Here, we asked whether Ptprd mutations cause aberrant neural development by perturbing neurogenesis in the murine cortex. We show that loss of Ptprd causes increases in neurogenic transit-amplifying intermediate progenitor cells and cortical neurons and perturbations in neuronal localization. These effects are intrinsic to neural precursor cells since acute Ptprd knockdown causes similar perturbations. PTPRD mediates these effects by dephosphorylating receptor tyrosine kinases, including TrkB and PDGFRß, and loss of Ptprd causes the hyperactivation of TrkB and PDGFRß and their downstream MEK-ERK signaling pathway in neural precursor cells. Moreover, inhibition of aberrant TrkB or MEK activation rescues the increased neurogenesis caused by knockdown or homozygous loss of Ptprd. These results suggest that PTPRD regulates receptor tyrosine kinases to ensure appropriate numbers of intermediate progenitor cells and neurons, suggesting a mechanism for its genetic association with neurodevelopmental disorders.


Subject(s)
Neurogenesis , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Alleles , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cerebral Cortex/embryology , Embryo, Mammalian/cytology , Gene Knockdown Techniques , HEK293 Cells , Humans , Matrix Attachment Region Binding Proteins/metabolism , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation , Receptor-Like Protein Tyrosine Phosphatases, Class 2/deficiency , Signal Transduction , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism
14.
FASEB J ; 22(10): 3617-27, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18591368

ABSTRACT

Niemann-Pick type C (NPC) disease is a fatal autosomal recessive disorder characterized by the accumulation of free cholesterol and glycosphingolipids in the endosomal-lysosomal system. Patients with NPC disease have markedly progressive neuronal loss, mainly of cerebellar Purkinje neurons. There is strong evidence indicating that cholesterol accumulation and trafficking defects activate apoptosis in NPC brains. The purpose of this study was to analyze the relevance of apoptosis and particularly the proapoptotic c-Abl/p73 system in cerebellar neuron degeneration in NPC disease. We used the NPC1 mouse model to evaluate c-Abl/p73 expression and activation in the cerebellum and the effect of therapy with the c-Abl-specific inhibitor imatinib. The proapoptotic c-Abl/p73 system and the p73 target genes are expressed in the cerebellums of NPC mice. Furthermore, inhibition of c-Abl with imatinib preserved Purkinje neurons and reduced general cell apoptosis in the cerebellum, improved neurological symptoms, and increased the survival of NPC mice. Moreover, this prosurvival effect correlated with reduced mRNA levels of p73 proapoptotic target genes. Our results suggest that the c-Abl/p73 pathway is involved in NPC neurodegeneration and show that treatment with c-Abl inhibitors is useful in delaying progressive neurodegeneration, supporting the use of imatinib for clinical treatment of patients with NPC disease.


Subject(s)
Apoptosis/drug effects , Cerebellar Cortex/drug effects , Niemann-Pick Disease, Type C/drug therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Animals , Benzamides , Cell Survival/drug effects , Cerebellar Cortex/metabolism , Cerebellar Cortex/pathology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression/drug effects , Imatinib Mesylate , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Motor Activity/drug effects , Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Nuclear Proteins/metabolism , Proteins/genetics , Proto-Oncogene Proteins c-abl/metabolism , Purkinje Cells/drug effects , Purkinje Cells/pathology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Tumor Protein p73 , Tumor Suppressor Proteins/metabolism
15.
Brain ; 131(Pt 9): 2425-42, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18559370

ABSTRACT

There is evidence that amyloid beta-protein (Abeta) deposits or Abeta intermediates trigger pathogenic factors in Alzheimer's disease patients. We have previously reported that c-Abl kinase activation involved in cell signalling regulates the neuronal death response to Abeta fibrils (Abeta(f)). In the present study we investigated the therapeutic potential of the selective c-Abl inhibitor STI571 on both the intrahippocampal injection of Abeta(f) and APPsw/PSEN1DeltaE9 transgenic mice Alzheimer's disease models. Injection of Abeta(f) induced an increase in the numbers of p73 and c-Abl immunoreactive cells in the hippocampal area near to the lesion. Chronic intraperitoneal administration of STI571 reduced the rat behavioural deficit induced by Abeta(f), as well as apoptosis and tau phosphorylation. Our in vitro studies suggest that inhibition of the c-Abl/p73 signalling pathway is the mechanism underlying of the effects of STI571 on Abeta-induced apoptosis for the following reasons: (i) Abeta(f) induces p73 phosphorylation, the TAp73 isoform levels increase so as to enhance its proapoptotic function, and all these effects where reduced by STI571; (ii) c-Abl kinase activity is required for neuronal apoptosis and (iii) STI571 prevents the Abeta-induced increase in the expression of apoptotic genes. Furthermore, in the Abeta-injected area there was a huge increase in phosphorylated p73 and a larger number of TAp73-positive cells, with these changes being prevented by STI571 coinjection. Moreover, the intraperitoneal administration of STI571 rescued the cognitive decline in APPsw/PSEN1DeltaE9 mice, p73 phosphorylation, tau phosphorylation and caspase-3 activation in neurons around Abeta deposits. Besides, we observed a decrease in the number and size of Abeta deposits in the APPsw/PSEN1DeltaE9-STI571-treated mice. These results are consistent with the role of the c-Abl/p73 signalling pathway in Abeta neurodegeneration, and suggest that STI571-like compounds would be effective in therapeutic treatments of Alzheimer disease.


Subject(s)
Alzheimer Disease/prevention & control , Apoptosis/drug effects , Pyrimidines/therapeutic use , tau Proteins/metabolism , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Peptides , Animals , Benzamides , Cells, Cultured , DNA-Binding Proteins/metabolism , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Imatinib Mesylate , Male , Maze Learning/drug effects , Mice , Mice, Transgenic , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Piperazines , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Protein p73 , Tumor Suppressor Proteins/metabolism
16.
Redox Biol ; 12: 274-284, 2017 08.
Article in English | MEDLINE | ID: mdl-28282615

ABSTRACT

MLN64 is a late endosomal cholesterol-binding membrane protein that has been implicated in cholesterol transport from endosomal membranes to the plasma membrane and/or mitochondria, in toxin-induced resistance, and in mitochondrial dysfunction. Down-regulation of MLN64 in Niemann-Pick C1 deficient cells decreased mitochondrial cholesterol content, suggesting that MLN64 functions independently of NPC1. However, the role of MLN64 in the maintenance of endosomal cholesterol flow and intracellular cholesterol homeostasis remains unclear. We have previously described that hepatic MLN64 overexpression increases liver cholesterol content and induces liver damage. Here, we studied the function of MLN64 in normal and NPC1-deficient cells and we evaluated whether MLN64 overexpressing cells exhibit alterations in mitochondrial function. We used recombinant-adenovirus-mediated MLN64 gene transfer to overexpress MLN64 in mouse liver and hepatic cells; and RNA interference to down-regulate MLN64 in NPC1-deficient cells. In MLN64-overexpressing cells, we found increased mitochondrial cholesterol content and decreased glutathione (GSH) levels and ATPase activity. Furthermore, we found decreased mitochondrial membrane potential and mitochondrial fragmentation and increased mitochondrial superoxide levels in MLN64-overexpressing cells and in NPC1-deficient cells. Consequently, MLN64 expression was increased in NPC1-deficient cells and reduction of its expression restore mitochondrial membrane potential and mitochondrial superoxide levels. Our findings suggest that MLN64 overexpression induces an increase in mitochondrial cholesterol content and consequently a decrease in mitochondrial GSH content leading to mitochondrial dysfunction. In addition, we demonstrate that MLN64 expression is increased in NPC cells and plays a key role in cholesterol transport into the mitochondria.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Cholesterol/metabolism , Liver/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/physiology , Niemann-Pick Diseases/metabolism , Animals , CHO Cells , Cricetulus , Dependovirus/genetics , Genetic Vectors/administration & dosage , Glutathione/metabolism , Hep G2 Cells , Humans , Liver/cytology , Membrane Potential, Mitochondrial , Mice , Mitochondria/metabolism , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/physiopathology , Superoxides/metabolism
17.
Stem Cell Reports ; 7(4): 719-734, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27618724

ABSTRACT

While epigenetic modifications have emerged as attractive substrates to integrate environmental changes into the determination of cell identity and function, specific signals that directly activate these epigenetic modifications remain unknown. Here, we examine the role of atypical protein kinase C (aPKC)-mediated Ser436 phosphorylation of CBP, a histone acetyltransferase, in adult hippocampal neurogenesis and memory. Using a knockin mouse strain (CbpS436A) in which the aPKC-CBP pathway is deficient, we observe impaired hippocampal neuronal differentiation, maturation, and memory and diminished binding of CBP to CREB in 6-month-old CbpS436A mice, but not at 3 months of age. Importantly, elevation of CREB activity rescues these deficits, and CREB activity is reduced whereas aPKC activity is increased in the murine hippocampus as they age from 3 to 6 months regardless of genotype. Thus, the aPKC-CBP pathway is a homeostatic compensatory mechanism that modulates hippocampal neurogenesis and memory in an age-dependent manner in response to reduced CREB activity.


Subject(s)
CREB-Binding Protein/metabolism , Hippocampus/metabolism , Neurogenesis , Protein Kinase C/metabolism , Signal Transduction , Age Factors , Animals , Biomarkers , Cell Differentiation , Memory , Mice , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Binding
18.
Cell Rep ; 17(4): 1022-1036, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27760310

ABSTRACT

Maternal diabetes is known to adversely influence brain development in offspring. Here, we provide evidence that this involves the circulating metabolite methylglyoxal, which is increased in diabetes, and its detoxifying enzyme, glyoxalase 1 (Glo1), which when mutated is associated with neurodevelopmental disorders. Specifically, when Glo1 levels were decreased in embryonic mouse cortical neural precursor cells (NPCs), this led to premature neurogenesis and NPC depletion embryonically and long-term alterations in cortical neurons postnatally. Increased circulating maternal methylglyoxal caused similar changes in embryonic cortical precursors and neurons and long-lasting changes in cortical neurons and NPCs in adult offspring. Depletion of embryonic and adult NPCs was also observed in murine offspring exposed to a maternal diabetic environment. Thus, the Glo1-methylglyoxal pathway integrates maternal and NPC metabolism to regulate neural development, and perturbations in this pathway lead to long-lasting alterations in adult neurons and NPC pools.


Subject(s)
Adult Stem Cells/metabolism , Diabetes Mellitus, Experimental/metabolism , Lactoylglutathione Lyase/metabolism , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/metabolism , Pyruvaldehyde/metabolism , Signal Transduction , Animals , Animals, Newborn , Behavior, Animal , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Diabetes Mellitus, Experimental/pathology , Diabetes, Gestational/metabolism , Diabetes, Gestational/pathology , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neurogenesis , Neurons/cytology , Neurons/metabolism , Pregnancy
19.
Neuron ; 91(5): 988-1004, 2016 Sep 07.
Article in English | MEDLINE | ID: mdl-27545711

ABSTRACT

The neural stem cell decision to self-renew or differentiate is tightly regulated by its microenvironment. Here, we have asked about this microenvironment, focusing on growth factors in the embryonic cortex at a time when it is largely comprised of neural precursor cells (NPCs) and newborn neurons. We show that cortical NPCs secrete factors that promote their maintenance, while cortical neurons secrete factors that promote differentiation. To define factors important for these activities, we used transcriptome profiling to identify ligands produced by NPCs and neurons, cell-surface mass spectrometry to identify receptors on these cells, and computational modeling to integrate these data. The resultant model predicts a complex growth factor environment with multiple autocrine and paracrine interactions. We tested this communication model, focusing on neurogenesis, and identified IFNγ, Neurturin (Nrtn), and glial-derived neurotrophic factor (GDNF) as ligands with unexpected roles in promoting neurogenic differentiation of NPCs in vivo.


Subject(s)
Cerebral Cortex/growth & development , Glial Cell Line-Derived Neurotrophic Factor/physiology , Interferon-gamma/physiology , Models, Neurological , Nerve Growth Factors/metabolism , Neurogenesis/physiology , Neurturin/physiology , Animals , Cell Differentiation/physiology , Cerebral Cortex/metabolism , Humans , Ligands , Mice , Neural Stem Cells/physiology , Neurons/metabolism , Neurons/physiology , Primary Cell Culture , Transcriptome/physiology
20.
Cell Cycle ; 14(20): 3270-81, 2015.
Article in English | MEDLINE | ID: mdl-26359534

ABSTRACT

p63 is a member of the p53 family that regulates the survival of neural precursors in the adult brain. However, the relative importance of p63 in the developing brain is still unclear, since embryonic p63(-/-) mice display no apparent deficits in neural development. Here, we have used a more definitive conditional knockout mouse approach to address this issue, crossing p63(fl/fl) mice to mice carrying a nestin-CreERT2 transgene that drives inducible recombination in neural precursors following tamoxifen treatment. Inducible ablation of p63 following tamoxifen treatment of mice on embryonic day 12 resulted in highly perturbed forebrain morphology including a thinner cortex and enlarged lateral ventricles 3 d later. While the normal cortical layers were still present following acute p63 ablation, cortical precursors and neurons were both reduced in number due to widespread cellular apoptosis. This apoptosis was cell-autonomous, since it also occurred when p63 was inducibly ablated in primary cultured cortical precursors. Finally, we demonstrate increased expression of the mRNA encoding another p53 family member, ΔNp73, in cortical precursors of p63(-/-) but not tamoxifen-treated p63(fl/fl);R26YFP(fl/fl);nestin-CreERT2(+/Ø) embryos. Since ΔNp73 promotes cell survival, then this compensatory increase likely explains the lack of an embryonic brain phenotype in p63(-/-) mice. Thus, p63 plays a key prosurvival role in the developing mammalian brain.


Subject(s)
Central Nervous System/embryology , Central Nervous System/metabolism , Embryonic Development/physiology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Phosphoproteins/deficiency , Trans-Activators/deficiency , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Pregnancy , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL