Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30.525
Filtrar
1.
J Mol Biol ; 435(24): 168354, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-37935253

RESUMO

Mutations causing loss of PTEN lipid phosphatase activity can promote cancer, benign tumors (PHTS), and neurodevelopmental disorders (NDDs). Exactly how they preferentially trigger distinct phenotypic outcomes has been puzzling. Here, we demonstrate that PTEN mutations differentially allosterically bias P loop dynamics and its connection to the catalytic site, affecting catalytic activity. NDD-related mutations are likely to sample conformations of the functional wild-type state, while sampled conformations for the strong, cancer-related driver mutation hotspots favor catalysis-primed conformations, suggesting that NDD mutations are likely to be weaker, and our large-scale simulations show why. Prenatal PTEN isoform expression data suggest exons 5 and 7, which harbor NDD mutations, as cancer-risk carriers. Since cancer requires more than a single mutation, our conformational and genomic analysis helps discover how same protein mutations can foster different clinical manifestations, articulates a role for co-occurring background latent driver mutations, and uncovers relationships of splicing isoform expression to life expectancy.


Assuntos
Transtorno Autístico , Encéfalo , Neoplasias , Humanos , Transtorno Autístico/genética , Encéfalo/enzimologia , Mutação , Neoplasias/genética , Isoformas de Proteínas/genética , PTEN Fosfo-Hidrolase/genética
2.
J Chem Neuroanat ; 133: 102345, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37778734

RESUMO

Thimet oligopeptides (THOP 1) is a metal-dependent peptidase involved in the metabolism of neuropeptides and the presentation of peptides via MHC-1. It has been shown to play a role in the regulation of protein-protein interactions and the metabolism of intracellular peptides. THOP 1 is associated with important biological processes such as metabolism and neurodegenerative diseases, among others. The objective of this study is to elucidate the distribution of THOP 1 in the Bufo marinus brain. The analysis of THOP 1 amino acid sequences indicates that they have been conserved throughout evolution, with significant homology observed across various phyla. When comparing amphibians with other species, more than 70% identity can be identified. Immunohistochemistry analysis of the toad's brain has demonstrated that the enzyme has a ubiquitous distribution, consistent with previous findings in mammals. THOP 1 can be found in important areas of the brain, such as bulb, thalamic nuclei, striatum, hypothalamus, and among others. Nonetheless, THOP 1 is consistently localized within the nucleus, a pattern also observed in the rat brain. Therefore, based on these results, the toad appears to be an excellent model for studying the general biology of THOP 1, given the substantial homology of this enzyme with mammals and its similarity in distribution within the brain.


Assuntos
Bufo marinus , Metaloendopeptidases , Animais , Ratos , Bufo marinus/metabolismo , Peptídeos/metabolismo , Núcleos Talâmicos/enzimologia , Metaloendopeptidases/metabolismo , Encéfalo/enzimologia
3.
Int J Mol Sci ; 24(17)2023 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-37686081

RESUMO

Proteins with extended polyglutamine regions are associated with several neurodegenerative disorders, including Huntington's disease. Intracellular proteolytic processing of these proteins is not well understood. In particular, it is unclear whether long polyglutamine fragments resulting from the proteolysis of these proteins can be potentially cleaved by the proteasome. Here, we studied the susceptibility of the glutamine-glutamine bond to proteolysis by the proteasome using oligoglutamine-containing peptides with a fluorophore/quencher pair. We found that the addition of the 11S proteasomal regulator (also known as PA28) significantly accelerated the hydrolysis of oligoglutamine-containing peptides by the 20S proteasome. Unexpectedly, a similar effect was observed for the 26S proteasome in the presence of the 11S regulator. LC/MS data revealed that the hydrolysis of our peptides with both 20S and 26S proteasomes leads to N-terminal fragments containing two or three glutamine residues and that the hydrolysis site does not change after the addition of the 11S regulator. This was confirmed by the docking experiment, which shows that the preferred hydrolysis site is located after the second/third glutamine residue. Inhibitory analysis revealed that trypsin-like specificity is mainly responsible for the proteasomal hydrolysis of the glutamine-glutamine bond. Together, our results indicate that both 20S and 26S proteasomes are capable of degrading the N-terminal part of oligoglutamine fragments, while the 11S regulator significantly accelerates the hydrolysis without changing its specificity. This data suggests that proteasome activity may be enhanced in relation to polyglutamine substrates present in neurons in the early stages of polyglutamine disorders.


Assuntos
Encéfalo , Peptídeos , Complexo de Endopeptidases do Proteassoma , Proteólise , Animais , Camundongos , Encéfalo/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Hidrólise , Peptídeos/metabolismo , Tripsina/metabolismo
4.
Sheng Wu Gong Cheng Xue Bao ; 39(7): 2656-2668, 2023 Jul 25.
Artigo em Chinês | MEDLINE | ID: mdl-37584122

RESUMO

Somatostatin (SST) is an inhibitory polypeptide hormone that plays an important role in a variety of biological processes. Somatostatin receptor 2 (SSTR2) is the most widely expressed somatostatin receptor. However, the specific cell types expressing Sstr2 in the tissues have not been investigated. In this study, we detected the expression pattern of SSTR2 protein in mouse at different development stages, including the embryonic 15.5 days and the postnatal 1, 7, 15 days as well as 3 and 6 months, by multicolour immunofluorescence analyses. We found that Sstr2 was expressed in some specific cells types of several tissues, including the neuronal cells and astrocytes in the brain, the mesenchymal cells, the hematopoietic cells, the early hematopoietic stem cells, and the B cells in the bone marrow, the macrophages, the type Ⅱ alveolar epithelial cells, and the airway ciliated cells in the lung, the epithelial cells and the neuronal cells in the intestine, the hair follicle cells, the gastric epithelial cells, the hematopoietic stem cells and the nerve fibre in the spleen, and the tubular epithelial cells in the kidney. This study identified the specific cell types expressing Sstr2 in mouse at different developmental stages, providing new insights into the physiological function of SST and SSTR2 in several cell types.


Assuntos
Encéfalo , Animais , Camundongos , Encéfalo/enzimologia , Encéfalo/metabolismo
5.
J Biol Chem ; 299(9): 105128, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37543361

RESUMO

Gliomas are the most prevalent primary tumor of the central nervous system. Despite advances in imaging technologies, neurosurgical techniques, and radiotherapy, a cure for high-grade glioma remains elusive. Several groups have reported that protein tyrosine phosphatase receptor type Z (PTPRZ) is highly expressed in glioblastoma, and that targeting PTPRZ attenuates tumor growth in mice. PTPRZ is modified with diverse glycan, including the PTPRZ-unique human natural killer-1 capped O-mannosyl core M2 glycans. However, the regulation and function of these unique glycans are unclear. Using CRISPR genome-editing technology, we first demonstrated that disruption of the PTPRZ gene in human glioma LN-229 cells resulted in profoundly reduced tumor growth in xenografted mice, confirming the potential of PTPRZ as a therapeutic target for glioma. Furthermore, multiple glycan analyses revealed that PTPRZ derived from glioma patients and from xenografted glioma expressed abundant levels of human natural killer-1-capped O-Man glycans via extrinsic signals. Finally, since deficiency of O-Man core M2 branching enzyme N-acetylglucosaminyltransferase IX (GnT-IX) was reported to reduce PTPRZ protein levels, we disrupted the GnT-IX gene in LN-229 cells and found a significant reduction of glioma growth both in vitro and in the xenograft model. These results suggest that the PTPR glycosylation enzyme GnT-IX may represent a promising therapeutic target for glioma.


Assuntos
Glioma , N-Acetilglucosaminiltransferases , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores , Animais , Humanos , Camundongos , Encéfalo/enzimologia , Encéfalo/fisiopatologia , Glioma/fisiopatologia , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Polissacarídeos/metabolismo , Linhagem Celular Tumoral , Feminino , Camundongos SCID , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/deficiência , Proteínas Tirosina Fosfatases Classe 5 Semelhantes a Receptores/metabolismo , Técnicas de Silenciamento de Genes
6.
Bull Exp Biol Med ; 175(2): 191-195, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37462806

RESUMO

Hypothermia in homeotherms significantly affects the neurotransmitter systems of the brain, including the cholinergic system. The function of the brain cholinergic system during prolonged moderate hypothermia is not known yet. We studied the effects of moderate hypothermia of various durations on the activity and kinetic parameters of synaptic acetylcholinesterase in rat brain. Immediately after body temperature decrease to 30°C, the efficiency of synaptic acetylcholinesterase catalysis significantly increases due to changes in both the maximum rate of reaction (Vmax; the rate of reaction when the enzyme is saturated with substrate) and Michaelis constant (Km). However, in the dynamics of prolonged hypothermia (1-3 h), it decreases to a level of intact animals, which was associated with normalization of the kinetic parameters of the enzyme. The detected changes in the kinetic parameters of the enzyme are compensatory and can be associated with both its reversible post-translational modifications and changes in the annular lipids.


Assuntos
Encéfalo , Hipotermia Induzida , Masculino , Animais , Ratos Wistar , Encéfalo/enzimologia , Acetilcolinesterase/metabolismo , Fatores de Tempo , Ratos , Cinética
7.
J Neurosci ; 43(19): 3567-3581, 2023 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-36977578

RESUMO

Metachromatic leukodystrophy (MLD) is a rare, inherited, demyelinating lysosomal storage disorder caused by mutations in the arylsulfatase-A gene (ARSA). In patients, levels of functional ARSA enzyme are diminished and lead to deleterious accumulation of sulfatides. Herein, we demonstrate that intravenous administration of HSC15/ARSA restored the endogenous murine biodistribution of the corresponding enzyme, and overexpression of ARSA corrected disease biomarkers and ameliorated motor deficits in Arsa KO mice of either sex. In treated Arsa KO mice, when compared with intravenously administered AAV9/ARSA, significant increases in brain ARSA activity, transcript levels, and vector genomes were observed with HSC15/ARSA Durability of transgene expression was established in neonate and adult mice out to 12 and 52 weeks, respectively. Levels and correlation between changes in biomarkers and ARSA activity required to achieve functional motor benefit was also defined. Finally, we demonstrated blood-nerve, blood-spinal and blood-brain barrier crossing as well as the presence of circulating ARSA enzyme activity in the serum of healthy nonhuman primates of either sex. Together, these findings support the use of intravenous delivery of HSC15/ARSA-mediated gene therapy for the treatment of MLD.SIGNIFICANCE STATEMENT Herein, we describe the method of gene therapy adeno-associated virus (AAV) capsid and route of administration selection leading to an efficacious gene therapy in a mouse model of metachromatic leukodystrophy. We demonstrate the therapeutic outcome of a new naturally derived clade F AAV capsid (AAVHSC15) in a disease model and the importance of triangulating multiple end points to increase the translation into higher species via ARSA enzyme activity and biodistribution profile (with a focus on the CNS) with that of a key clinically relevant biomarker.


Assuntos
Arilsulfatases , Terapia Genética , Leucodistrofia Metacromática , Animais , Camundongos , Macaca fascicularis , Arilsulfatases/genética , Camundongos Knockout , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/fisiopatologia , Leucodistrofia Metacromática/terapia , Modelos Animais de Doenças , Dependovirus/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Encéfalo/enzimologia , Transtornos Motores/genética , Transtornos Motores/terapia , Administração Intravenosa , Biomarcadores/análise , Barreira Hematoencefálica , Masculino , Feminino , Humanos
8.
Science ; 379(6634): 795-802, 2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-36821679

RESUMO

Interfacing electronics with neural tissue is crucial for understanding complex biological functions, but conventional bioelectronics consist of rigid electrodes fundamentally incompatible with living systems. The difference between static solid-state electronics and dynamic biological matter makes seamless integration of the two challenging. To address this incompatibility, we developed a method to dynamically create soft substrate-free conducting materials within the biological environment. We demonstrate in vivo electrode formation in zebrafish and leech models, using endogenous metabolites to trigger enzymatic polymerization of organic precursors within an injectable gel, thereby forming conducting polymer gels with long-range conductivity. This approach can be used to target specific biological substructures and is suitable for nerve stimulation, paving the way for fully integrated, in vivo-fabricated electronics within the nervous system.


Assuntos
Biopolímeros , Encéfalo , Condutividade Elétrica , Enzimas , Sistema Nervoso Periférico , Animais , Biopolímeros/biossíntese , Encéfalo/enzimologia , Eletrodos , Eletrônica , Enzimas/metabolismo , Sanguessugas , Modelos Animais , Sistema Nervoso Periférico/enzimologia , Polimerização , Peixe-Zebra
9.
Science ; 379(6634): 758-759, 2023 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-36821689

RESUMO

An electrically conducting soft polymer is synthesized within living tissue.


Assuntos
Biopolímeros , Encéfalo , Eletrônica , Sistema Nervoso Periférico , Animais , Biopolímeros/biossíntese , Condutividade Elétrica , Géis , Encéfalo/enzimologia , Sistema Nervoso Periférico/enzimologia
10.
Aging Cell ; 22(3): e13780, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36644807

RESUMO

The contribution of cellular senescence to the behavioral changes observed in the elderly remains elusive. Here, we observed that aging is associated with a decline in protein phosphatase 2A (PP2A) activity in the brains of zebrafish and mice. Moreover, drugs activating PP2A reversed age-related behavioral changes. We developed a transgenic zebrafish model to decrease PP2A activity in the brain through knockout of the ppp2r2c gene encoding a regulatory subunit of PP2A. Mutant fish exhibited the behavioral phenotype observed in old animals and premature accumulation of neural cells positive for markers of cellular senescence, including senescence-associated ß-galactosidase, elevated levels cdkn2a/b, cdkn1a, senescence-associated secretory phenotype gene expression, and an increased level of DNA damage signaling. The behavioral and cell senescence phenotypes were reversed in mutant fish through treatment with the senolytic ABT263 or diverse PP2A activators as well as through cdkn1a or tp53 gene ablation. Senomorphic function of PP2A activators was demonstrated in mouse primary neural cells with downregulated Ppp2r2c. We conclude that PP2A reduction leads to neural cell senescence thereby contributing to age-related behavioral changes and that PP2A activators have senotherapeutic properties against deleterious behavioral effects of brain aging.


Assuntos
Comportamento Animal , Encéfalo , Senescência Celular , Envelhecimento Cognitivo , Neurônios , Proteína Fosfatase 2 , Senoterapia , Animais , Camundongos , Compostos de Anilina/farmacologia , Animais Geneticamente Modificados , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , beta-Galactosidase/genética , beta-Galactosidase/metabolismo , Biomarcadores/metabolismo , Encéfalo/enzimologia , Senescência Celular/efeitos dos fármacos , Senescência Celular/genética , Senescência Celular/fisiologia , Envelhecimento Cognitivo/fisiologia , Inibidor de Quinase Dependente de Ciclina p15/genética , Inibidor de Quinase Dependente de Ciclina p15/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Dano ao DNA , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Modelos Animais , Mutação , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/fisiologia , Cultura Primária de Células , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Senoterapia/farmacologia , Sulfonamidas/farmacologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra
11.
Nature ; 611(7937): 827-834, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36418452

RESUMO

Vacuolar-type adenosine triphosphatases (V-ATPases)1-3 are electrogenic rotary mechanoenzymes structurally related to F-type ATP synthases4,5. They hydrolyse ATP to establish electrochemical proton gradients for a plethora of cellular processes1,3. In neurons, the loading of all neurotransmitters into synaptic vesicles is energized by about one V-ATPase molecule per synaptic vesicle6,7. To shed light on this bona fide single-molecule biological process, we investigated electrogenic proton-pumping by single mammalian-brain V-ATPases in single synaptic vesicles. Here we show that V-ATPases do not pump continuously in time, as suggested by observing the rotation of bacterial homologues8 and assuming strict ATP-proton coupling. Instead, they stochastically switch between three ultralong-lived modes: proton-pumping, inactive and proton-leaky. Notably, direct observation of pumping revealed that physiologically relevant concentrations of ATP do not regulate the intrinsic pumping rate. ATP regulates V-ATPase activity through the switching probability of the proton-pumping mode. By contrast, electrochemical proton gradients regulate the pumping rate and the switching of the pumping and inactive modes. A direct consequence of mode-switching is all-or-none stochastic fluctuations in the electrochemical gradient of synaptic vesicles that would be expected to introduce stochasticity in proton-driven secondary active loading of neurotransmitters and may thus have important implications for neurotransmission. This work reveals and emphasizes the mechanistic and biological importance of ultraslow mode-switching.


Assuntos
Encéfalo , Mamíferos , ATPases Vacuolares Próton-Translocadoras , Animais , Trifosfato de Adenosina/metabolismo , Encéfalo/enzimologia , Encéfalo/metabolismo , Mamíferos/metabolismo , Prótons , Vesículas Sinápticas/enzimologia , Vesículas Sinápticas/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo , Neurotransmissores/metabolismo , Transmissão Sináptica , Fatores de Tempo , Cinética
12.
Neurocrit Care ; 37(1): 293-301, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35534658

RESUMO

BACKGROUND: According to international guidelines, neuroprognostication in comatose patients after cardiac arrest (CA) is performed using a multimodal approach. However, patients undergoing extracorporeal membrane oxygenation (ECMO) may have longer pharmacological sedation and show alteration in biological markers, potentially challenging prognostication. Here, we aimed to assess whether routinely used predictors of poor neurological outcome also exert an acceptable performance in patients undergoing ECMO after CA. METHODS: This observational retrospective study of our registry includes consecutive comatose adults after CA. Patients deceased within 36 h and not undergoing prognostic tests were excluded. Veno-arterial ECMO was initiated in patients < 80 years old presenting a refractory CA, with a no flow < 5 min and a low flow ≤ 60 min on admission. Neuroprognostication test performance (including pupillary reflex, electroencephalogram, somatosensory-evoked potentials, neuron-specific enolase) toward mortality and poor functional outcome (Cerebral Performance Categories [CPC] score 3-5) was compared between patients undergoing ECMO and those without ECMO. RESULTS: We analyzed 397 patients without ECMO and 50 undergoing ECMO. The median age was 65 (interquartile range 54-74), and 69.8% of patients were men. Most had a cardiac etiology (67.6%); 52% of the patients had a shockable rhythm, and the median time to return of an effective circulation was 20 (interquartile range 10-28) minutes. Compared with those without ECMO, patients receiving ECMO had worse functional outcome (74% with CPC scores 3-5 vs. 59%, p = 0.040) and a nonsignificant higher mortality (60% vs. 47%, p = 0.080). Apart from the neuron-specific enolase level (higher in patients with ECMO, p < 0.001), the presence of prognostic items (pupillary reflex, electroencephalogram background and reactivity, somatosensory-evoked potentials, and myoclonus) related to unfavorable outcome (CPC score 3-5) in both groups was similar, as was the prevalence of at least any two such items concomitantly. The specificity of each these variables toward poor outcome was between 92 and 100% in both groups, and of the combination of at least two items, it was 99.3% in patients without ECMO and 100% in those with ECMO. The predictive performance (receiver operating characteristic curve) of their combination toward poor outcome was 0.822 (patients without ECMO) and 0.681 (patients with ECMO) (p = 0.134). CONCLUSIONS: Pending a prospective assessment on a larger cohort, in comatose patients after CA, the performance of prognostic factors seems comparable in patients with ECMO and those without ECMO. In particular, the combination of at least two poor outcome criteria appears valid across these two groups.


Assuntos
Encéfalo , Coma , Oxigenação por Membrana Extracorpórea , Parada Cardíaca , Adulto , Idoso , Idoso de 80 Anos ou mais , Encéfalo/enzimologia , Encéfalo/fisiopatologia , Coma/etiologia , Coma/fisiopatologia , Coma/terapia , Eletroencefalografia , Feminino , Parada Cardíaca/complicações , Humanos , Masculino , Fosfopiruvato Hidratase/metabolismo , Prognóstico , Estudos Prospectivos , Estudos Retrospectivos
13.
Chemosphere ; 294: 133791, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35104548

RESUMO

Several studies have indicated that hospital effluents can produce genotoxic and mutagenic effects, cytotoxicity, hematological and histological alterations, embryotoxicity, and oxidative stress in diverse water organisms, but research on the neurotoxic effects hospital wastewater materials can generate in fish is still scarce. To fill the above-described knowledge gap, this study aimed to determine whether the exposure of adult zebrafish (Danio rerio) to several proportions (0.1%, 2.5%, 3.5%) of a hospital effluent can disrupt behavior or impair redox status and acetylcholinesterase content in the brain. After 96 h of exposure to the effluent, we observed a decrease in total distance traveled and an increase in frozen time compared to the control group. Moreover, we also observed a significant increase in the levels of reactive oxygen species in the brains of the fish, especially in hydroperoxide and protein carbonyl content, relative to the control group. Our results also demonstrated that hospital effluents significantly inhibited the activity of the AChE enzyme in the brains of the fish. Our Pearson correlation demonstrated that the response to acetylcholinesterase at the lowest proportions (0.1% and 2.5%) is positively related to the oxidative stress response and the behavioral changes observed. The cohort of our studies demonstrated that the exposure of adult zebrafish to a hospital effluent induced oxidative stress and decreased acetylcholinesterase activity in the brain of these freshwater organisms, which can lead to alterations in their behavior.


Assuntos
Acetilcolinesterase , Comportamento Animal , Estresse Oxidativo , Poluentes Químicos da Água , Peixe-Zebra , Acetilcolinesterase/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Hospitais , Humanos , México , Estresse Oxidativo/efeitos dos fármacos , Carbonilação Proteica/efeitos dos fármacos , Natação , Poluentes Químicos da Água/toxicidade , Peixe-Zebra/metabolismo
14.
J Neurosci ; 42(10): 1945-1957, 2022 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-35101965

RESUMO

Phosphatase and tensin homolog (PTEN) is a major negative regulator of the phosphatidylinositol-3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) pathway. Loss-of-function mutations in PTEN have been found in a subset of patients with macrocephaly and autism spectrum disorder (ASD). PTEN loss in neurons leads to somal hypertrophy, aberrant migration, dendritic overgrowth, increased spine density, and hyperactivity of neuronal circuits. These neuronal overgrowth phenotypes are present on Pten knock-out (KO) and reconstitution with autism-associated point mutations. The mechanism underlying dendritic overgrowth in Pten deficient neurons is unclear. In this study, we examined how Pten loss impacts microtubule (MT) dynamics in both sexes using retroviral infection and transfection strategies to manipulate PTEN expression and tag the plus-end MT binding protein, end-binding protein 3 (EB3). We found Pten KO neurons sprout more new processes over time compared with wild-type (WT) neurons. We also found an increase in MT polymerization rate in Pten KO dendritic growth cones. Reducing MT polymerization rate to the WT level was sufficient to reduce dendritic overgrowth in Pten KO neurons in vitro and in vivo Finally, we found that rescue of dendritic overgrowth via inhibition of MT polymerization was sufficient to improve the performance of Pten KO mice in a spatial memory task. Taken together, our data suggests that one factor underlying PTEN loss dependent dendritic overgrowth is increased MT polymerization. This opens the possibility for an intersectional approach targeting MT polymerization and mTOR with low doses of inhibitors to achieve therapeutic gains with minimal side effects in pathologies associated with loss of neuronal PTEN function.SIGNIFICANCE STATEMENT Loss of Pten function because of genetic deletion or expression of mutations associated with autism spectrum disorder (ASD), results in overgrowth of neurons including increased total dendritic length and branching. We have discovered that this overgrowth is accompanied by increased rate of microtubule (MT) polymerization. The increased polymerization rate is insensitive to acute inhibition of mechanistic target of rapamycin (mTOR)C1 or protein synthesis. Direct pharmacological inhibition of MT polymerization can slow the polymerization rate in Pten knock-out (KO) neurons to rates seen in wild-type (WT) neurons. Correction of the MT polymerization rate rescues increased total dendritic arborization and spatial memory. Our studies suggest that phosphatase and tensin homolog (PTEN) inhibits dendritic growth through parallel regulation of protein synthesis and cytoskeletal polymerization.


Assuntos
Transtorno do Espectro Autista , Encéfalo , Microtúbulos , PTEN Fosfo-Hidrolase , Animais , Transtorno do Espectro Autista/enzimologia , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/patologia , Encéfalo/citologia , Encéfalo/enzimologia , Encéfalo/metabolismo , Feminino , Humanos , Masculino , Camundongos , Microtúbulos/metabolismo , Plasticidade Neuronal/fisiologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Polimerização , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo
15.
Mol Psychiatry ; 27(3): 1542-1551, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35087195

RESUMO

Mounting evidence supports the role of the Ras/mitogen-activated protein kinase (Ras/MAPK) pathway in neurodevelopmental disorders. Here, the authors used a genetics-first approach to examine how Ras/MAPK pathogenic variants affect the functional organization of the brain and cognitive phenotypes including weaknesses in attention and inhibition. Functional MRI was used to examine resting state functional connectivity (RSFC) in association with Ras/MAPK pathogenic variants in children with Noonan syndrome (NS). Participants (age 4-12 years) included 39 children with NS (mean age 8.44, SD = 2.20, 25 females) and 49 typically developing (TD) children (mean age 9.02, SD = 9.02, 33 females). Twenty-eight children in the NS group and 46 in the TD group had usable MRI data and were included in final analyses. The results indicated significant hyperconnectivity for the NS group within canonical visual, ventral attention, left frontoparietal and limbic networks (p < 0.05 FWE). Higher connectivity within canonical left frontoparietal and limbic networks positively correlated with cognitive function within the NS but not the TD group. Further, the NS group demonstrated significant group differences in seed-based striatal-frontal connectivity (Z > 2.6, p < 0.05 FWE). Hyperconnectivity within canonical brain networks may represent an intermediary phenotype between Ras/MAPK pathogenic variants and cognitive phenotypes, including weaknesses in attention and inhibition. Altered striatal-frontal connectivity corresponds with smaller striatal volume and altered white matter connectivity previously documented in children with NS. These results may indicate delayed maturation and compensatory mechanisms and they are important for understanding the pathophysiology underlying cognitive phenotypes in NS and in the broader population of children with neurodevelopmental disorders.


Assuntos
Sistema de Sinalização das MAP Quinases , Proteínas Quinases Ativadas por Mitógeno , Substância Branca , Proteínas ras , Atenção/fisiologia , Encéfalo/enzimologia , Encéfalo/patologia , Feminino , Humanos , Imageamento por Ressonância Magnética , Substância Branca/enzimologia , Substância Branca/patologia , Proteínas ras/metabolismo
16.
Int J Biol Sci ; 18(2): 693-706, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35002518

RESUMO

The aggregation of amyloid-ß (Aß) peptides into oligomers and fibrils is a key pathological feature of Alzheimer's disease (AD). An increasing amount of evidence suggests that oligomeric Aß might be the major culprit responsible for various neuropathological changes in AD. Death-associated protein kinase 1 (DAPK1) is abnormally elevated in brains of AD patients and plays an important role in modulating tau homeostasis by regulating prolyl isomerase Pin1 phosphorylation. However, it remains elusive whether and how Aß species influence the function of DAPK1, and whether this may further affect the function and phosphorylation of tau in neurons. Herein, we demonstrated that Aß aggregates (both oligomers and fibrils) prepared from synthetic Aß42 peptides were able to upregulate DAPK1 protein levels and thereby its function through heat shock protein 90 (HSP90)-mediated protein stabilization. DAPK1 activation not only caused neuronal apoptosis, but also phosphorylated Pin1 at the Ser71 residue, leading to tau accumulation and phosphorylation at multiple AD-related sites in primary neurons. Both DAPK1 knockout (KO) and the application of a specific DAPK1 inhibitor could effectively protect primary neurons against Aß aggregate-induced cell death and tau dysregulation, corroborating the critical role of DAPK1 in mediating Aß aggregation-induced neuronal damage. Our study suggests a mechanistic link between Aß oligomerization and tau hyperphosphorylation mediated by DAPK1, and supports the role of DAPK1 as a promising target for early intervention in AD.


Assuntos
Doença de Alzheimer/enzimologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/enzimologia , Proteínas Quinases Associadas com Morte Celular/metabolismo , Neurônios/enzimologia , Fragmentos de Peptídeos/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/genética , Animais , Apoptose/genética , Encéfalo/patologia , Proteínas Quinases Associadas com Morte Celular/deficiência , Proteínas Quinases Associadas com Morte Celular/genética , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/genética , Fosforilação , Proteínas tau/genética , Proteínas tau/metabolismo
17.
Sci Rep ; 12(1): 181, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34996949

RESUMO

A major concern with preterm birth is the risk of neurodevelopmental disability. Poor cerebral circulation leading to periods of hypoxia is believed to play a significant role in the etiology of preterm brain injury, with the first three days of life considered the period when the brain is most vulnerable. This study focused on monitoring cerebral perfusion and metabolism during the first 72 h after birth in preterm infants weighing less than 1500 g. Brain monitoring was performed by combining hyperspectral near-infrared spectroscopy to assess oxygen saturation and the oxidation state of cytochrome c oxidase (oxCCO), with diffuse correlation spectroscopy to monitor cerebral blood flow (CBF). In seven of eight patients, oxCCO remained independent of CBF, indicating adequate oxygen delivery despite any fluctuations in cerebral hemodynamics. In the remaining infant, a significant correlation between CBF and oxCCO was found during the monitoring periods on days 1 and 3. This infant also had the lowest baseline CBF, suggesting the impact of CBF instabilities on metabolism depends on the level of blood supply to the brain. In summary, this study demonstrated for the first time how continuous perfusion and metabolic monitoring can be achieved, opening the possibility to investigate if CBF/oxCCO monitoring could help identify preterm infants at risk of brain injury.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/enzimologia , Circulação Cerebrovascular , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Recém-Nascido Prematuro , Imagem Óptica , Consumo de Oxigênio , Oxigênio/sangue , Espectroscopia de Luz Próxima ao Infravermelho , Peso ao Nascer , Estabilidade Enzimática , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Recém-Nascido de muito Baixo Peso , Masculino , Oxirredução , Valor Preditivo dos Testes , Estudo de Prova de Conceito , Fatores de Tempo
18.
Sci Rep ; 12(1): 249, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-34997032

RESUMO

Older adult patients with sepsis frequently experience cognitive impairment. The roles of brain neutrophil gelatinase-associated lipocalin (NGAL) and iron in older sepsis patients remain unknown. We investigated the effects of lipopolysaccharide-induced sepsis on novel object recognition test, NGAL levels, an inflammatory mediator tumor necrosis factor-α (TNFα) levels, and iron ion levels in the hippocampus and cortex of young and aged rats. The effect of an iron chelator deferoxamine pretreatment on aged sepsis rats was also examined. Young sepsis-survivor rats did not show impaired novel object recognition, TNFα responses, or a Fe2+/Fe3+ imbalance. They showed hippocampal and cortical NGAL level elevations. Aged sepsis-survivor rats displayed a decreased object discrimination index, elevation of NGAL levels and Fe2+/Fe3+ ratio, and no TNFα responses. Pretreatment with deferoxamine prevented the reduction in the object recognition of aged sepsis-survivor rats. The elevation in hippocampal and cortical NGAL levels caused by lipopolysaccharide was not influenced by deferoxamine pretreatment. The lipopolysaccharide-induced Fe2+/Fe3+ ratio elevation was blocked by deferoxamine pretreatment. In conclusion, our findings suggest that iron homeostasis in the cortex and hippocampus contributes to the maintenance of object recognition ability in older sepsis survivors.


Assuntos
Comportamento Animal , Encéfalo/enzimologia , Disfunção Cognitiva/enzimologia , Ferro/metabolismo , Lipocalina-2/metabolismo , Reconhecimento Psicológico , Sepse/enzimologia , Fatores Etários , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/fisiopatologia , Disfunção Cognitiva/fisiopatologia , Disfunção Cognitiva/prevenção & controle , Disfunção Cognitiva/psicologia , Desferroxamina/farmacologia , Modelos Animais de Doenças , Homeostase , Masculino , Teste de Campo Aberto , Ratos Sprague-Dawley , Reconhecimento Psicológico/efeitos dos fármacos , Sepse/tratamento farmacológico , Sepse/fisiopatologia , Sepse/psicologia , Sideróforos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
19.
Nature ; 602(7895): 112-116, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35046577

RESUMO

The biological basis of male-female brain differences has been difficult to elucidate in humans. The most notable morphological difference is size, with male individuals having on average a larger brain than female individuals1,2, but a mechanistic understanding of how this difference arises remains unknown. Here we use brain organoids3 to show that although sex chromosomal complement has no observable effect on neurogenesis, sex steroids-namely androgens-lead to increased proliferation of cortical progenitors and an increased neurogenic pool. Transcriptomic analysis and functional studies demonstrate downstream effects on histone deacetylase activity and the mTOR pathway. Finally, we show that androgens specifically increase the neurogenic output of excitatory neuronal progenitors, whereas inhibitory neuronal progenitors are not increased. These findings reveal a role for androgens in regulating the number of excitatory neurons and represent a step towards understanding the origin of sex-related brain differences in humans.


Assuntos
Androgênios/farmacologia , Encéfalo/citologia , Excitabilidade Cortical/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Organoides/citologia , Organoides/efeitos dos fármacos , Caracteres Sexuais , Potenciais de Ação/efeitos dos fármacos , Androgênios/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Encéfalo/metabolismo , Contagem de Células , Feminino , Perfilação da Expressão Gênica , Histona Desacetilases/genética , Humanos , Masculino , Inibição Neural/efeitos dos fármacos , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Organoides/enzimologia , Organoides/metabolismo , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética
20.
Endocrinology ; 163(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34967909

RESUMO

Reproductive dysfunction in women has been linked to high caloric diet (HCD)-feeding and obesity. Central resistance to leptin and insulin have been shown to accompany diet-induced infertility in rodent studies, and we have previously shown that deleting suppressor of cytokine signaling 3, which is a negative regulator of leptin signaling, from all forebrain neurons partially protects mice from HCD-induced infertility. In this study, we were interested in exploring the role of protein tyrosine phosphatase 1B (PTP1B), which is a negative regulator of both leptin and insulin signaling, in the pathophysiology of HCD-induced obesity and infertility. To this end, we generated male and female neuron-specific PTP1B knockout mice and compared their body weight gain, food intake, glucose tolerance, and fertility relative to control littermates under both normal calorie diet and HCD feeding conditions. Both male and female mice with neuronal PTP1B deletion exhibited slower body weight gain in response to HCD feeding, yet only male knockout mice exhibited improved glucose tolerance compared with controls. Neuronal PTP1B deletion improved the time to first litter in HCD-fed mice but did not protect female mice from eventual HCD-induced infertility. While the mice fed a normal caloric diet remained fertile throughout the 150-day period of assessment, HCD-fed females became infertile after producing only a single litter, regardless of their genotype. These data show that neuronal PTP1B deletion is able to partially protect mice from HCD-induced obesity but is not a critical mediator of HCD-induced infertility.


Assuntos
Encéfalo/enzimologia , Infertilidade Feminina/prevenção & controle , Neurônios/enzimologia , Obesidade/prevenção & controle , Proteína Tirosina Fosfatase não Receptora Tipo 1/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 1/fisiologia , Animais , Cruzamentos Genéticos , Ingestão de Energia , Feminino , Infertilidade Feminina/etiologia , Masculino , Camundongos Endogâmicos DBA , Camundongos Knockout , Camundongos Transgênicos , Obesidade/enzimologia , Obesidade/etiologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/genética , Maturidade Sexual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...