Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.838
Filtrar
1.
Int J Mol Sci ; 25(8)2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38673939

RESUMO

Polyglutamine (polyQ)-encoding CAG repeat expansions represent a common disease-causing mutation responsible for several dominant spinocerebellar ataxias (SCAs). PolyQ-expanded SCA proteins are toxic for cerebellar neurons, with Purkinje cells (PCs) being the most vulnerable. RNA interference (RNAi) reagents targeting transcripts with expanded CAG reduce the level of various mutant SCA proteins in an allele-selective manner in vitro and represent promising universal tools for treating multiple CAG/polyQ SCAs. However, it remains unclear whether the therapeutic targeting of CAG expansion can be achieved in vivo and if it can ameliorate cerebellar functions. Here, using a mouse model of SCA7 expressing a mutant Atxn7 allele with 140 CAGs, we examined the efficacy of short hairpin RNAs (shRNAs) targeting CAG repeats expressed from PHP.eB adeno-associated virus vectors (AAVs), which were introduced into the brain via intravascular injection. We demonstrated that shRNAs carrying various mismatches with the CAG target sequence reduced the level of polyQ-expanded ATXN7 in the cerebellum, albeit with varying degrees of allele selectivity and safety profile. An shRNA named A4 potently reduced the level of polyQ-expanded ATXN7, with no effect on normal ATXN7 levels and no adverse side effects. Furthermore, A4 shRNA treatment improved a range of motor and behavioral parameters 23 weeks after AAV injection and attenuated the disease burden of PCs by preventing the downregulation of several PC-type-specific genes. Our results show the feasibility of the selective targeting of CAG expansion in the cerebellum using a blood-brain barrier-permeable vector to attenuate the disease phenotype in an SCA mouse model. Our study represents a significant advancement in developing CAG-targeting strategies as a potential therapy for SCA7 and possibly other CAG/polyQ SCAs.


Assuntos
Ataxina-7 , Dependovirus , Modelos Animais de Doenças , Peptídeos , Fenótipo , RNA Interferente Pequeno , Ataxias Espinocerebelares , Expansão das Repetições de Trinucleotídeos , Animais , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/terapia , Ataxias Espinocerebelares/metabolismo , Peptídeos/genética , Dependovirus/genética , Camundongos , Ataxina-7/genética , Ataxina-7/metabolismo , Expansão das Repetições de Trinucleotídeos/genética , RNA Interferente Pequeno/genética , Vetores Genéticos/genética , Vetores Genéticos/administração & dosagem , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Camundongos Transgênicos , Cerebelo/metabolismo , Cerebelo/patologia , Humanos , Terapia Genética/métodos , Alelos
2.
Dis Model Mech ; 17(5)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38616770

RESUMO

Dystonia is thought to arise from abnormalities in the motor loop of the basal ganglia; however, there is an ongoing debate regarding cerebellar involvement. We adopted an established cerebellar dystonia mouse model by injecting ouabain to examine the contribution of the cerebellum. Initially, we examined whether the entopeduncular nucleus (EPN), substantia nigra pars reticulata (SNr), globus pallidus externus (GPe) and striatal neurons were activated in the model. Next, we examined whether administration of a dopamine D1 receptor agonist and dopamine D2 receptor antagonist or selective ablation of striatal parvalbumin (PV, encoded by Pvalb)-expressing interneurons could modulate the involuntary movements of the mice. The cerebellar dystonia mice had a higher number of cells positive for c-fos (encoded by Fos) in the EPN, SNr and GPe, as well as a higher positive ratio of c-fos in striatal PV interneurons, than those in control mice. Furthermore, systemic administration of combined D1 receptor agonist and D2 receptor antagonist and selective ablation of striatal PV interneurons relieved the involuntary movements of the mice. Abnormalities in the motor loop of the basal ganglia could be crucially involved in cerebellar dystonia, and modulating PV interneurons might provide a novel treatment strategy.


Assuntos
Corpo Estriado , Modelos Animais de Doenças , Distonia , Interneurônios , Parvalbuminas , Proteínas Proto-Oncogênicas c-fos , Receptores de Dopamina D2 , Animais , Interneurônios/metabolismo , Interneurônios/efeitos dos fármacos , Parvalbuminas/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Distonia/patologia , Distonia/metabolismo , Distonia/fisiopatologia , Corpo Estriado/patologia , Corpo Estriado/metabolismo , Receptores de Dopamina D2/metabolismo , Receptores de Dopamina D1/metabolismo , Cerebelo/patologia , Cerebelo/metabolismo , Ouabaína/farmacologia , Camundongos Endogâmicos C57BL , Camundongos , Masculino
3.
Zhonghua Bing Li Xue Za Zhi ; 53(5): 452-457, 2024 May 08.
Artigo em Chinês | MEDLINE | ID: mdl-38678325

RESUMO

Objective: To investigate the role of RNA m6A methylation in mediating cerebellar dysplasia through analyzing the phenotypes of the mouse cerebella and the expression of several key m6A regulators upon hypobaric hypoxia treatment. Methods: Five-day old C57/BL6 mice were exposed to hypobaric hypoxia for 9 days. The status of mouse cerebellar development was analyzed by comparing the body weights, brain weights and histological features. Immunostaining of cell-type-specific markers was performed to analyze the cerebellar morphology. Real-time PCR, Western blot and immunohistochemical staining were performed to detect the expression of key m6A regulators in the mouse cerebella. Results: Compared with the control, the body weights, brain weights and cerebellar volumes of hypobaric hypoxic mice were significantly reduced (P<0.01). The expression of specific markers in different cells, including NeuN (mature neuron), Calbindin-D28K (Purkinje cell) and GFAP (astrocyte), was decreased in hypobaric hypoxic mouse cerebella (P<0.01), accompanied with disorganized cellular structure. The expression of methyltransferase METTL3 was significantly down-regulated in the cerebella of hypobaric hypoxic mice (P<0.05). Conclusions: Hypobaric hypoxia stimulation causes mouse cerebellar dysplasia, with structural abnormalities in mature granular neurons, Purkinje cells and astrocytes. Expression of METTL3 is decreased in hypobaric hypoxic mice cerebellum compared with that of normobaric normoxic mice, suggesting that its mediated RNA m6A methylation may play an important role in hypobaric hypoxia-induced mouse cerebellar dysplasia.


Assuntos
Calbindinas , Cerebelo , Proteínas de Ligação a DNA , Hipóxia , Metiltransferases , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso , Células de Purkinje , Animais , Camundongos , Cerebelo/metabolismo , Hipóxia/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Calbindinas/metabolismo , Calbindinas/genética , Metiltransferases/metabolismo , Metiltransferases/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteína Glial Fibrilar Ácida/genética , Astrócitos/metabolismo , Regulação para Baixo , Metilação , Adenosina/metabolismo , Adenosina/análogos & derivados , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/genética
4.
Sci Adv ; 10(17): eade1650, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38669326

RESUMO

While the kinesin-2 motors KIF3A and KIF3B have essential roles in ciliogenesis and Hedgehog (HH) signal transduction, potential role(s) for another kinesin-2 motor, KIF17, in HH signaling have yet to be explored. Here, we investigated the contribution of KIF17 to HH-dependent cerebellar development, where Kif17 is expressed in both HH-producing Purkinje cells and HH-responding cerebellar granule neuron progenitors (CGNPs). Germline Kif17 deletion in mice results in cerebellar hypoplasia due to reduced CGNP proliferation, a consequence of decreased HH pathway activity mediated through decreased Sonic HH (SHH) protein. Notably, Purkinje cell-specific Kif17 deletion partially phenocopies Kif17 germline mutants. Unexpectedly, CGNP-specific Kif17 deletion results in the opposite phenotype-increased CGNP proliferation and HH target gene expression due to altered GLI transcription factor processing. Together, these data identify KIF17 as a key regulator of HH-dependent cerebellar development, with dual and opposing roles in HH-producing Purkinje cells and HH-responding CGNPs.


Assuntos
Cerebelo , Cerebelo/anormalidades , Proteínas Hedgehog , Cinesinas , Malformações do Sistema Nervoso , Células de Purkinje , Animais , Cinesinas/metabolismo , Cinesinas/genética , Cerebelo/metabolismo , Cerebelo/crescimento & desenvolvimento , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Camundongos , Células de Purkinje/metabolismo , Transdução de Sinais , Proliferação de Células , Camundongos Knockout , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/citologia , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteína GLI1 em Dedos de Zinco/genética , Deficiências do Desenvolvimento
5.
Cell Tissue Res ; 396(2): 255-267, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38502237

RESUMO

Joubert syndrome (JS) is a recessively inherited congenital ataxia characterized by hypotonia, psychomotor delay, abnormal ocular movements, intellectual disability, and a peculiar cerebellar and brainstem malformation, the "molar tooth sign." Over 40 causative genes have been reported, all encoding for proteins implicated in the structure or functioning of the primary cilium, a subcellular organelle widely present in embryonic and adult tissues. In this paper, we developed an in vitro neuronal differentiation model using patient-derived induced pluripotent stem cells (iPSCs), to evaluate possible neurodevelopmental defects in JS. To this end, iPSCs from four JS patients harboring mutations in distinct JS genes (AHI1, CPLANE1, TMEM67, and CC2D2A) were differentiated alongside healthy control cells to obtain mid-hindbrain precursors and cerebellar granule cells. Differentiation was monitored over 31 days through the detection of lineage-specific marker expression by qRT-PCR, immunofluorescence, and transcriptomics analysis. All JS patient-derived iPSCs, regardless of the mutant gene, showed a similar impairment to differentiate into mid-hindbrain and cerebellar granule cells when compared to healthy controls. In addition, analysis of primary cilium count and morphology showed notable ciliary defects in all differentiating JS patient-derived iPSCs compared to controls. These results confirm that patient-derived iPSCs are an accessible and relevant in vitro model to analyze cellular phenotypes connected to the presence of JS gene mutations in a neuronal context.


Assuntos
Anormalidades Múltiplas , Diferenciação Celular , Cerebelo , Cerebelo/anormalidades , Anormalidades do Olho , Células-Tronco Pluripotentes Induzidas , Doenças Renais Císticas , Neurônios , Retina , Retina/anormalidades , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Humanos , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Cerebelo/patologia , Cerebelo/metabolismo , Neurônios/metabolismo , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Retina/metabolismo , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Doenças Renais Císticas/metabolismo , Masculino , Feminino , Mutação/genética , Cílios/metabolismo
6.
Neuroreport ; 35(6): 374-379, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38526932

RESUMO

Lethal giant larvae 1 (LGL1) is originally recognized as a tumor suppressor, implicated in maintaining cell polarity in Drosophila and mammalian cells. Cell polarity plays a crucial role in tumorigenesis. We previously established Pax2-LGL1 -/- conditional knockout mice but did not focus on the tumorigenesis in cerebellar primordium. HE staining was used to detect the morphological structure of the cerebellar primordium during early embryonic development in Pax2-LGL1 -/- mice. Immunofluorescence assays were used to detect the expression of polar molecules. TUNEL staining assessed tissue apoptosis. Our findings reveal that deletion of LGL1 leads to the emergence of neuroblastoma-like tissues within the cerebellum primordium during early embryogenesis. This outcome can be attributed to alterations in expression patterns of polar molecules Cdc42 and ß-catenin following early deletion of LGL1, resulting in loss of cell polarity among neuroepithelial cells and subsequent formation of tumor-like tissues. However, further histological examination demonstrated that these tumor-like tissues disappear from embryonic day 15.5 onwards within the cerebellar primordium of Pax2-LGL1 -/- mice due to apoptosis-mediated cellular compensation. Our data emphasize the importance of LGL1 in maintaining neuroepithelial cell polarity and reveal a novel role for LGL1 in regulating tumorigenesis and ablation in the cerebellar primordium.


Assuntos
Apoptose , Cerebelo , Glicoproteínas , Animais , Camundongos , Carcinogênese , Cerebelo/metabolismo , Glicoproteínas/metabolismo , Mamíferos/metabolismo , Camundongos Knockout
7.
J Neurosci ; 44(15)2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38383496

RESUMO

Almost all medulloblastomas (MB) of the Wingless/Int-1 (WNT) type are characterized by hotspot mutations in CTNNB1, and mouse models have convincingly demonstrated the tumor-initiating role of these mutations. Additional alterations in SMARCA4 are detected in ∼20% of WNT MB, but their functional role is mostly unknown. We, therefore, amended previously described brain lipid binding protein (Blbp)-cre::Ctnnb1(ex3)fl/wt mice by the introduction of floxed Smarca4 alleles. Unexpectedly, mutated and thereby stabilized ß-catenin on its own induced severe developmental phenotypes in male and female Blbp-cre::Ctnnb1(ex3)fl/wt mice in our hands, including a thinned cerebral cortex, hydrocephalus, missing cerebellar layering, and cell accumulations in the brainstem and cerebellum. An additional loss of SMARCA4 even resulted in prenatal death for most mice. Respective Blbp-cre::Ctnnb1(ex3)fl/wt::Smarca4fl/rec mutants (male and female) developed large proliferative lesions in the cerebellum evolving from E13.5 to E16.5. Histological and molecular analysis of these lesions by DNA methylation profiling and single-cell RNA sequencing suggested an origin in early undifferentiated SOX2-positive cerebellar progenitors. Furthermore, upregulated WNT signaling, altered actin/cytoskeleton organization, and reduced neuronal differentiation were evident in mutant cells. In vitro, cells harboring alterations in both Ctnnb1 and Smarca4 were negatively selected and did not show tumorigenic potential after transplantation in adult female recipient mice. However, in cerebellar explant cultures, mutant cells displayed significantly increased proliferation, suggesting an important role of the embryonic microenvironment in the development of lesions. Altogether, these results represent an important first step toward the unraveling of tumorigenic mechanisms induced by aberrant WNT signaling and SMARCA4 deficiency.


Assuntos
Neoplasias Cerebelares , Meduloblastoma , Animais , Feminino , Masculino , Camundongos , beta Catenina/genética , beta Catenina/metabolismo , Cerebelo/metabolismo , Mutação/genética , Transdução de Sinais , Microambiente Tumoral
8.
Sci Rep ; 14(1): 3236, 2024 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-38332227

RESUMO

Machado-Joseph disease (MJD) is a neurodegenerative disorder characterized by widespread neuronal death affecting the cerebellum. Cell therapy can trigger neuronal replacement and neuroprotection through bystander effects providing a therapeutic option for neurodegenerative diseases. Here, human control (CNT) and MJD iPSC-derived neuroepithelial stem cells (NESC) were established and tested for their therapeutic potential. Cells' neuroectodermal phenotype was demonstrated. Brain organoids obtained from the Control NESC showed higher mRNA levels of genes related to stem cells' bystander effects, such as BDNF, NEUROD1, and NOTCH1, as compared with organoids produced from MJD NESC, suggesting that Control NESC have a higher therapeutic potential. Graft-derived glia and neurons, such as cells positive for markers of cerebellar neurons, were detected six months after NESC transplantation in mice cerebella. The graft-derived neurons established excitatory and inhibitory synapses in the host cerebella, although CNT neurons exhibited higher excitatory synapse numbers compared with MJD neurons. Cell grafts, mainly CNT NESC, sustained the bystander effects through modulation of inflammatory interleukins (IL1B and IL10), neurotrophic factors (NGF), and neurogenesis-related proteins (Msi1 and NeuroD1), for six months in the mice cerebella. Altogether this study demonstrates the long-lasting therapeutic potential of human iPSC-derived NESC in the cerebellum.


Assuntos
Células-Tronco Pluripotentes Induzidas , Doença de Machado-Joseph , Camundongos , Animais , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Efeito Espectador , Neurônios/metabolismo , Cerebelo/metabolismo , Doença de Machado-Joseph/metabolismo
10.
Nat Genet ; 56(3): 383-394, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38291334

RESUMO

Brain region-specific degeneration and somatic expansions of the mutant Huntingtin (mHTT) CAG tract are key features of Huntington's disease (HD). However, the relationships among CAG expansions, death of specific cell types and molecular events associated with these processes are not established. Here, we used fluorescence-activated nuclear sorting (FANS) and deep molecular profiling to gain insight into the properties of cell types of the human striatum and cerebellum in HD and control donors. CAG expansions arise at mHTT in striatal medium spiny neurons (MSNs), cholinergic interneurons and cerebellar Purkinje neurons, and at mutant ATXN3 in MSNs from SCA3 donors. CAG expansions in MSNs are associated with higher levels of MSH2 and MSH3 (forming MutSß), which can inhibit nucleolytic excision of CAG slip-outs by FAN1. Our data support a model in which CAG expansions are necessary but may not be sufficient for cell death and identify transcriptional changes associated with somatic CAG expansions and striatal toxicity.


Assuntos
Corpo Estriado , Doença de Huntington , Humanos , Animais , Cerebelo/metabolismo , Doença de Huntington/genética , Modelos Animais de Doenças
11.
J Neuroimmunol ; 387: 578293, 2024 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-38266443

RESUMO

Purkinje cell cytoplasmic autoantibody type 1 (PCA1), also known as anti-Yo, is a 'high-risk' paraneoplastic antibody, associated with rapidly progressive cerebellar syndrome. In patients with this syndrome, various MRI abnormalities have been documented, including atrophy in the cerebellum and brainstem, T2 hyperintensity in the brainstem and spinal cord, and cranial nerve enhancement. This report introduces an imaging finding, cerebellar leptomeningeal enhancement, which was observed in all three cases at early stages. Despite neurological deterioration, all patients underwent immunotherapy, and subsequent follow-up MRI revealed resolution of the leptomeningeal enhancement, suggesting that this feature is distinct from meningeal carcinomatosis.


Assuntos
Doenças Cerebelares , Degeneração Paraneoplásica Cerebelar , Síndromes Paraneoplásicas , Humanos , Degeneração Paraneoplásica Cerebelar/diagnóstico por imagem , Degeneração Paraneoplásica Cerebelar/metabolismo , Células de Purkinje/metabolismo , Autoanticorpos , Proteínas do Tecido Nervoso , Cerebelo/metabolismo , Doenças Cerebelares/diagnóstico por imagem , Doenças Cerebelares/metabolismo
12.
Int J Dev Neurosci ; 84(2): 122-133, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38238938

RESUMO

Dietary polyphenol consumption is associated with a wide range of neuroprotective effects by improving mitochondrial function and signaling. Consequently, the use of polyphenol supplementation has been investigated as an approach to prevent neurodevelopmental diseases during gestation; however, the data obtained are still very inconclusive, mostly because of the difficulty of choosing the correct doses and period of administration to properly prevent neurodegenerative diseases without undermining normal brain development. Thus, we aimed to evaluate the effect of naringin supplementation during the third week of gestation on mitochondrial health and signaling in the cerebellum of 21-day-old offspring. The offspring born to naringin-supplemented dams displayed higher mitochondrial mass, membrane potential, and superoxide content in the cerebellum without protein oxidative damage. Such alterations were associated with dynamin-related protein 1 (DRP1) and phosphorylated AKT (p-AKT) downregulation, whereas the sirtuin 3 (SIRT3) levels were strongly upregulated. Our findings suggest that high dietary polyphenol supplementation during gestation may reduce mitochondrial fission and affect mitochondrial dynamics even 3 weeks after delivery via SIRT3 and p-AKT. Although the offspring born to naringin dams did not present neurobehavioral defects, the mitochondrial alterations elicited by naringin may potentially interfere during neurodevelopment and need to be further investigated.


Assuntos
Flavanonas , Sirtuína 3 , Ratos , Animais , Feminino , Gravidez , Ratos Wistar , Sirtuína 3/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Cerebelo/metabolismo , Suplementos Nutricionais , Mitocôndrias/metabolismo , Polifenóis/metabolismo
13.
Exp Brain Res ; 242(3): 619-637, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38231387

RESUMO

Cilia are organelles extend from cells to sense external signals for tuning intracellular signaling for optimal cellular functioning. They have evolved sensory and motor roles in various cells for tissue organization and homeostasis in development and post-development. More than a thousand genes are required for cilia function. Mutations in them cause multisystem disorders termed ciliopathies. The null mutations in CC2D2A result in Meckel syndrome (MKS), which is embryonic lethal, whereas patients who have missense mutations in the C2 domain of CC2D2A display Joubert syndrome (JBTS). They survive with blindness and mental retardation. How C2 domain defects cause disease conditions is not understood. To answer this question, C2 domain of Cc2d2a (mice gene) was knocked down (KD) in IMCD-3 cells by shRNA. This resulted in defective cilia morphology observed by immunofluorescence analysis. To further probe the cellular signaling alteration in affected cells, gene expression profiling was done by RNAseq and compared with the controls. Bioinformatics analysis revealed that the differentially expressed genes (DEGs) have functions in cilia. Among the 61 cilia DEGs identified, 50 genes were downregulated and 11 genes were upregulated. These cilia genes are involved in cilium assembly, protein trafficking to the cilium, intraflagellar transport (IFT), cellular signaling like polarity patterning, and Hedgehog signaling pathway. This suggests that the C2 domain of CC2D2A plays a critical role in cilia assembly and molecular signaling hosted in cilia for cellular homeostasis. Taken together, the missense mutations in the C2 domain of CC2D2A seen in JBTS might have affected cilia-mediated signaling in neurons of the retina and brain.


Assuntos
Anormalidades Múltiplas , Cerebelo , Anormalidades do Olho , Doenças Renais Císticas , Doenças Renais Policísticas , Retina , Animais , Camundongos , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Domínios C2 , Cerebelo/metabolismo , Cerebelo/anormalidades , Cílios/genética , Cílios/metabolismo , Proteínas do Citoesqueleto/genética , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Mutação/genética , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Retina/anormalidades
14.
Purinergic Signal ; 20(2): 181-192, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37458955

RESUMO

L-Glutamate (L-Glu) is an amino acid present in the diet that plays a fundamental role in the central nervous system, as the main excitatory neurotransmitter participating in learning and memory processes. In addition, the nucleoside adenosine has a crucial role in L-Glu metabolism, by regulating the liberation of this neurotransmitter through four different receptors: A1, A2A, A2B and A3, which activate (A2A and A2B) or inhibit (A1 and A3) adenylate cyclase pathway. L-Glu at high concentrations can act as a neurotoxin and induce oxidative stress. The study of the oxidative stress correlated with an excess of L-Glu consumption during maternity is key to understand its effects on foetuses and neonates. Previous studies have shown that there is a change in the receptor levels in the brain of pregnant rats and their foetuses when mothers are administered L-Glu during gestation; however, its effect on the cerebellum is unknown. Cerebellum is known to be responsible for motor, cognitive and emotional functions, so its possible involvement after L-Glu consumption is an important issue to study. Therefore, the aim of the present work was to study the effect of L-Glu exposure during gestation and lactation on oxidative stress biomarkers and neurotransmitter receptors from the cerebellum of foetuses and neonates. After maternal L-Glu intake during gestation, oxidative stress was increased, as the ionotropic L-Glu receptors, and GluR1 AMPA subunit levels were altered in foetuses. A1 adenosine receptor suffered changes after L-Glu treatment during gestation, lactation or both, in lactating neonate cerebellum, while adenylate cyclase activity remain unaltered. Further studies will be necessary to elucidate the importance of L-Glu intake and its possible excitotoxicity in the cerebellum of Wistar rats during the pregnancy period and their involvement in long-term neurodegeneration.


Assuntos
Ácido Glutâmico , Efeitos Tardios da Exposição Pré-Natal , Humanos , Animais , Ratos , Feminino , Gravidez , Ácido Glutâmico/metabolismo , Lactação , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Ratos Wistar , Adenosina/metabolismo , Receptores de AMPA , Adenilil Ciclases/metabolismo , Adenilil Ciclases/farmacologia , Cerebelo/metabolismo , Feto/metabolismo , Estresse Oxidativo , Neurotransmissores/metabolismo , Neurotransmissores/farmacologia
15.
J Chem Neuroanat ; 135: 102367, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38043916

RESUMO

BACKGROUND: Chronic use of tramadol can cause neurotoxic effects and subsequently cause neurodegeneration in the cerebellum. The main damage mechanisms identified are oxidative stress and inflammation. Currently, we investigated the effects of coenzyme Q10 (CoQ10) in attenuates of neurodegeneration in the cerebellum induced by chronic exposure to tramadol. MATERIAL AND METHODS: Seventy-two male mature albino rats were allocated into four equal groups, including; non-treated group, CoQ10 group (which received CoQ10 at 200 mg/kg/day orally for three weeks), tramadol group (which received tramadol hydrochloride at 50 mg/kg/day orally for three weeks), and tramadol+CoQ10 group (which received tramadol and CoQ10 at the same doses as the previous groups). Tissue samples were obtained for stereological, immunohistochemical, biochemical, and molecular evaluations. Also, functional tests were performed to evaluate behavioral properties. RESULTS: We found a significant increase in stereological parameters, antioxidant factors (catalase, glutathione, and superoxide dismutase), and behavioral function scores in the tramadol+CoQ10 group compared to the tramadol group (p < 0.05). In addition, malondialdehyde levels, the density of apoptotic cells, as well as the expression of pro-inflammatory (tumor necrosis factor-alpha, interleukin 1 beta, and interleukin 6) and autophagy (lysosome-associated membrane protein 2, autophagy-related 5, beclin 1, and autophagy-related 12) genes were considerably reduced in the tramadol+CoQ10 group compared to the tramadol group (p < 0.05). CONCLUSION: We conclude that the administration of CoQ10 has neuroprotective effects in the cerebellum of rats that have chronic exposure to tramadol.


Assuntos
Tramadol , Ratos , Masculino , Animais , Tramadol/farmacologia , Ubiquinona/farmacologia , Ubiquinona/metabolismo , Ubiquinona/uso terapêutico , Antioxidantes/farmacologia , Estresse Oxidativo , Cerebelo/metabolismo
16.
Sci Rep ; 13(1): 19831, 2023 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-37963945

RESUMO

Adenosine kinase (ADK), the major adenosine-metabolizing enzyme, plays a key role in brain development and disease. In humans, mutations in the Adk gene have been linked to developmental delay, stunted growth, and intellectual disability. To better understand the role of ADK in brain development, it is important to dissect the specific roles of the two isoforms of the enzyme expressed in the cytoplasm (ADK-S) and cell nucleus (ADK-L). We, therefore, studied brain development in Adk-tg transgenic mice, which only express ADK-S in the absence of ADK-L throughout development. In the mutant animals, we found a reduction in the overall brain, body size, and weight during fetal and postnatal development. As a major developmental abnormality, we found a profound change in the foliation pattern of the cerebellum. Strikingly, our results indicated aberrant Purkinje cells arborization at P9 and accelerated cell death at P6 and P9. We found defects in cerebellar cell proliferation and migration using a bromodeoxyuridine (BrdU)-based cell proliferation assay at postnatal day 7. Our data demonstrate that dysregulation of ADK expression during brain development profoundly affects brain growth and differentiation.


Assuntos
Adenosina Quinase , Encéfalo , Camundongos , Animais , Humanos , Adenosina Quinase/genética , Adenosina Quinase/metabolismo , Encéfalo/metabolismo , Camundongos Transgênicos , Cerebelo/metabolismo , Isoformas de Proteínas/metabolismo
17.
Biomolecules ; 13(11)2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-38002281

RESUMO

We recently identified protein kinase N1 (PKN1) as a negative gatekeeper of neuronal AKT protein kinase activity during postnatal cerebellar development. The developing cerebellum is specifically vulnerable to hypoxia-ischemia (HI), as it occurs during hypoxic-ischemic encephalopathy, a condition typically caused by oxygen deprivation during or shortly after birth. In that context, activation of the AKT cell survival pathway has emerged as a promising new target for neuroprotective interventions. Here, we investigated the role of PKN1 in an in vitro model of HI, using postnatal cerebellar granule cells (Cgc) derived from Pkn1 wildtype and Pkn1-/- mice. Pkn1-/- Cgc showed significantly higher AKT phosphorylation, resulting in reduced caspase-3 activation and improved survival after HI. Pkn1-/- Cgc also showed enhanced axonal outgrowth on growth-inhibitory glial scar substrates, further pointing towards a protective phenotype of Pkn1 knockout after HI. The specific PKN1 phosphorylation site S374 was functionally relevant for the enhanced axonal outgrowth and AKT interaction. Additionally, PKN1pS374 shows a steep decrease during cerebellar development. In summary, we demonstrate the pathological relevance of the PKN1-AKT interaction in an in vitro HI model and establish the relevant PKN1 phosphorylation sites, contributing important information towards the development of specific PKN1 inhibitors.


Assuntos
Hipóxia-Isquemia Encefálica , Fármacos Neuroprotetores , Animais , Camundongos , Hipóxia-Isquemia Encefálica/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Hipóxia , Cerebelo/metabolismo , Animais Recém-Nascidos
18.
J Trace Elem Med Biol ; 80: 127318, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37864919

RESUMO

BACKGROUND: Aluminum and nickel are potent neurotoxicants to which humans are constantly exposed. Previous studies have demonstrated that these two metals can affect the motor system, but their effects on the cerebellum, a central nervous system region with the highest number of neurons, have remained largely unexplored. Therefore, we conducted a study to investigate the adverse effects of Al, Ni, and Al+Ni in vivo. METHODS: In our study, seven male Sprague Dawley rats per group were orally exposed to deionized water, 0.2 mg/kg of Ni, 1 mg/kg of Al, and 0.2 mg/kg of Ni + 1 mg/kg of Al (as a binary heavy metals mixture; HMM), respectively. RESULTS: Ni, Al, and HMM exposed rats accumulated higher levels of Al and Ni compared to controls, and HMM treated animals had higher levels of Ca and Fe in the cerebellum (p < 0.05). Malondialdehyde (MDA) levels were significantly (p < 0.05) higher in the HMM, Ni, and Al treated groups compared to the control group that received deionized water. Superoxide dismutase (SOD), catalase (CAT), glutathione (GSH), and glutathione peroxidase (GPx) activities were significantly (p < 0.05) reduced in the HMM, Ni, and Al treated groups compared to the control group that received deionized water. Ni, Al, and HMM significantly (p < 0.05) shortened the length of time of the grip in comparison to the control. Nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) levels were significantly decreased in the nickel, Al, and heavy metal mixture groups compared with the control group. Moreover, there was a significant decrease in the activity of acetylcholinesterase (AChE) and a increase in cyclooxygenase-2 (COX-2) activity in the Ni, Al, and HMM treated groups compared to the control group. CONCLUSION: HMM exposed animals had significantly poorer performance in the Rotarod test (p < 0.05) than controls. Al and Ni induced impairment of cerebellar function at various levels.


Assuntos
Metais Pesados , Transtornos Motores , Humanos , Ratos , Masculino , Animais , Acetilcolinesterase/metabolismo , Níquel/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fator de Crescimento Neural/metabolismo , Fator de Crescimento Neural/farmacologia , Estresse Oxidativo , Ratos Sprague-Dawley , Metais Pesados/farmacologia , Antioxidantes/metabolismo , Cerebelo/metabolismo , Catalase/metabolismo , Glutationa/metabolismo , Superóxido Dismutase/metabolismo , Água/farmacologia
19.
J Toxicol Sci ; 48(7): 429-439, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37394656

RESUMO

Granule cell-selective toxicity of methylmercury in the cerebellum is one of the main unresolved issues in the pathogenesis of Minamata disease. Rats were orally administered methylmercury chloride (10 mg/kg/day) for 5 consecutive days, and their brains were harvested on days 1, 7, 14, 21, or 28 after the last administration for histological examination of the cerebellum. It was found that methylmercury caused a marked degenerative change to the granule cell layers but not to the Purkinje cell layers. The generative change of the granule cell layer was due to cell death, including apoptosis, which occurred at day 21 and beyond after the methylmercury administration. Meanwhile, cytotoxic T-lymphocytes and macrophages had infiltrated the granule cell layer. Additionally, granule cells are shown to be a cell type susceptible to TNF-α. Taken together, these results suggest that methylmercury causes small-scale damage to granule cells, triggering the infiltration of cytotoxic T-lymphocytes and macrophages into the granule cell layer, which secrete tumor necrosis factor-α (TNF-α) to induce apoptosis in granule cells. This chain is established based on the susceptibility of granule cells to methylmercury, the ability of cytotoxic T lymphocytes and macrophages to synthesize and secrete TNF-α, and the sensitivity of granule cells to TNF-α and methylmercury. We propose to call the pathology of methylmercury-induced cerebellar damage the "inflammation hypothesis."


Assuntos
Compostos de Metilmercúrio , Ratos , Animais , Compostos de Metilmercúrio/toxicidade , Compostos de Metilmercúrio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Cerebelo/metabolismo , Neurônios , Apoptose
20.
J Physiol ; 601(17): 3905-3920, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37431690

RESUMO

Kinesin family member 2C (KIF2C)/mitotic centromere-associated kinesin (MCAK), is thought to be oncogenic as it is involved in tumour progression and metastasis. Moreover, it also plays a part in neurodegenerative conditions like Alzheimer's disease and psychiatric disorders such as suicidal schizophrenia. Our previous study conducted on mice demonstrated that KIF2C is widely distributed in various regions of the brain, and is localized in synaptic spines. Additionally, it regulates microtubule dynamic properties through its own microtubule depolymerization activity, thereby affecting AMPA receptor transport and cognitive behaviour in mice. In this study, we show that KIF2C regulates the transport of mGlu1 receptors in Purkinje cells by binding to Rab8. KIF2C deficiency in Purkinje cells results in abnormal gait, reduced balance ability and motor incoordination in male mice. These data suggest that KIF2C is essential for maintaining normal transport and synaptic function of mGlu1 and motor coordination in mice. KEY POINTS: KIF2C is localized in synaptic spines of hippocampus neurons, and regulates excitatory transmission, synaptic plasticity and cognitive behaviour. KIF2C is extensively expressed in the cerebellum, and we investigated its functions in development and synaptic transmission of cerebellar Purkinje cells. KIF2C deficiency in Purkinje cells alters the expression of metabotropic glutamate receptor 1 (mGlu1) and the AMPA receptor GluA2 subunit at Purkinje cell synapses, and changes excitatory synaptic transmission, but not inhibitory transmission. KIF2C regulates the transport of mGlu1 receptors in Purkinje cells by binding to Rab8. KIF2C deficiency in Purkinje cells affects motor coordination, but not social behaviour in male mice.


Assuntos
Células de Purkinje , Receptores de Glutamato Metabotrópico , Masculino , Animais , Camundongos , Células de Purkinje/fisiologia , Receptores de AMPA/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Cerebelo/metabolismo , Proteínas de Transporte/metabolismo , Sinapses/metabolismo , Proteínas de Ciclo Celular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA