Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.553
Filtrar
1.
Cereb Cortex ; 34(13): 94-103, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38696597

RESUMEN

Autism (or autism spectrum disorder) was initially defined as a psychiatric disorder, with the likely cause maternal behavior (the very destructive "refrigerator mother" theory). It took several decades for research into brain mechanisms to become established. Both neuropathological and imaging studies found differences in the cerebellum in autism spectrum disorder, the most widely documented being a decreased density of Purkinje cells in the cerebellar cortex. The popular interpretation of these results is that cerebellar neuropathology is a critical cause of autism spectrum disorder. We challenge that view by arguing that if fewer Purkinje cells are critical for autism spectrum disorder, then any condition that causes the loss of Purkinje cells should also cause autism spectrum disorder. We will review data on damage to the cerebellum from cerebellar lesions, tumors, and several syndromes (Joubert syndrome, Fragile X, and tuberous sclerosis). Collectively, these studies raise the question of whether the cerebellum really has a role in autism spectrum disorder. Autism spectrum disorder is now recognized as a genetically caused developmental disorder. A better understanding of the genes that underlie the differences in brain development that result in autism spectrum disorder is likely to show that these genes affect the development of the cerebellum in parallel with the development of the structures that do underlie autism spectrum disorder.


Asunto(s)
Cerebelo , Humanos , Cerebelo/patología , Trastorno del Espectro Autista/patología , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Trastorno del Espectro Autista/diagnóstico por imagen , Animales , Trastorno Autístico/patología , Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Células de Purkinje/patología
2.
Int J Mol Sci ; 25(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38673939

RESUMEN

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.


Asunto(s)
Ataxina-7 , Dependovirus , Modelos Animales de Enfermedad , Péptidos , Fenotipo , ARN Interferente Pequeño , Ataxias Espinocerebelosas , Expansión de Repetición de Trinucleótido , Animales , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/terapia , Ataxias Espinocerebelosas/metabolismo , Péptidos/genética , Dependovirus/genética , Ratones , Ataxina-7/genética , Ataxina-7/metabolismo , Expansión de Repetición de Trinucleótido/genética , ARN Interferente Pequeño/genética , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Células de Purkinje/metabolismo , Células de Purkinje/patología , Ratones Transgénicos , Cerebelo/metabolismo , Cerebelo/patología , Humanos , Terapia Genética/métodos , Alelos
3.
Zhonghua Bing Li Xue Za Zhi ; 53(5): 452-457, 2024 May 08.
Artículo en Chino | MEDLINE | ID: mdl-38678325

RESUMEN

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.


Asunto(s)
Calbindinas , Cerebelo , Proteínas de Unión al ADN , Hipoxia , Metiltransferasas , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso , Células de Purkinje , Animales , Ratones , Cerebelo/metabolismo , Hipoxia/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Células de Purkinje/metabolismo , Células de Purkinje/patología , Calbindinas/metabolismo , Calbindinas/genética , Metiltransferasas/metabolismo , Metiltransferasas/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Astrocitos/metabolismo , Regulación hacia Abajo , Metilación , Adenosina/metabolismo , Adenosina/análogos & derivados , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/genética
4.
Sci Adv ; 10(17): eade1650, 2024 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-38669326

RESUMEN

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.


Asunto(s)
Cerebelo , Cerebelo/anomalías , Proteínas Hedgehog , Cinesinas , Malformaciones del Sistema Nervioso , Células de Purkinje , Animales , Cinesinas/metabolismo , Cinesinas/genética , Cerebelo/metabolismo , Cerebelo/crecimiento & desarrollo , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Ratones , Células de Purkinje/metabolismo , Transducción de Señal , Proliferación Celular , Ratones Noqueados , Regulación del Desarrollo de la Expresión Génica , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Proteína con Dedos de Zinc GLI1/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Discapacidades del Desarrollo
5.
Handb Clin Neurol ; 200: 409-417, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38494293

RESUMEN

Gynecologic and breast malignancies are the cancers most commonly associated with paraneoplastic neurologic syndromes, of which the foremost is Yo [Purkinje cell antibody, type 1 (PCA-1)] paraneoplastic cerebellar degeneration. Yo syndrome affects women in the sixth decade and manifests as a subacute severe cerebellar ataxia. The association of the typical clinical picture with the detection of Yo antibodies in a patient's serum or CSF defines the diagnosis. Yo syndrome is always associated with a cancer, and the search for the underlying tumor should focus on ovarian and breast cancers and be repeated overtime if negative. The Yo autoantibodies are directed against the Yo antigens, aberrantly overexpressed by tumor cells with frequent somatic mutations and gene amplifications. The massive infiltration of these tumors by immune cells suggests that they are the site of the immune tolerance breakdown, leading to the destruction of Purkinje cells harboring the Yo antigens. Despite a growing understanding of the immunologic mechanisms, efficient therapeutic options are still lacking. Anti-Ri and antiamphiphysin syndromes are rarer and associated with breast cancers; a wide variety of other rare paraneoplastic neurologic syndromes have been described in association with gynecologic and breast malignancies that, though sharing some similarities, may have specific immune and genetics features leading to the immune tolerance breakdown.


Asunto(s)
Neoplasias de la Mama , Degeneración Cerebelosa Paraneoplásica , Femenino , Humanos , Neoplasias de la Mama/complicaciones , Degeneración Cerebelosa Paraneoplásica/etiología , Degeneración Cerebelosa Paraneoplásica/diagnóstico , Autoanticuerpos , Células de Purkinje/patología
6.
Neurosci Lett ; 826: 137733, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38492880

RESUMEN

Etomidate (ET) is a widely used intravenous imidazole general anesthetic, which depresses the cerebellar neuronal activity by modulating various receptors activity and synaptic transmission. In this study, we investigated the effects of ET on the cerebellar climbing fiber-Purkinje cells (CF-PC) plasticity in vitro in mice using whole-cell recording technique and pharmacological methods. Our results demonstrated that CF tetanic stimulation produced a mGluR1-dependent long-term depression (LTD) of CF-PC excitatory postsynaptic currents (EPSCs), which was enhanced by bath application of ET (10 µM). Blockade of mGluR1 receptor with JNJ16259685, ET triggered the tetanic stimulation to induce a CF-PC LTD accompanied with an increase in paired-pulse ratio (PPR). The ET-triggered CF-PC LTD was abolished by extracellular administration of an N-methyl-(D)-aspartate (NMDA) receptor antagonist, D-APV, as well as by intracellular blockade of NMDA receptors activity with MK801. Furthermore, blocking cannabinoids 1 (CB1) receptor with AM251 or chelating intracellular Ca2+ with BAPTA, ET failed to trigger the CF-PC LTD. Moreover, the ET-triggered CF-PC LTD was abolished by inhibition of protein kinase A (PKA), but not by inhibition of protein kinase C inhibiter. The present results suggest that ET acts on postsynaptic NMDA receptor resulting in an enhancement of the cerebellar CF-PC LTD through CB1 receptor/PKA cascade in vitro in mice. These results provide new evidence and possible mechanism for ET anesthesia to affect motor learning and motor coordination by regulating cerebellar CF-PC LTD.


Asunto(s)
Etomidato , Ratones , Animales , Etomidato/farmacología , Receptor Cannabinoide CB1/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Depresión Sináptica a Largo Plazo/fisiología , Sinapsis/fisiología , Cerebelo/fisiología , Plasticidad Neuronal/fisiología , Células de Purkinje/fisiología , Transmisión Sináptica , Anestésicos Intravenosos/farmacología
7.
J Neuroinflammation ; 21(1): 49, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-38355633

RESUMEN

BACKGROUND: Myeloid-derived suppressor cells (MDSCs) constitute a recently discovered bone-marrow-derived cell type useful for dealing with neuroinflammatory disorders. However, these cells are only formed during inflammatory conditions from immature myeloid cells (IMCs) that acquire immunosuppressive activity, thus being commonly gathered from diseased animals. Then, to obtain a more clinically feasible source, we characterized IMCs directly derived from healthy bone marrow and proved their potential immunosuppressive activity under pathological conditions in vitro. We then explored their neuroprotective potential in a model of human cerebellar ataxia, the Purkinje Cell Degeneration (PCD) mouse, as it displays a well-defined neurodegenerative and neuroinflammatory process that can be also aggravated by invasive surgeries. METHODS: IMCs were obtained from healthy bone marrow and co-cultured with activated T cells. The proliferation and apoptotic rate of the later were analyzed with Tag-it Violet. For in vivo studies, IMCs were transplanted by stereotactic surgery into the cerebellum of PCD mice. We also used sham-operated animals as controls of the surgical effects, as well as their untreated counterparts. Motor behavior of mice was assessed by rotarod test. The Purkinje cell density was measured by immunohistochemistry and cell death assessed with the TUNEL technique. We also analyzed the microglial phenotype by immunofluorescence and the expression pattern of inflammation-related genes by qPCR. Parametric tests were applied depending on the specific experiment: one or two way ANOVA and Student's T test. RESULTS: IMCs were proven to effectively acquire immunosuppressive activity under pathological conditions in vitro, thus acting as MDSCs. Concerning in vivo studios, sham-operated PCD mice suffered detrimental effects in motor coordination, Purkinje cell survival and microglial activation. After intracranial administration of IMCs into the cerebellum of PCD mice, no special benefits were detected in the transplanted animals when compared to untreated mice. Nonetheless, this transplant almost completely prevented the impairments caused by the surgery in PCD mice, probably by the modulation of the inflammatory patterns. CONCLUSIONS: Our work comprise two main translational findings: (1) IMCs can be directly used as they behave as MDSCs under pathological conditions, thus avoiding their gathering from diseased subjects; (2) IMCs are promising adjuvants when performing neurosurgery.


Asunto(s)
Cerebelo , Células Mieloides , Ratones , Humanos , Animales , Células Mieloides/metabolismo , Células de Purkinje/patología , Monocitos , Inmunosupresores
8.
J Neuroimmunol ; 387: 578293, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-38266443

RESUMEN

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.


Asunto(s)
Enfermedades Cerebelosas , Degeneración Cerebelosa Paraneoplásica , Síndromes Paraneoplásicos , Humanos , Degeneración Cerebelosa Paraneoplásica/diagnóstico por imagen , Degeneración Cerebelosa Paraneoplásica/metabolismo , Células de Purkinje/metabolismo , Autoanticuerpos , Proteínas del Tejido Nervioso , Cerebelo/metabolismo , Enfermedades Cerebelosas/diagnóstico por imagen , Enfermedades Cerebelosas/metabolismo
9.
Cerebellum ; 23(1): 181-196, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36729270

RESUMEN

Modeling paraneoplastic neurological diseases to understand the immune mechanisms leading to neuronal death is a major challenge given the rarity and terminal access of patients' autopsies. Here, we present a pilot study aiming at modeling paraneoplastic cerebellar degeneration with Yo autoantibodies (Yo-PCD). Female mice were implanted with an ovarian carcinoma cell line expressing CDR2 and CDR2L, the known antigens recognized by anti-Yo antibodies. To boost the immune response, we also immunized the mice by injecting antigens with diverse adjuvants and immune checkpoint inhibitors. Ataxia and gait instability were assessed in treated mice as well as autoantibody levels, Purkinje cell density, and immune infiltration in the cerebellum. We observed the production of anti-Yo antibodies in the CSF and serum of all immunized mice. Brain immunoreaction varied depending on the site of implantation of the tumor, with subcutaneous administration leading to a massive infiltration of immune cells in the meningeal spaces, choroid plexus, and cerebellar parenchyma. However, we did not observe massive Purkinje cell death nor any motor impairments in any of the experimental groups. Self-sustained neuro-inflammation might require a longer time to build up in our model. Unusual tumor antigen presentation and/or intrinsic, species-specific factors required for pro-inflammatory engagement in the brain may also constitute strong limitations to achieve massive recruitment of antigen-specific T-cells and killing of antigen-expressing neurons in this mouse model.


Asunto(s)
Ataxia Cerebelosa , Degeneración Cerebelosa Paraneoplásica , Humanos , Ratones , Femenino , Animales , Proyectos Piloto , Cerebelo/patología , Células de Purkinje/metabolismo , Ataxia Cerebelosa/patología , Autoanticuerpos
10.
J Physiol ; 601(23): 5147-5164, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37837315

RESUMEN

Many neurons that fire high-frequency action potentials express specialized voltage-gated Na channel complexes that not only conduct transient current upon depolarization, but also pass resurgent current upon repolarization. The resurgent current is associated with recovery of transient current, even at moderately negative potentials where fast inactivation is usually absorbing. The combined results of many experimental studies have led to the hypothesis that resurgent current flows upon repolarization when an endogenous blocking protein that occludes open channels at depolarized potentials is expelled by inwardly permeating Na ions. Additional observations have suggested that the position of the voltage sensor of domain IV regulates the affinity of the channel for the putative blocker. To test the effectiveness of a kinetic scheme incorporating these features, here we develop and justify a Markov model with states grounded in known Na channel conformations. Simulations were designed to investigate whether including a permeation-dependent unblocking rate constant and two open-blocked states, superimposed on conformations and voltage-sensitive movements present in all voltage-gated Na channels, is sufficient to account for the unusual gating of channels with a resurgent component. Optimizing rate constant parameters against a wide range of experimental data from cerebellar Purkinje cells demonstrates that a kinetic scheme for Na channels incorporating the novel aspects of a permeation-dependent unblock, as well as distinct high- and low-affinity blocked states, reproduces all the attributes of experimentally recorded Na currents in a physiologically plausible manner.


Asunto(s)
Células de Purkinje , Canales de Sodio , Canales de Sodio/metabolismo , Células de Purkinje/fisiología , Neuronas/fisiología , Potenciales de Acción
11.
Sci Rep ; 13(1): 15908, 2023 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-37741947

RESUMEN

The noradrenergic fibers of the locus coeruleus, together with mossy fibers and climbing fibers, comprise the three types of cerebellar afferents that modulate the cerebellar neuronal circuit. We previously demonstrated that noradrenaline (NA) modulated synaptic transmission in the mouse cerebellar cortex via adrenergic receptors (ARs). In the present study, we investigated the effect of NA on facial stimulation-evoked cerebellar molecular layer interneuron (MLI)-Purkinje cell (PC) synaptic transmission in urethane-anesthetized mice using an in vivo cell-attached recording technique and a pharmacological method. MLI-PC synaptic transmission was induced by air-puff stimulation (duration: 60 ms) of the ipsilateral whisker pad, which exhibited positive components (P1 and P2) accompanied by a pause in simple spike activity. Cerebellar molecular layer application of NA (15 µM) decreased the amplitude and area under the curve of P1, and the pause in simple spike activity, but increased the P2/P1 ratio. The NA-induced decrease in P1 amplitude was concentration-dependent, and the half-inhibitory concentration was 10.94 µM. The NA-induced depression of facial stimulation-evoked MLI-PC GABAergic synaptic transmission was completely abolished by blockade of α-ARs or α2-ARs, but not by antagonism of α1-ARs or ß-ARs. Bath application of an α2-AR agonist inhibited MLI-PC synaptic transmission and attenuated the effect of NA on the synaptic response. NA-induced depression of MLI-PC synaptic transmission was completely blocked by a mixture of α2A- and 2B-AR antagonists, and was abolished by inhibition of protein kinase A. In addition, electrical stimulation of the molecular layer evoked MLI-PC GABAergic synaptic transmission in the presence of an AMPA receptor antagonist, which was inhibited by NA through α2-ARs. Our results indicate that NA inhibits MLI-PC GABAergic synaptic transmission by reducing GABA release via an α2-AR/PKA signaling pathway.


Asunto(s)
Norepinefrina , Células de Purkinje , Animales , Ratones , Norepinefrina/farmacología , Transducción de Señal , Transmisión Sináptica , Interneuronas , Proteínas Quinasas Dependientes de AMP Cíclico
12.
Int J Mol Sci ; 24(16)2023 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-37628946

RESUMEN

Perfluorooctane sulfonate (PFOS) has been used in a wide variety of industrial and commercial products. The adverse effects of PFOS on the developing brain are becoming of a great concern. However, the molecular mechanisms of PFOS on brain development have not yet been clarified. We investigated the effect of early-life exposure to PFOS on brain development and the mechanism involved. We investigated the change in thyroid hormone (TH)-induced dendrite arborization of Purkinje cells in the primary culture of newborn rat cerebellum. We further examined the mechanism of PFOS on TH signaling by reporter gene assay, quantitative RT-PCR, and type 2 iodothyronine deiodinase (D2) assay. As low as 10-7 M PFOS suppressed thyroxine (T4)-, but not triiodothyronine (T3)-induced dendrite arborization of Purkinje cells. Reporter gene assay showed that PFOS did not affect TRα1- and TRß1-mediated transcription in CV-1 cells. RT-PCR showed that PFOS suppressed D2 mRNA expression in the absence of T4 in primary cerebellar cells. D2 activity was also suppressed by PFOS in C6 glioma-derived cells. These results indicate that early-life exposure of PFOS disrupts TH-mediated cerebellar development possibly through the disruption of D2 activity and/or mRNA expression, which may cause cerebellar dysfunction.


Asunto(s)
Cerebelo , Yoduro Peroxidasa , Animales , Ratas , Yoduro Peroxidasa/genética , Células de Purkinje , ARN Mensajero
13.
Exp Neurol ; 369: 114511, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37634697

RESUMEN

Purkinje cells are the sole output neurons of the cerebellar cortex and play central roles in the integration of cerebellum-related motor coordination and memory. The loss or dysfunction of Purkinje cells due to cerebellar atrophy leads to severe ataxia. Here we used in vivo transplantation to examine the function of human iPS cell-derived cerebellar progenitors in adult transgenic mice in which Purkinje-specific cell death occurs due to cytotoxicity of polyglutamines. Transplantation using cerebellar organoids (42-48 days in culture), which are rich in neural progenitors, showed a viability of >50% 4 weeks after transplantation. STEM121+ grafted cells extended their processes toward the deep cerebellar nuclei, superior cerebellar peduncle, and vestibulocerebellar nuclei. The transplanted cells were mostly located in the white matter, and they were not found in the Purkinje cell layer. MAP2-positive fibers seen in the molecular layer of cerebellar cortex received VGluT2 inputs from climbing fibers. Transplanted neural progenitors overgrew in the host cerebellum but were suppressed by pretreatment with the γ-secretase inhibitor DAPT. Hyperproliferation was also suppressed by transplantation with more differentiated organoids (86 days in culture) or KIRREL2-positive cells purified by FACS sorting. Transplanted cells expressed Purkinje cell markers, GABA, CALB1 and L7, though they did not show fan-shaped morphology. We attempted to improve neuronal integration of stem cell-derived cerebellar progenitors by transplantation into the adult mouse, but this was not successfully achieved. Our findings in the present study contribute to regenerative medical application for cerebellar degeneration and provide new insights into cerebellar development in future.


Asunto(s)
Células Madre Pluripotentes Inducidas , Células de Purkinje , Humanos , Ratones , Animales , Células de Purkinje/metabolismo , Cerebelo , Corteza Cerebelosa/fisiología , Ratones Transgénicos
14.
J Physiol ; 601(17): 3905-3920, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37431690

RESUMEN

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.


Asunto(s)
Células de Purkinje , Receptores de Glutamato Metabotrópico , Masculino , Animales , Ratones , Células de Purkinje/fisiología , Receptores AMPA/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Cerebelo/metabolismo , Proteínas Portadoras/metabolismo , Sinapsis/metabolismo , Proteínas de Ciclo Celular/metabolismo
15.
Int. j. morphol ; 41(3): 825-830, jun. 2023. ilus, tab
Artículo en Inglés | LILACS | ID: biblio-1514291

RESUMEN

SUMMARY: The cerebellum is a crucial area of the hindbrain that plays an essential role in balancing, excitement control, and subtle and accurate functions. Studies have shown that long-term use of D-galactose in mice, as with the symptoms of aging, causes morphological and functional disorders in the brain. This study was performed to evaluate the changes in the cerebellum cortex tissue and the measurement of reactive oxygen species (ROS) in the cerebellum following the induction of aging in mice by D-galactose. Accordingly, subjects were randomly assigned into two groups: Normal saline group and Aging group (D-galactose). To create an aging model, D- galactose, and saline solution (sodium chloride 0.9 %) were used. After completing the preparation and passage of the tissue, the cerebellum specimens were cut in 5 microns thickness and then stained with hematoxylin-eosin stain and finally examined under a Nikon microscope. Quantitative variables were analyzed by SPSS software using T-test. In the observations of cerebellum tissue samples, in the aged induced group by D-galactose, the most changes were observed in the Neuron purkinjense (Purkinje cells) layer. In the observations of the cerebellum tissue samples of aging group induced by D-galactose, the most changes were observed in the Neuron purkinjense, and the arrangement and placement of these cells were disorientated. The nucleus positioning was not central, and the Neuron purkinjense induced by aging were seen in different morphological forms. Necrosis, Chromatolysis, and Pyknosis were found. Based on the results, D-galactose (induction of aging) causes pathological changes in the cerebellar cortex, especially in the Neuron purkinjense layer.


El cerebelo es un área crucial del rombencéfalo que desempeña un papel esencial en el equilibrio, el control de la excitación y las funciones sutiles y precisas. Los estudios han demostrado que el uso a largo plazo de D-galactosa en ratones, al igual que con los síntomas del envejecimiento, provoca trastornos morfológicos y funcionales en el cerebro. Este estudio se realizó para evaluar los cambios en el tejido de la corteza del cerebelo y la medición de especies reactivas de oxígeno (ROS) en el cerebelo luego de la inducción del envejecimiento en ratones por D-galactosa. En consecuencia, los sujetos fueron asignados aleatoriamente a dos grupos: grupo de solución salina normal y grupo de envejecimiento (D-galactosa). Para crear un modelo de envejecimiento, se utilizaron D-galactosa y solución salina (cloruro de sodio al 0,9 %). Después de completar la preparación y el paso del tejido, las muestras de cerebelo se cortaron en un grosor de 5 µm y luego se tiñeron con tinción de hematoxilina-eosina y finalmente se examinaron bajo un microscopio Nikon. Las variables cuantitativas se analizaron mediante el software SPSS utilizando la prueba T. En las observaciones de muestras de tejido de cerebelo, en el grupo envejecido inducido por D-galactosa, la mayoría de los cambios se observaron en la capa de neuronas purkinjenses (células de Purkinje). En las observaciones de las muestras de tejido del cerebelo del grupo de envejecimiento inducidas por D-galactosa, la mayoría de los cambios se observaron en las neuronas purkinjenses, y la disposición y ubicación de estas células estaban desorientadas. El posicionamiento del núcleo no era central y las neuronas purkinjenses inducidas por el envejecimiento se observaban en diferentes formas morfológicas. Se encontró necrosis, cromatólisis y picnosis. Según los resultados, la D-galactosa (inducción del envejecimiento) provoca cambios patológicos en la corteza cerebelosa, especialmente en la capa de neuronas purkinjenses.


Asunto(s)
Animales , Masculino , Ratones , Envejecimiento , Cerebelo/patología , Galactosa/administración & dosificación , Células de Purkinje , Cerebelo/citología , Especies Reactivas de Oxígeno , Modelos Animales , Ratones Endogámicos BALB C
16.
Pharmacol Res ; 191: 106773, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37068531

RESUMEN

Specific medications to combat cerebellar ataxias, a group of debilitating movement disorders characterized by difficulty with walking, balance and coordination, are still lacking. Notably, cerebellar microglial activation appears to be a common feature in different types of ataxic patients and rodent models. However, direct evidence that cerebellar microglial activation in vivo is sufficient to induce ataxia is still lacking. Here, by employing chemogenetic approaches to manipulate cerebellar microglia selectively and directly, we found that specific chemogenetic activation of microglia in the cerebellar vermis directly leads to ataxia symptoms in wild-type mice and aggravated ataxic motor deficits in 3-acetylpyridine (3-AP) mice, a classic mouse model of cerebellar ataxia. Mechanistically, cerebellar microglial proinflammatory activation induced by either chemogenetic M3D(Gq) stimulation or 3-AP modeling hyperexcites Purkinje cells (PCs), which consequently triggers ataxia. Blockade of microglia-derived TNF-α, one of the most important proinflammatory cytokines, attenuates the hyperactivity of PCs driven by microglia. Moreover, chemogenetic inhibition of cerebellar microglial activation or suppression of cerebellar microglial activation by PLX3397 and minocycline reduces the production of proinflammatory cytokines, including TNF-α, to effectively restore the overactivation of PCs and alleviate motor deficits in 3-AP mice. These results suggest that cerebellar microglial activation may aggravate the neuroinflammatory response and subsequently induce dysfunction of PCs, which in turn triggers ataxic motor deficits. Our findings thus reveal a causal relationship between proinflammatory activation of cerebellar microglia and ataxic motor symptoms, which may offer novel evidence for therapeutic intervention for cerebellar ataxias by targeting microglia and microglia-derived inflammatory mediators.


Asunto(s)
Ataxia Cerebelosa , Ratones , Animales , Ataxia Cerebelosa/inducido químicamente , Células de Purkinje/fisiología , Microglía , Factor de Necrosis Tumoral alfa/farmacología , Cerebelo , Citocinas
17.
J Chem Neuroanat ; 130: 102268, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36989922

RESUMEN

The cytotoxicity of chemotherapeutic drugs is known due to its non-selective effect not only on cancer cells but also on healthy cells. This study investigated the cerebellar alteration in rats prenatally exposed to cyclophosphamide (SK, 20 mg/kg). We also evaluated the neuroprotective potential of Ginkgo biloba (GB, 80 mg/kg/day) against possible biological changes caused by SK in the cerebellar tissues. Twenty adult female rats (weighing 230-280 g, 12 weeks old) were divided into five groups: control, sham, SK, GB, and SK + GB. After mating, pregnant rats was treated with SK in the SK and SK + GB groups and GB in the GB and SK + GB groups from day 13 to day 21 of gestation. After parturition, eight female rats were randomly selected from each group. On day 32 after birth, the cerebellar tissues were dissected and then examined under light microscope using stereological and histopathological methods. Stereological findings showed that the total number of Purkinje cells and granular cells were significantly decreased in the SK group than the control group (p < 0.05). In addition, the mean volumes of molecular layer, granular layer, white matter, and cerebellum were significantly decreased in the SK group compared to the control group (p < 0.05). In the SK + GB group, the total number Purkinje cell, and granular cells, as well as the mean volumes of molecular layer, granular layer, white matter, and cerebellum were significantly increased than the SK group (p < 0.05). Histopathological evaluation also confirmed our stereological findings in the cerebellar tissues. Our results showed that prenatal exposure to SK caused significant changes in the cerebellar architectures of rats, and that GB administration significantly attenuated the deleterious effect of SK on the cerebellar tissues.


Asunto(s)
Ginkgo biloba , Efectos Tardíos de la Exposición Prenatal , Embarazo , Humanos , Ratas , Animales , Femenino , Efectos Tardíos de la Exposición Prenatal/patología , Células de Purkinje/patología , Cerebelo/patología , Ciclofosfamida/toxicidad
18.
Mol Neurobiol ; 60(6): 3071-3085, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36790604

RESUMEN

Chronic hyperammonemia is a main contributor to the cognitive and motor impairment in patients with hepatic encephalopathy. Sustained hyperammonemia induces the TNFα expression in Purkinje neurons, mediated by NF-κB activation. The aims were the following: (1) to assess if enhanced TrkB activation by BDNF is responsible for enhanced NF-κB activation in Purkinje neurons in hyperammonemic rats, (2) to assess if this is associated with increased content of NF-κB modulated proteins such as TNFα, HMGB1, or glutaminase I, (3) to assess if these changes are due to enhanced activation of the TNFR1-S1PR2-CCR2-BDNF-TrkB pathway, (4) to analyze if increased activation of NF-κB is mediated by the PI3K-AKT pathway. It is shown that, in the cerebellum of hyperammonemic rats, increased BDNF levels enhance TrkB activation in Purkinje neurons leading to activation of PI3K, which enhances phosphorylation of AKT and of IκB, leading to increased nuclear translocation of NF-κB which enhances TNFα, HMGB1, and glutaminase I content. To assess if the changes are due to enhanced activation of the TNFR1-S1PR2-CCR2 pathway, we blocked TNFR1 with R7050, S1PR2 with JTE-013, and CCR2 with RS504393. These changes are reversed by blocking TrkB, PI3K, or the TNFR1-SP1PR2-CCL2-CCR2-BDNF-TrkB pathway at any step. In hyperammonemic rats, increased levels of BDNF enhance TrkB activation in Purkinje neurons, leading to activation of the PI3K-AKT-IκB-NF-κB pathway which increased the content of glutaminase I, HMGB1, and TNFα. Enhanced activation of this TrkB-PI3K-AKT-NF-κB pathway would contribute to impairing the function of Purkinje neurons and motor function in hyperammonemic rats and likely in cirrhotic patients with minimal or clinical hepatic encephalopathy.


Asunto(s)
Proteína HMGB1 , Encefalopatía Hepática , Hiperamonemia , Ratas , Animales , FN-kappa B/metabolismo , Células de Purkinje/metabolismo , Microglía/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína HMGB1/metabolismo , Hiperamonemia/complicaciones , Hiperamonemia/metabolismo , Glutaminasa/metabolismo , Encefalopatía Hepática/complicaciones , Encefalopatía Hepática/metabolismo
19.
Hum Mol Genet ; 32(10): 1647-1659, 2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-36621975

RESUMEN

The shaker rat carries a naturally occurring mutation leading to progressive ataxia characterized by Purkinje cell (PC) loss. We previously reported on fine-mapping the shaker locus to the long arm of the rat X chromosome. In this work, we sought to identify the mutated gene underlying the shaker phenotype and confirm its identity by functional complementation. We fine-mapped the candidate region and analyzed cerebellar transcriptomes, identifying a XM_217630.9 (Slc9a6):c.[191_195delinsA] variant in the Slc9a6 gene that segregated with disease. We generated an adeno-associated virus (AAV) targeting Slc9a6 expression to PCs using the mouse L7-6 (L7) promoter. We administered the AAV prior to the onset of PC degeneration through intracerebroventricular injection and found that it reduced the shaker motor, molecular and cellular phenotypes. Therefore, Slc9a6 is mutated in shaker and AAV-based gene therapy may be a viable therapeutic strategy for Christianson syndrome, also caused by Slc9a6 mutation.


Asunto(s)
Ataxia Cerebelosa , Discapacidad Intelectual , Ratas , Ratones , Animales , Células de Purkinje , Ataxia Cerebelosa/genética , Ataxia/genética , Mutación , Discapacidad Intelectual/genética
20.
Cerebellum ; 22(2): 240-248, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35262839

RESUMEN

Although radiation is a strategy widely used to inhibit cancer progression, which includes those of the neck and head, there are still few experimental reports on radiation effects in the cerebellum, particularly on the morphology of its cortex layers and on the Matrix metalloproteinases' (MMPs') expression, which, recently, seems to be involved in the progression of some mental disorders. Therefore, in the present study, we evaluated the morphology of the cerebellum close to the expression of MMP-9 from 4 up to 60 days after a 15-Gy X-ray single dose of X-ray irradiation had been applied to the heads of healthy adult male rats. The cerebellum of the control and irradiated groups was submitted for an analysis of cell Purkinje count, nuclear perimeter, and chromatin density using morphometric estimatives obtained from the Feulgen histochemistry reaction. In addition, immunolocalization and estimative for MMP-9 expression were determined in the cerebellar cortex on days 4, 9, 14, 25, and 60 after the irradiation procedure. Results demonstrated that irradiation produced a significant reduction in the total number of Purkinje cells and a reduction in their nuclear perimeter, along with an increase in chromatin condensation and visible nuclear fragmentation, which was also detected in the granular layer. MMP-9 expression was significantly increased on 4, 9, and 14 days, being detected around the Purkinje cells and in parallel fibres at the molecular layer. We conclude that the effects of a single dose of 15-Gy X-ray irradiation in the cerebellum were an increase in MMP-9 expression in the first 2 weeks after irradiation, especially surrounding the Purkinje cells and in the molecular layers, with morphological changes in the Purkinje cell and granular cell layers, suggesting a continuous cell loss throughout the days evaluated after irradiation.


Asunto(s)
Corteza Cerebelosa , Metaloproteinasa 9 de la Matriz , Ratas , Animales , Masculino , Rayos X , Células de Purkinje , Cerebelo , Cromatina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA