Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 185
Filtrar
1.
Cell Rep ; 43(8): 114612, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39110592

RESUMEN

Atypical sensory processing is common in autism, but how neural coding is disrupted in sensory cortex is unclear. We evaluate whisker touch coding in L2/3 of somatosensory cortex (S1) in Cntnap2-/- mice, which have reduced inhibition. This classically predicts excess pyramidal cell spiking, but this remains controversial, and other deficits may dominate. We find that c-fos expression is elevated in S1 of Cntnap2-/- mice under spontaneous activity conditions but is comparable to that of control mice after whisker stimulation, suggesting normal sensory-evoked spike rates. GCaMP8m imaging from L2/3 pyramidal cells shows no excess whisker responsiveness, but it does show multiple signs of degraded somatotopic coding. This includes broadened whisker-tuning curves, a blurred whisker map, and blunted whisker point representations. These disruptions are greater in noisy than in sparse sensory conditions. Tuning instability across days is also substantially elevated in Cntnap2-/-. Thus, Cntnap2-/- mice show no excess sensory-evoked activity, but a degraded and unstable tactile code in S1.


Asunto(s)
Trastorno Autístico , Modelos Animales de Enfermedad , Proteínas de la Membrana , Proteínas del Tejido Nervioso , Corteza Somatosensorial , Vibrisas , Animales , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/fisiopatología , Ratones , Trastorno Autístico/fisiopatología , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Ratones Noqueados , Tacto/fisiología , Ratones Endogámicos C57BL , Células Piramidales/metabolismo , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo
2.
Cell Rep ; 43(8): 114531, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39058591

RESUMEN

Spontaneous and sensory-evoked activity sculpts developing circuits. Yet, how these activity patterns intersect with cellular programs regulating the differentiation of neuronal subtypes is not well understood. Through electrophysiological and in vivo longitudinal analyses, we show that C-X-C motif chemokine ligand 14 (Cxcl14), a gene previously characterized for its association with tumor invasion, is expressed by single-bouquet cells (SBCs) in layer I (LI) of the somatosensory cortex during development. Sensory deprivation at neonatal stages markedly decreases Cxcl14 expression. Additionally, we report that loss of function of this gene leads to increased intrinsic excitability of SBCs-but not LI neurogliaform cells-and augments neuronal complexity. Furthermore, Cxcl14 loss impairs sensory map formation and compromises the in vivo recruitment of superficial interneurons by sensory inputs. These results indicate that Cxcl14 is required for LI differentiation and demonstrate the emergent role of chemokines as key players in cortical network development.


Asunto(s)
Diferenciación Celular , Quimiocinas CXC , Interneuronas , Corteza Somatosensorial , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/citología , Animales , Interneuronas/metabolismo , Quimiocinas CXC/metabolismo , Quimiocinas CXC/genética , Ratones , Ratones Endogámicos C57BL
3.
Aging Cell ; 23(8): e14177, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38760908

RESUMEN

Aging is generally associated with declining somatosensory function, which seems at odds with the high prevalence of chronic pain in older people. This discrepancy is partly related to the high prevalence of degenerative diseases such as osteoarthritis in older people. However, whether aging alters pain processing in the primary somatosensory cortex (S1), and if so, whether it promotes pain chronification is largely unknown. Herein, we report that older mice displayed prolonged nociceptive behavior following nerve injury when compared with mature adult mice. The expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) in S1 was decreased in older mice, whereas PGC-1α haploinsufficiency promoted prolonged nociceptive behavior after nerve injury. Both aging and PGC-1α haploinsufficiency led to abnormal S1 neural dynamics, revealed by intravital two-photon calcium imaging. Manipulating S1 neural dynamics affected nociceptive behavior after nerve injury: chemogenetic inhibition of S1 interneurons aggravated nociceptive behavior in naive mice; chemogenetic activation of S1 interneurons alleviated nociceptive behavior in older mice. More interestingly, adeno-associated virus-mediated expression of PGC-1α in S1 interneurons ameliorated aging-associated chronification of nociceptive behavior as well as aging-related S1 neural dynamic changes. Taken together, our results showed that aging-associated decrease of PGC-1α promotes pain chronification, which might be harnessed to alleviate the burden of chronic pain in older individuals.


Asunto(s)
Envejecimiento , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Animales , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Envejecimiento/metabolismo , Ratones , Masculino , Ratones Endogámicos C57BL , Corteza Somatosensorial/metabolismo , Dolor Crónico/metabolismo
4.
Cell Rep ; 43(5): 114197, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38733587

RESUMEN

Interneurons (INs), specifically those in disinhibitory circuits like somatostatin (SST) and vasoactive intestinal peptide (VIP)-INs, are strongly modulated by the behavioral context. Yet, the mechanisms by which these INs are recruited during active states and whether their activity is consistent across sensory cortices remain unclear. We now report that in mice, locomotor activity strongly recruits SST-INs in the primary somatosensory (S1) but not the visual (V1) cortex. This diverse engagement of SST-INs cannot be explained by differences in VIP-IN function but is absent in the presence of visual input, suggesting the involvement of feedforward sensory pathways. Accordingly, inactivating the somatosensory thalamus, but not decreasing VIP-IN activity, significantly reduces the modulation of SST-INs by locomotion. Model simulations suggest that the differences in SST-INs across behavioral states can be explained by varying ratios of VIP- and thalamus-driven activity. By integrating feedforward activity with neuromodulation, SST-INs are anticipated to be crucial for adapting sensory processing to behavioral states.


Asunto(s)
Interneuronas , Somatostatina , Péptido Intestinal Vasoactivo , Animales , Interneuronas/metabolismo , Interneuronas/fisiología , Somatostatina/metabolismo , Ratones , Péptido Intestinal Vasoactivo/metabolismo , Corteza Somatosensorial/fisiología , Corteza Somatosensorial/metabolismo , Masculino , Ratones Endogámicos C57BL , Locomoción/fisiología , Conducta Animal/fisiología , Corteza Visual/fisiología , Corteza Visual/metabolismo , Tálamo/fisiología , Tálamo/metabolismo
5.
Zhen Ci Yan Jiu ; 49(5): 480-486, 2024 May 25.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38764119

RESUMEN

OBJECTIVES: To observe the activation state and neuronal types of somatosensory cortex and the primary motor cortex induced by electroacupuncture (EA) stimulation of "Sibai" (ST2) and "Quanliao" (SI18) acupoints in mice. METHODS: Male C57BL/6J mice were randomly divided into blank control and EA groups, with 6 mice in each group. Rats of the EA group received EA stimulation (2 Hz, 0.6 mA) at ST2 and SI18 for 30 minutes. Samples were collected after EA intervention, and immunofluorescence staining was performed to quantify the expression of the c-Fos gene (proportion of c-Fos positive cells) in the somatosensory cortex and primary motor cortex. The co-labelled cells of calcium/calmodulin-dependent protein kinase Ⅱ (CaMKⅡ) and gamma-aminobutyric acid (GABA) in the somatosensory cortex and primary motor cortex were observed and counted by using microscope after immunofluorescence staining. Another 10 mice were used to detect the calcium activity of excitatory neurons in the somatosensory cortex and primary motor cortex by fiber photometry. RESULTS: In comparison with the blank control group, the number of c-Fos positive cells, and the proportion of c-Fos and CaMKⅡ co-labelled cells in both the somatosensory cortex and primary motor cortex were significantly increased after EA stimulation (P<0.05). No significant changes were found in the proportion of c-Fos and GABA co-labeled cells in both the somatosensory cortex and primary motor cortex after EA. Results of fiber optic calcium imaging technology showed that the spontaneous calcium activity of excitatory neurons in both somatosensory cortex and primary motor cortex were obviously increased during EA compared with that before EA (P<0.01), and strikingly reduced after cessation of EA compared with that during EA (P<0.05). CONCLUSIONS: Under physiological conditions, EA of ST2 and SI18 can effectively activate excitatory neurons in the somatosensory cortex and primary motor cortex.


Asunto(s)
Puntos de Acupuntura , Electroacupuntura , Ratones Endogámicos C57BL , Neuronas , Animales , Masculino , Ratones , Neuronas/metabolismo , Corteza Sensoriomotora/metabolismo , Humanos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Corteza Motora/metabolismo , Corteza Somatosensorial/metabolismo
6.
Mol Ther ; 32(6): 1721-1738, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38566414

RESUMEN

Recombinant adeno-associated viruses (AAVs) allow rapid and efficient gene delivery to the nervous system, are widely used in neuroscience research, and are the basis of FDA-approved neuron-targeting gene therapies. Here we find that an innate immune response to the AAV genome reduces dendritic length and complexity and disrupts synaptic transmission in mouse somatosensory cortex. Dendritic loss is apparent 3 weeks after injection of experimentally relevant viral titers, is not restricted to a particular capsid serotype, transgene, promoter, or production facility, and cannot be explained by responses to surgery or transgene expression. AAV-associated dendritic loss is accompanied by a decrease in the frequency and amplitude of miniature excitatory postsynaptic currents and an increase in the proportion of GluA2-lacking, calcium-permeable AMPA receptors. The AAV genome is rich in unmethylated CpG DNA, which is recognized by the innate immunoreceptor Toll-like receptor 9 (TLR9), and acutely blocking TLR9 preserves dendritic complexity and AMPA receptor subunit composition in AAV-injected mice. These results reveal unexpected impacts of an immune response to the AAV genome on neuronal structure and function and identify approaches to improve the safety and efficacy of AAV-mediated gene delivery in the nervous system.


Asunto(s)
Dendritas , Dependovirus , Vectores Genéticos , Inmunidad Innata , Transmisión Sináptica , Receptor Toll-Like 9 , Animales , Dependovirus/genética , Ratones , Dendritas/metabolismo , Receptor Toll-Like 9/metabolismo , Receptor Toll-Like 9/genética , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Receptores AMPA/genética , Receptores AMPA/metabolismo , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/inmunología , Genoma Viral
7.
Zhen Ci Yan Jiu ; 49(4): 331-340, 2024 Apr 25.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38649200

RESUMEN

OBJECTIVES: To observe whether acupuncture up-regulates chemokine CXC ligand 1 (CXCL1) in the brain to play an analgesic role through CXCL1/chemokine CXC receptor 2 (CXCR2) signaling in adjuvant induced arthritis (AIA) rats, so as to reveal its neuro-immunological mechanism underlying improvement of AIA. METHODS: BALB/c mice with relatively stable thermal pain reaction were subjected to planta injection of complete Freund adjuvant (CFA) for establishing AIA model, followed by dividing the AIA mice into simple AF750 (fluorochrome) and AF750+CXCL1 groups (n=2 in each group). AF750 labeled CXCL1 recombinant protein was then injected into the mouse's tail vein to induce elevation of CXCL1 level in blood for simulating the effect of acupuncture stimulation which has been demonstrated by our past study. In vivo small animal imaging technology was used to observe the AF750 and AF750+CXCL1-labelled target regions. After thermal pain screening, the Wistar rats with stable pain reaction were subjected to AIA modeling by injecting CFA into the rat's right planta, then were randomized into model and manual acupuncture groups (n=12 in each group). Other 12 rats that received planta injection of saline were used as the control group. Manual acupuncture (uniform reinforcing and reducing manipulations) was applied to bilateral "Zusanli" (ST36) for 4×2 min, with an interval of 5 min between every 2 min, once daily for 7 days. The thermal pain threshold was assessed by detecting the paw withdrawal latency (PWL) using a thermal pain detector. The contents of CXCL1 in the primary somatosensory cortex (S1), medial prefrontal cortex, nucleus accumbens, amygdala, periaqueductal gray and rostroventromedial medulla regions were assayed by using ELISA, and the expression levels of CXCL1, CXCR2 and mu-opioid receptor (MOR) mRNA in the S1 region were detected using real time-quantitative polymerase chain reaction. The immune-fluorescence positive cellular rate of CXCL1 and CXCR2 in S1 region was observed after immunofluorescence stain. The immunofluorescence double-stain of CXCR2 and astrocyte marker glial fibrillary acidic protein (GFAP) or neuron marker NeuN or MOR was used to determine whether there is a co-expression between them. RESULTS: In AIA mice, results of in vivo experiments showed no obvious enrichment signal of AF750 or AF750+CXCL1 in any organ of the body, while in vitro experiments showed that there was a stronger fluorescence signal of CXCL1 recombinant protein in the brain. In rats, compared with the control group, the PWL from day 0 to day 7 was significantly decreased (P<0.01) and the expression of CXCR2 mRNA in the S1 region significantly increased in the model group (P<0.05), while in comparison with the model group, the PWL from day 2 to day 7, CXCL1 content, CXCR2 mRNA expression and CXCR2 content, and MOR mRNA expression in the S1 region were significantly increased in the manual acupuncture group (P<0.05, P<0.01). Immunofluorescence stain showed that CXCR2 co-stained with NeuN and MOR in the S1 region, indicating that CXCR2 exists in neurons and MOR-positive neurons but not in GFAP positive astrocytes. CONCLUSIONS: Acupuncture can increase the content of CXCL1 in S1 region, up-regulate CXCR2 on neurons in the S1 region and improve MOR expression in S1 region of AIA rats, which may contribute to its effect in alleviating inflammatory pain.


Asunto(s)
Terapia por Acupuntura , Artritis Experimental , Quimiocina CXCL1 , Receptores de Interleucina-8B , Corteza Somatosensorial , Animales , Humanos , Masculino , Ratones , Ratas , Puntos de Acupuntura , Artritis Experimental/terapia , Artritis Experimental/metabolismo , Artritis Experimental/genética , Quimiocina CXCL1/metabolismo , Quimiocina CXCL1/genética , Inflamación/terapia , Inflamación/metabolismo , Inflamación/genética , Ratones Endogámicos BALB C , Dolor/metabolismo , Dolor/genética , Manejo del Dolor , Ratas Wistar , Receptores de Interleucina-8B/metabolismo , Receptores de Interleucina-8B/genética , Transducción de Señal , Corteza Somatosensorial/metabolismo
8.
Zhen Ci Yan Jiu ; 49(4): 341-348, 2024 Apr 25.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-38649201

RESUMEN

OBJECTIVES: To study the regularity of central response to thermal needle stimulation of "Zusanli" (ST36) at different temperature, and to analyze the temperature difference of central responses. METHODS: Six male C57BL/6j adult mice were used in the present study. For observing activities of neurons in the hindlimb region of left primary somatosensory cortex (S1HL, A/P=0.46 mm, M/L=1.32 mm, D/V=-0.14 mm) by using a fast high-resolution miniature two-photon microscopy (FHIRM-TPM), the mice were anesthetized with 3% isoflurane (inhalation), with its head fixed in a stereotaxic apparatus, then, adeno-associated virus (AAV-hSyn-GCaMP6f-WPRE-hGHpA, for showing intracellular calcium transients in neurons transfected) was injected into the left S1HL region using a micro-syringe after scalp surgical operation. The mice's right ST36 were stimulated using internal thermal needles with the temperature being 43 ℃, or 45 ℃, or 47 ℃, separately. Image J software and MATLAB 2020b software were used to process the image data of neuronal calcium activity (Ca2+ signaling) in the left S1HL region, including the instant maximum calcium peak value (ΔF/F) in 2 s, instant calcium spike frequency in 2 s, short-term calcium peak value (ΔF/F) in 3.5 min, short-term calcium spike frequency in 3.5 min, calcium peak duration in 3.5 min, maximum calcium peak value (ΔF/F) at the 1st , 2nd and 3rd min, and calcium spike frequency at the 1st, 2nd and 3rd min after thermal needle stimulation. RESULTS: In comparison with the normal temperature needle stimulation, the instant intracellular maximum calcium peak value, instant calcium spike frequency, short-term maximum calcium peak value, short-term calcium spike frequency, and calcium peak duration of S1HL neurons in response to 43 ℃, 45 ℃ and 47 ℃ internal thermal needle stimulation of ST36 were significantly increased (P<0.001, P<0.01). Comparison among the 43 ℃, 45 ℃ and 47 ℃ thermal needle stimulation showed that the 45 ℃ thermal needle stimulation was obviously superior to 43 ℃ and 47 ℃ thermal needle stimulation in increasing instant calcium spike frequency, short-term calcium spike frequency and calcium peak duration of S1HL neurons (P<0.001, P<0.01). The 47 ℃ thermal needle stimulation was stronger than 43 ℃ and 45 ℃ thermal needle stimulation in increasing the instant maximum calcium peak value (P<0.001). The maximum calcium peak value was apparently higher (P<0.001) at the 2nd min than that at the 1st and 3rd min after 43 ℃, 45 ℃ and 47 ℃ thermal needle stimulation. No significant differences were found in the short-term maximum calcium peak value among the 3 thermal needle stimulation and in the calcium spike frequency among the 3 time points after 43 ℃, 45 ℃ and 47 ℃ thermal needle stimulation. CONCLUSIONS: S1HL neurons respond to all 43 ℃, 45 ℃ and 47 ℃ thermal needle stimulation of ST36 in mice, while more actively to 45 ℃ thermal needle stimulation.


Asunto(s)
Miembro Posterior , Ratones Endogámicos C57BL , Neuronas , Corteza Somatosensorial , Animales , Ratones , Masculino , Neuronas/fisiología , Corteza Somatosensorial/fisiología , Corteza Somatosensorial/metabolismo , Puntos de Acupuntura , Humanos , Agujas , Calor , Temperatura
9.
Neurobiol Dis ; 183: 106170, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37257662

RESUMEN

Cytoplasmic dynein is an important intracellular motor protein that plays an important role in neuronal growth, axonal polarity formation, dendritic differentiation, and dendritic spine development among others. The intermediate chain of dynein, encoded by Dync1i1, plays a vital role in the dynein complex. Therefore, we assessed the behavioral and related neuronal activities in mice with dync1i1 gene knockout. Neuronal activities in primary somatosensory cortex were recorded by in vivo electrophysiology and manipulated by optogenetic and chemogenetics. Nociception of mechanical, thermal, and cold pain in Dync1i1-/- mice were impaired. The activities of parvalbumin (PV) interneurons and gamma oscillation in primary somatosensory were also impaired when exposed to mechanical nociceptive stimulation. This neuronal dysfunction was rescued by optogenetic activation of PV neurons in Dync1i1-/- mice, and mimicked by suppressing PV neurons using chemogenetics in WT mice. Impaired pain sensations in Dync1i1-/- mice were correlated with impaired gamma oscillations due to a loss of interneurons, especially the PV type. This genotype-driven approach revealed an association between impaired pain sensation and cytoplasmic dynein complex.


Asunto(s)
Parvalbúminas , Corteza Somatosensorial , Ratones , Animales , Parvalbúminas/metabolismo , Corteza Somatosensorial/metabolismo , Dineínas Citoplasmáticas/metabolismo , Dineínas/metabolismo , Interneuronas/metabolismo , Umbral del Dolor
10.
Brain Res ; 1800: 148179, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36511312

RESUMEN

The tish (telencephalic internal structural heterotopia) rat is a naturally occurring and unique model of a malformation of cortical development (MCD) arising from a sponeantous mutation in the Eml1 gene. Tish rats are characterized by a macroscopic bilateral heterotopic dysplastic cortex (HDCx) and an overlaying, intact normotopic neocortex (NNCx). These two cortices are functional and have been reported to innervate and establish connections with subcortical regions including the thalamus, resulting in a dual-cortical representation. Additionally, impaired GABAergic neurotransmission and early-onset spike wave discharge bursts have been reported in developing tish rats. Perineuronal nets (PNNs) are specialized extraceullar matrix structures that predominately surround and stabilize parvalbumin-positive (PV+) GABAergic interneurons and are essential components of the neural landscape. Here, we report a significant reduction in the average number of WFA+-PNNs in the normotopic somatosensory cortex (NSSCx) of the tish rat at two developmental time points, P16 and P35, corresponding to a decrease in the number of PV+ interneurons ensheathed by a PNN in the NSSCx. Compared with control animals, PNN expression was partially, but significantly restored following treatment with insulin-like growth factor 1 (IGF-1). These data suggest that the 'dual cortical representation' in the setting of an MCD reduces the cortical activation necessary for proper PNN expression likely contributing to the impairments in GABAergic neurotransmission and network excitability previously identified in the tish rat.


Asunto(s)
Neocórtex , Corteza Somatosensorial , Ratas , Animales , Corteza Somatosensorial/metabolismo , Matriz Extracelular/metabolismo , Neocórtex/metabolismo , Transmisión Sináptica , Interneuronas/metabolismo , Parvalbúminas/metabolismo
11.
Pharmacol Res ; 178: 106144, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35304260

RESUMEN

The glutamate delta family of receptors is composed of GluD1 and GluD2 and serve as synaptic organizers. We have previously demonstrated several autism-like molecular and behavioral phenotypes including an increase in dendritic spines in GluD1 knockout mice. Based on previous reports we evaluated whether disruption of autophagy mechanisms may account for these phenotypes. Mouse model with conditional deletion of GluD1 from excitatory neurons in the corticolimbic regions was utilized. GluD1 loss led to overactive Akt-mTOR pathway, higher p62 and a lower LC3-II/LC3-I ratio in the somatosensory cortex suggesting reduced autophagy. Excitatory elements were increased in number but had immature phenotype based on puncta size, lower AMPA subunit GluA1 expression and impaired development switch from predominantly GluN2B to mixed GluN2A/GluN2B subunit expression. Overactive Akt-mTOR signaling and impaired autophagy was also observed in dorsal striatum upon conditional ablation of GluD1 and in the prefrontal cortex and hippocampus in constitutive knockout. Finally, cognitive deficits in novel object recognition test and fear conditioning were observed in mice with conditional ablation of GluD1 from the corticolimbic regions. Together, these results demonstrate a novel function of GluD1 in the regulation of autophagy pathway which may underlie autism phenotypes and is relevant to the genetic association of GluD1 coding, GRID1 gene with autism and other developmental disorders.


Asunto(s)
Ácido Glutámico , Receptores de Glutamato , Corteza Somatosensorial , Animales , Autofagia , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Corteza Somatosensorial/metabolismo , Sinapsis/fisiología , Serina-Treonina Quinasas TOR/metabolismo
12.
Proc Natl Acad Sci U S A ; 118(52)2021 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-34930843

RESUMEN

Immediate-early gene (IEG) expression has been used to identify small neural ensembles linked to a particular experience, based on the principle that a selective subset of activated neurons will encode specific memories or behavioral responses. The majority of these studies have focused on "engrams" in higher-order brain areas where more abstract or convergent sensory information is represented, such as the hippocampus, prefrontal cortex, or amygdala. In primary sensory cortex, IEG expression can label neurons that are responsive to specific sensory stimuli, but experience-dependent shaping of neural ensembles marked by IEG expression has not been demonstrated. Here, we use a fosGFP transgenic mouse to longitudinally monitor in vivo expression of the activity-dependent gene c-fos in superficial layers (L2/3) of primary somatosensory cortex (S1) during a whisker-dependent learning task. We find that sensory association training does not detectably alter fosGFP expression in L2/3 neurons. Although training broadly enhances thalamocortical synaptic strength in pyramidal neurons, we find that synapses onto fosGFP+ neurons are not selectively increased by training; rather, synaptic strengthening is concentrated in fosGFP- neurons. Taken together, these data indicate that expression of the IEG reporter fosGFP does not facilitate identification of a learning-specific engram in L2/3 in barrel cortex during whisker-dependent sensory association learning.


Asunto(s)
Aprendizaje por Asociación/fisiología , Memoria/fisiología , Plasticidad Neuronal , Proteínas Proto-Oncogénicas c-fos , Corteza Somatosensorial , Animales , Femenino , Genes Inmediatos-Precoces/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Transgénicos , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/fisiología
13.
Neurosci Lett ; 765: 136268, 2021 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-34571088

RESUMEN

Absence epilepsy is classified as a childhood generalized epilepsy syndrome with distinctive electroencephalographic patterns. The Wistar Albino Glaxo originating from Rijswijk (WAG/Rij) strain is a very well validated animal model of absence epilepsy that also shows behavioral deficits. In addition to the gastrointestinal system, VIP is highly expressed throughout numerous brain regions, and it plays crucial roles as a neurotransmitter and as a neuromodulatory, neurotrophic and neuroprotective factor in both the central and peripheral nervous systems. In this study, adult WAG/Rij rats were divided into two groups (n = 10): a group that was administered VIP (25 ng/kg i.p.) every 2 days for 15 days and an age-matched control group that was administered physiological saline. Electrical brain activity and behavior (depressive- like behavior, learning and memory and anxiety) were investigated in both groups. In addition, the extracellular concentrations of GABA and glutamate and the GABA/glutamate ratio were measured by high-performance liquid chromatography in microdialysate samples collected from the somatosensorial cortex of WAG/Rij rats. Our results demonstrated that VIP treatment significantly suppressed the total duration and number of spike wave discharges in WAG/Rij rats. However, VIP had no significant effect on behavior. VIP increased the extracellular concentration of GABA and the GABA/glutamate ratio in the somatosensory cortex. In conclusion, VIP has suppressive effects on absence seizures, possibly by increasing the GABA concentration and inducing the transformation of glutamate to GABA in the somatosensory cortex of WAG/Rij rats.


Asunto(s)
Epilepsia Tipo Ausencia/metabolismo , Convulsiones/metabolismo , Corteza Somatosensorial/metabolismo , Péptido Intestinal Vasoactivo/farmacología , Ácido gamma-Aminobutírico/metabolismo , Animales , Femenino , Ratas , Ratas Wistar , Corteza Somatosensorial/efectos de los fármacos
14.
Mol Neurobiol ; 58(9): 4727-4744, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34165684

RESUMEN

Neuronal regeneration is crucial for maintaining intact neural interactions for perpetuation of cognitive and emotional functioning. The NRG1-ErbB receptor signaling is a key pathway for regeneration in adult brain and also associated with learning and mood stabilization by modulating synaptic transmission. Extreme glycemic stress is known to affect NRG1-ErbB-mediated regeneration in brain; yet, it remains unclear how the ErbB receptor subtypes are differentially affected due to such metabolic variations. Here, we assessed the alterations in NRG1, ErbB receptor subtypes to study the regenerative potential, both in rodents as well as in neuronal and glial cell models of hyperglycemia and hypoglycemic insults during hyperglycemia. The pro-oxidant and anti-oxidant status leading to degenerative changes in brain regions were determined. The spatial memory and anxiogenic behaviour of experimental rodents were tested using 'T' maze and Elevated Plus Maze. Our data revealed that the extreme glycemic discrepancies during diabetes and recurrent hypoglycemia lead to altered expression of NRG1, ErbB receptor subtypes, Syntaxin1 and Olig1 that shows association with impaired regeneration, synaptic dysfunction, demyelination, cognitive deficits and anxiety.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cognición/fisiología , Diabetes Mellitus Experimental/metabolismo , Receptores ErbB/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neurregulina-1/metabolismo , Animales , Glucemia , Cuerpo Calloso/metabolismo , Hipocampo/metabolismo , Masculino , Regeneración Nerviosa/fisiología , Neuronas/metabolismo , Ratas , Ratas Wistar , Corteza Somatosensorial/metabolismo
15.
Int J Mol Sci ; 22(10)2021 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-34065959

RESUMEN

Brain tissue may be especially sensitive to electromagnetic phenomena provoking signs of neural stress in cerebral activity. Fifty-four adult female Sprague-Dawley rats underwent ELISA and immunohistochemistry testing of four relevant anatomical areas of the cerebrum to measure biomarkers indicating induction of heat shock protein 70 (HSP-70), glucocorticoid receptors (GCR) or glial fibrillary acidic protein (GFAP) after single or repeated exposure to 2.45 GHz radiation in the experimental set-up. Neither radiation regime caused tissue heating, so thermal effects can be ruled out. A progressive decrease in GCR and HSP-70 was observed after acute or repeated irradiation in the somatosensory cortex, hypothalamus and hippocampus. In the limbic cortex; however, values for both biomarkers were significantly higher after repeated exposure to irradiation when compared to control animals. GFAP values in brain tissue after irradiation were not significantly different or were even lower than those of nonirradiated animals in all brain regions studied. Our results suggest that repeated exposure to 2.45 GHz elicited GCR/HSP-70 dysregulation in the brain, triggering a state of stress that could decrease tissue anti-inflammatory action without favoring glial proliferation and make the nervous system more vulnerable.


Asunto(s)
Cerebro/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Biomarcadores/metabolismo , Cerebro/efectos de la radiación , Femenino , Regulación de la Expresión Génica/efectos de la radiación , Hipocampo/metabolismo , Hipocampo/efectos de la radiación , Hipotálamo/metabolismo , Hipotálamo/efectos de la radiación , Ratas , Ratas Sprague-Dawley , Corteza Somatosensorial/metabolismo , Corteza Somatosensorial/efectos de la radiación
16.
Exp Neurol ; 342: 113734, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33945789

RESUMEN

Disruptions to either sulfate supply or sulfation enzymes can affect brain development and have long-lasting effects on brain function, yet our understanding of the molecular mechanisms governing this are incomplete. Perineuronal nets (PNNs) are highly sulfated, specialized extracellular matrix structures that regulate the maturation of synaptic connections and neuronal plasticity. We have previously shown that mice heterozygous for the brain sulfate transporter Slc13a4 have abnormal social interactions, memory, exploratory behaviors, stress and anxiety of postnatal origin, pointing to potential deficits in PNN biology, and implicate SLC13A4 as a critical factor required for regulating normal synaptic connectivity and function. Here, we sought to investigate aberrant PNN formation as a potential mechanism contributing to the functional deficits displayed by Slc13a4+/- mice. Following social interactions, we reveal reduced neuronal activation in the somatosensory cortex of Slc13a4+/- mice, and altered inhibitory and excitatory postsynaptic currents. In line with this, we found a reduction in parvalbumin-expressing neurons decorated with PNNs, as well as reduced expression of markers for PNN maturation. Finally, we reveal that postnatal administration of N-acetylcysteine prevented PNN abnormalities from manifesting in Slc13a4+/- adult animals. Collectively, these data highlight a central role for postnatal SLC13A4 in normal PNN formation, circuit function and subsequent animal behavior.


Asunto(s)
Acetilcisteína/administración & dosificación , Red Nerviosa/efectos de los fármacos , Red Nerviosa/metabolismo , Nervios Periféricos/efectos de los fármacos , Nervios Periféricos/metabolismo , Transportadores de Sulfato/metabolismo , Simportadores/metabolismo , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Femenino , Depuradores de Radicales Libres/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/metabolismo , Transportadores de Sulfato/genética , Simportadores/genética
17.
JCI Insight ; 6(5)2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33561011

RESUMEN

BACKGROUNDType 1 diabetes (T1D) is a risk factor for dementia and structural brain changes. It remains to be determined whether transient insulin deprivation that frequently occurs in insulin-treated individuals with T1D alters brain function.METHODSWe therefore performed functional and structural magnetic resonance imaging, magnetic resonance spectroscopy, and neuropsychological testing at baseline and following 5.4 ± 0.6 hours of insulin deprivation in 14 individuals with T1D and compared results with those from 14 age-, sex-, and BMI-matched nondiabetic (ND) participants with no interventions.RESULTSInsulin deprivation in T1D increased blood glucose, and ß-hydroxybutyrate, while reducing bicarbonate levels. Participants with T1D showed lower baseline brain N-acetyl aspartate and myo-inositol levels but higher cortical fractional anisotropy, suggesting unhealthy neurons and brain microstructure. Although cognitive functions did not differ between participants with T1D and ND participants at baseline, significant changes in fine motor speed as well as attention and short-term memory occurred following insulin deprivation in participants with T1D. Insulin deprivation also reduced brain adenosine triphosphate levels and altered the phosphocreatine/adenosine triphosphate ratio. Baseline differences in functional connectivity in brain regions between participants with T1D and ND participants were noted, and on insulin deprivation further alterations in functional connectivity between regions, especially cortical and hippocampus-caudate regions, were observed. These alterations in functional connectivity correlated to brain metabolites and to changes in cognition.CONCLUSIONTransient insulin deprivation therefore caused alterations in executive aspects of cognitive function concurrent with functional connectivity between memory regions and the sensory cortex. These findings have important clinical implications, as many patients with T1D inadvertently have periods of transient insulin deprivation.TRIAL REGISTRATIONClinicalTrials.gov NCT03392441.FUNDINGClinical and Translational Science Award (UL1 TR002377) from the National Center for Advancing Translational Science; NIH grants (R21 AG60139 and R01 AG62859); the Mayo Foundation.


Asunto(s)
Disfunción Cognitiva/metabolismo , Diabetes Mellitus Tipo 1 , Insulina/metabolismo , Memoria , Corteza Somatosensorial/metabolismo , Adulto , Glucemia/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/fisiopatología , Femenino , Humanos , Masculino , Proyectos Piloto , Ciencia Traslacional Biomédica , Adulto Joven
18.
Cell Rep ; 34(8): 108774, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33626343

RESUMEN

Extensive hierarchical yet highly reciprocal interactions among cortical areas are fundamental for information processing. However, connectivity rules governing the specificity of such corticocortical connections, and top-down feedback projections in particular, are poorly understood. We analyze synaptic strength from functionally relevant brain areas to diverse neuronal types in the primary somatosensory cortex (S1). Long-range projections from different areas preferentially engage specific sets of GABAergic neurons in S1. Projections from other somatosensory cortices strongly recruit parvalbumin (PV)-positive GABAergic neurons and lead to PV neuron-mediated feedforward inhibition of pyramidal neurons in S1. In contrast, inputs from whisker-related primary motor cortex are biased to vasoactive intestinal peptide (VIP)-positive GABAergic neurons and potentially result in VIP neuron-mediated disinhibition. Regardless of the input areas, somatostatin-positive neurons receive relatively weak long-range inputs. Computational analyses suggest that a characteristic combination of synaptic inputs to different GABAergic IN types in S1 represents a specific long-range input area.


Asunto(s)
Neuronas GABAérgicas/metabolismo , Interneuronas/metabolismo , Inhibición Neural , Células Piramidales/metabolismo , Corteza Somatosensorial/metabolismo , Transmisión Sináptica , Vibrisas/inervación , Ácido gamma-Aminobutírico/metabolismo , Animales , Femenino , Masculino , Ratones Transgénicos , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Técnicas de Trazados de Vías Neuroanatómicas , Parvalbúminas/metabolismo , Corteza Somatosensorial/citología , Péptido Intestinal Vasoactivo/metabolismo
19.
PLoS Biol ; 18(2): e3000613, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32027647

RESUMEN

Cortical interneurons expressing vasoactive intestinal polypeptide (VIP) and choline acetyltransferase (ChAT) are sparsely distributed throughout the neocortex, constituting only 0.5% of its neuronal population. The co-expression of VIP and ChAT suggests that these VIP/ChAT interneurons (VChIs) can release both γ-aminobutyric acid (GABA) and acetylcholine (ACh). In vitro physiological studies quantified the response properties and local connectivity patterns of the VChIs; however, the function of VChIs has not been explored in vivo. To study the role of VChIs in cortical network dynamics and their long-range connectivity pattern, we used in vivo electrophysiology and rabies virus tracing in the barrel cortex of mice. We found that VChIs have a low spontaneous spiking rate (approximately 1 spike/s) in the barrel cortex of anesthetized mice; nevertheless, they responded with higher fidelity to whisker stimulation than the neighboring layer 2/3 pyramidal neurons (Pyrs). Analysis of long-range inputs to VChIs with monosynaptic rabies virus tracing revealed that direct thalamic projections are a significant input source to these cells. Optogenetic activation of VChIs in the barrel cortex of awake mice suppresses the sensory responses of excitatory neurons in intermediate amplitudes of whisker deflections while increasing the evoked spike latency. The effect of VChI activation on the response was similar for both high-whisking (HW) and low-whisking (LW) conditions. Our findings demonstrate that, despite their sparsity, VChIs can effectively modulate sensory processing in the cortical microcircuit.


Asunto(s)
Colina O-Acetiltransferasa/metabolismo , Interneuronas/fisiología , Corteza Somatosensorial/citología , Péptido Intestinal Vasoactivo/metabolismo , Animales , Colina O-Acetiltransferasa/genética , Potenciales Evocados , Potenciales Postsinápticos Inhibidores , Integrasas/genética , Interneuronas/metabolismo , Ratones , Ratones Transgénicos , Vías Nerviosas , Neuronas/metabolismo , Neuronas/fisiología , Optogenética , Corteza Somatosensorial/metabolismo , Péptido Intestinal Vasoactivo/genética , Núcleos Talámicos Ventrales/metabolismo , Vibrisas
20.
J Neurosci Res ; 98(3): 481-490, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31670406

RESUMEN

Luminopsins (LMOs) are chimeric proteins consisting of a luciferase fused to an opsin that provide control of neuronal activity, allowing for less cumbersome and less invasive optogenetic manipulation. It was previously shown that both an external light source and the luciferase substrate, coelenterazine (CTZ), could modulate activity of LMO-expressing neurons, although the magnitudes of the photoresponses remained subpar. In this study, we created an enhanced iteration of the excitatory luminopsin LMO3, termed eLMO3, that has improved membrane targeting due to the insertion of a Golgi trafficking signal sequence. In cortical neurons in culture, the expression of eLMO3 resulted in significant reductions in the formation of intracellular aggregates, as well as in a significant increase in total photocurrents. Furthermore, we corroborated the findings with injections of adeno-associated viral vectors into the deep layers of the somatosensory cortex (the barrel cortex) of male mice. We observed greatly reduced numbers of intracellular puncta in eLMO3-expressing cortical neurons compared to those expressing the original LMO3. Finally, we quantified CTZ-driven behavior, namely whisker-touching behavior, in male mice with LMO3 expression in the barrel cortex. After CTZ administration, mice with eLMO3 displayed significantly longer whisker responses than mice with LMO3. In summary, we have engineered the superior LMO by resolving membrane trafficking defects, and we demonstrated improved membrane targeting, greater photocurrents, and greater functional responses to stimulate with CTZ.


Asunto(s)
Imidazoles/administración & dosificación , Luciferasas/metabolismo , Sustancias Luminiscentes/administración & dosificación , Neuronas/metabolismo , Opsinas/metabolismo , Optogenética/métodos , Transporte de Proteínas , Pirazinas/administración & dosificación , Corteza Somatosensorial/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Femenino , Mediciones Luminiscentes , Masculino , Potenciales de la Membrana , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Cultivo Primario de Células , Corteza Somatosensorial/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA