The cholinergic system extensively innervates mature prefrontal cortex (PFC) and is critical to sustain cortical activation during wakefulness and REM sleep. The implication of nicotinic receptors (nAChRs) in these processes is increasingly recognized. Moreover, nAChRs regulate developing cortical circuits during the first postnatal weeks. First, we studied mice expressing β2-V287L, a mutant subunit of the nAChR linked to Autosomal Dominant Nocturnal Frontal Lobe Epilepsy (ADNFLE). β2-V287L exerts the epileptogenic action by altering the synaptic stabilization during sensitive phases of network maturation, since its expression during the first two postnatal weeks is necessary to develop the epileptic phenotype. The peak of postnatal expression of nAChRs coincides with the excitatory to inhibitory GABAergic switch, which is determined by the progressive decrease in the ratio between the expression of the Na+/K+/Cl- cotransporter-1 (NKCC1) and the K+/Cl- cotransporter-2 (KCC2). How these transporters are regulated in PFC and thalamus is largely unknown. We first studied the distribution of NKCC1 and KCC2 in developing wild-type (WT) mice. The amount of NKCC1 and KCC2 in PFC and somatosensory cortex progressively increased during the first two postnatal weeks. NKCC1 was found in neurons as well as astrocytes. KCC2 was mainly localized in neuronal somata at birth, and subsequently migrated to the somatodendritic membranes. Next, in mice expressing β2-V287L, the KCC2 amount in PFC layer V was lower than in the control littermates at postnatal day 8 (P8), but reached higher amounts by P60. Consistently, the time course of the GABAergic switch was delayed in PFC layer V of mice carrying β2-V287L, as measured by perforated patch method. Irrespective of genotype, NKCC1 and KCC2 were highly expressed in the neuropil of thalamic nuclei at birth. Their amount remained high in the adult sensory nuclei, whereas a significant decrease of KCC2 was observed in the reticular nucleus by P40. Such a decrease was more pronounced in mice expressing β2-V287L. Our results indicate that β2-containing nAChRs interact with KCC2 during synaptogenesis as well as in mature circuits, which may contribute to the pathogenesis of ADNFLE. Second, we explored some aspects of the complex interplay between the main neurotransmitters involved in cortical arousal, i. e. acetylcholine (ACh), norepinephrine (NE) and orexin A (OrxA), in order to characterize their combined effect on the PFC layer V circuit. In particular, by patch-clamp methods, we investigated how these neurotransmitters regulate glutamate release onto pyramidal neurons in PFC layer V of adult wild-type mice. Tonic administration of ACh stimulated glutamate release, mainly through α4-containing nAChRs. Moreover, OrxA increased the glutamate release onto pyramidal neurons, through Orx Receptor 1 (OrxR1). Low concentrations of NE (10 nM) were also effective in increasing glutamatergic release. We conclude that these neuromodulators cooperate in regulating glutamatergic transmission in PFC. Such interaction may be crucial in the cellular mechanisms underlying the cognitive and executive functions in this area, in normal and pathological conditions, which are often accompanied by alterations in the theta rhythms, largely determined by pyramidal neurons dynamics. These results may yield new insights into the complex regulation of the PFC microcircuit and lay the basis for further investigations on the pathogenic mechanisms responsible of altered cortical activity in sleep-related disorders such as frontal epilepsy and narcolepsy with cataplexy.

Il sistema colinergico proietta in modo diffuso alla corteccia prefrontale (PFC) matura e sostiene l’attivazione corticale durante la veglia e il sonno REM. Il ruolo dei recettori nicotinici (nAChRs) nel mediare questi processi è sempre più riconosciuto. Inoltre, i nAChRs regolano anche lo sviluppo dei circuiti corticali durante le prime settimane di vita. Innanzitutto abbiamo studiato topi esprimenti β2-V287L, una subunità mutata del nAChR associata all’epilessia notturna autosomica dominante del lobo frontale (ADNFLE). β2-V287L svolge la sua azione epilettogenica alterando la stabilizzazione sinaptica durante le fasi critiche della maturazione circuitale, infatti, la sua espressione durante le prime due settimane di vita è necessaria per lo sviluppo del fenotipo epilettico nel topo adulto. Questa fase critica, inoltre, coincide con lo switch del GABA da eccitatorio a inibitorio, determinato dalla diminuzione progressiva del rapporto tra l’espressione del cotrasportatore-1 Na+/K+/Cl- (NKCC1) e del cotrasportatore-2 (KCC2) K+/Cl-. La regolazione di questi trasportatori nella PFC e nel talamo è ancora largamente sconosciuta. Abbiamo indagato la distribuzione di NKCC1 e KCC2 in topi wild-type (WT) durante lo sviluppo. I livelli di entrambi i trasportatori aumentano progressivamente durante le prime due settimane di vita sia nella PFC che nella corteccia somato-sensoriale. Alla nascita, KCC2 è localizzato principalmente nei corpi cellulari neuronali e in seguito migra verso le membrane somato-dendritiche. In topi esprimenti β2-V287L, i livelli di KCC2 nel V strato della PFC a 8 giorni (P8) sono inferiori rispetto ai topi controllo, ma raggiungono livelli di espressione maggiori a P60. Anche la tempistica dello switch del GABA, misurato tramite patch perforato, risulta ritardato nel V strato della PFC nei topi mutati. Al momento della nascita NKCC1 e KCC2 sono molto espressi nel neuropilo dei nuclei talamici, indipendentemente dal genotipo. Il loro livello d’espressione rimane alto nei nuclei sensoriali nell’adulto, mentre KCC2 diminuisce significativamente nel nucleo reticolare a P40. Tale riduzione è più pronunciata nei topi esprimenti β2-V287L. Questi risultati indicano che i nAChRs contenenti la subunità β2 interagiscono con KCC2 durante la sinaptogenesi e questo può contribuire alla patogenesi dell’ADNFLE. In secondo luogo, abbiamo esplorato alcuni aspetti della complessa interazione tra i principali neurotrasmettitori che partecipano all’attivazione corticale, ovvero acetilcolina (ACh), noradrenalina (NE) e oressina A (OrxA), allo scopo di caratterizzare il loro effetto sull’eccitabilità del V strato della PFC. In particolare, tramite patch-clamp in topi WT adulti, abbiamo indagato il loro ruolo nel modulare il rilascio di glutammato sui neuroni piramidali del V strato. La somministrazione tonica di ACh stimola il rilascio di glutammato, principalmente attraverso i nAChRs contenenti la subunità α4. Inoltre, l’OrxA è in grado di stimolare il rilascio di glutammato sui neuroni piramidali attivando i recettori 1 dell’Orx (OrxR1). Anche basse concentrazioni di NE (10 nM) sono efficaci nell’aumentare il rilascio di glutammato. Possiamo concludere che questi neuromodulatori cooperano nel regolare la trasmissione glutammatergica nella PFC. Tale interazione potrebbe essere cruciale nella regolazione dei meccanismi cellulari alla base delle funzioni cognitive ed esecutive svolte da questa regione. Questo avviene sia in condizioni normali che patologiche, le quali sono spesso accompagnate da alterazioni dei ritmi theta, generati da dinamiche proprie dei neuroni piramidali. Questi risultati possono aiutare a comprendere la complessa regolazione dei microcircuiti della PFC, e pongono le basi per ulteriori indagini sui meccanismi patogenetici responsabili di anomalie corticali, tipiche di alcuni disordini legati al sonno, come l’epilessia frontale e la narcolessia con cataplessia.

(2018). Heteromeric nicotinic receptors regulate developing and mature prefrontal circuits: interaction with other neuromodulators, and implications for sleep-related hypermotor epilepsy.. (Tesi di dottorato, Università degli Studi di Milano-Bicocca, 2018).

Heteromeric nicotinic receptors regulate developing and mature prefrontal circuits: interaction with other neuromodulators, and implications for sleep-related hypermotor epilepsy.

COATTI, AURORA
2018

Abstract

The cholinergic system extensively innervates mature prefrontal cortex (PFC) and is critical to sustain cortical activation during wakefulness and REM sleep. The implication of nicotinic receptors (nAChRs) in these processes is increasingly recognized. Moreover, nAChRs regulate developing cortical circuits during the first postnatal weeks. First, we studied mice expressing β2-V287L, a mutant subunit of the nAChR linked to Autosomal Dominant Nocturnal Frontal Lobe Epilepsy (ADNFLE). β2-V287L exerts the epileptogenic action by altering the synaptic stabilization during sensitive phases of network maturation, since its expression during the first two postnatal weeks is necessary to develop the epileptic phenotype. The peak of postnatal expression of nAChRs coincides with the excitatory to inhibitory GABAergic switch, which is determined by the progressive decrease in the ratio between the expression of the Na+/K+/Cl- cotransporter-1 (NKCC1) and the K+/Cl- cotransporter-2 (KCC2). How these transporters are regulated in PFC and thalamus is largely unknown. We first studied the distribution of NKCC1 and KCC2 in developing wild-type (WT) mice. The amount of NKCC1 and KCC2 in PFC and somatosensory cortex progressively increased during the first two postnatal weeks. NKCC1 was found in neurons as well as astrocytes. KCC2 was mainly localized in neuronal somata at birth, and subsequently migrated to the somatodendritic membranes. Next, in mice expressing β2-V287L, the KCC2 amount in PFC layer V was lower than in the control littermates at postnatal day 8 (P8), but reached higher amounts by P60. Consistently, the time course of the GABAergic switch was delayed in PFC layer V of mice carrying β2-V287L, as measured by perforated patch method. Irrespective of genotype, NKCC1 and KCC2 were highly expressed in the neuropil of thalamic nuclei at birth. Their amount remained high in the adult sensory nuclei, whereas a significant decrease of KCC2 was observed in the reticular nucleus by P40. Such a decrease was more pronounced in mice expressing β2-V287L. Our results indicate that β2-containing nAChRs interact with KCC2 during synaptogenesis as well as in mature circuits, which may contribute to the pathogenesis of ADNFLE. Second, we explored some aspects of the complex interplay between the main neurotransmitters involved in cortical arousal, i. e. acetylcholine (ACh), norepinephrine (NE) and orexin A (OrxA), in order to characterize their combined effect on the PFC layer V circuit. In particular, by patch-clamp methods, we investigated how these neurotransmitters regulate glutamate release onto pyramidal neurons in PFC layer V of adult wild-type mice. Tonic administration of ACh stimulated glutamate release, mainly through α4-containing nAChRs. Moreover, OrxA increased the glutamate release onto pyramidal neurons, through Orx Receptor 1 (OrxR1). Low concentrations of NE (10 nM) were also effective in increasing glutamatergic release. We conclude that these neuromodulators cooperate in regulating glutamatergic transmission in PFC. Such interaction may be crucial in the cellular mechanisms underlying the cognitive and executive functions in this area, in normal and pathological conditions, which are often accompanied by alterations in the theta rhythms, largely determined by pyramidal neurons dynamics. These results may yield new insights into the complex regulation of the PFC microcircuit and lay the basis for further investigations on the pathogenic mechanisms responsible of altered cortical activity in sleep-related disorders such as frontal epilepsy and narcolepsy with cataplexy.
BECCHETTI, ANDREA
AMADEO, ALIDA
nAChR;; β2-V287L;; Fr2;; neuromodulation;; EPSC
nAChR;; β2-V287L;; Fr2;; neuromodulation;; EPSC
BIO/09 - FISIOLOGIA
English
16-mar-2018
MEDICINA TRASLAZIONALE E MOLECOLARE - DIMET - 76R
30
2016/2017
open
(2018). Heteromeric nicotinic receptors regulate developing and mature prefrontal circuits: interaction with other neuromodulators, and implications for sleep-related hypermotor epilepsy.. (Tesi di dottorato, Università degli Studi di Milano-Bicocca, 2018).
File in questo prodotto:
File Dimensione Formato  
phd_unimib_718138.pdf

Accesso Aperto

Descrizione: tesi di dottorato
Tipologia di allegato: Doctoral thesis
Dimensione 4.11 MB
Formato Adobe PDF
4.11 MB Adobe PDF Visualizza/Apri

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/10281/199023
Citazioni
  • Scopus ND
  • ???jsp.display-item.citation.isi??? ND
Social impact