Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.853
Filter
1.
Cell Commun Signal ; 22(1): 361, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39010083

ABSTRACT

BACKGROUND: Breast cancer is one of the most lethal cancers in women. Despite significant advances in the diagnosis and treatment of breast cancer, many patients still succumb to this disease, and thus, novel effective treatments are urgently needed. Natural product coumarin has been broadly investigated since it reveals various biological properties in the medicinal field. Accumulating evidence indicates that histone deacetylase inhibitors (HDACIs) are promising novel anti-breast cancer agents. However, most current HDACIs exhibit only moderate effects against solid tumors and are associated with severe side effects. Thus, to develop more effective HDACIs for breast cancer therapy, hydroxamate of HDACIs was linked to coumarin core, and coumarin-hydroxamate hybrids were designed and synthesized. METHODS: A substituted coumarin moiety was incorporated into the classic hydroxamate HDACIs by the pharmacophore fusion strategy. ZN444B was identified by using the HDACI screening kit and cell viability assay. Molecular docking was performed to explore the binding mode of ZN444B with HDAC1. Western blot, immunofluorescent staining, cell viability, colony formation and cell migration and flow cytometry assays were used to analyze the anti-breast cancer effects of ZN444B in vitro. Orthotopic studies in mouse models were applied for preclinical evaluation of efficacy and toxicity in vivo. Proteomic analysis, dual-luciferase reporter assay, chromatin immunoprecipitation, co-immunoprecipitation, immunofluorescent staining assays along with immunohistochemical (IHC) analysis were used to elucidate the molecular basis of the actions of ZN444B. RESULTS: We synthesized and identified a novel coumarin-hydroxamate conjugate, ZN444B which possesses promising anti-breast cancer activity both in vitro and in vivo. A molecular docking model showed that ZN444B binds to HDAC1 with high affinity. Further mechanistic studies revealed that ZN444B specifically decreases FOS-like antigen 2 (FOSL2) mRNA levels by inhibiting the deacetylase activity of HDAC1 on Sp1 at K703 and abrogates the binding ability of Sp1 to the FOSL2 promoter. Furthermore, FOSL2 expression positively correlates with breast cancer progression and metastasis. Silencing FOSL2 expression decreases the sensitivity of breast cancer cells to ZN444B treatment. In addition, ZN444B shows no systemic toxicity in mice. CONCLUSIONS: Our findings highlight the potential of FOSL2 as a new biomarker and therapeutic target for breast cancer and that targeting the HDAC1-Sp1-FOSL2 signaling axis with ZN444B may be a promising therapeutic strategy for breast cancer.


Subject(s)
Breast Neoplasms , Coumarins , Histone Deacetylase 1 , Hydroxamic Acids , Signal Transduction , Coumarins/chemistry , Coumarins/pharmacology , Humans , Histone Deacetylase 1/metabolism , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Animals , Signal Transduction/drug effects , Hydroxamic Acids/pharmacology , Hydroxamic Acids/chemistry , Hydroxamic Acids/therapeutic use , Sp1 Transcription Factor/metabolism , Mice , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/chemistry , Cell Line, Tumor , Molecular Docking Simulation , Cell Proliferation/drug effects , Mice, Nude , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , Mice, Inbred BALB C , Cell Movement/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Drug Discovery
2.
Mol Brain ; 17(1): 46, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39049098

ABSTRACT

The ventrolateral periaqueductal gray (vlPAG) serves as a central hub for descending pain modulation. It receives upstream projections from the medial prefrontal cortex (mPFC) and the ventrolateral orbitofrontal cortex (vlOFC), and projects downstream to the locus coeruleus (LC) and the rostroventral medulla (RVM). While much research has focused on upstream circuits and the LC-RVM connection, less is known about the PAG-LC circuit and its involvement in neuropathic pain. Here we examined the intrinsic electrophysiological properties of vlPAG-LC projecting neurons in Sham and spared nerve injury (SNI) operated mice. Injection of the retrotracer Cholera Toxin Subunit B (CTB-488) into the LC allowed the identification of LC-projecting neurons in the vlPAG. Electrophysiological recordings from CTB-488 positive cells revealed that both GABAergic and glutamatergic cells that project to the LC exhibited reduced intrinsic excitability after peripheral nerve injury. By contrast, CTB-488 negative cells did not exhibit alterations in firing properties after SNI surgery. An SNI-induced reduction of LC projecting cells was confirmed with c-fos labeling. Hence, SNI induces plasticity changes in the vlPAG that are consistent with a reduction in the descending modulation of pain signals.


Subject(s)
Locus Coeruleus , Mice, Inbred C57BL , Neurons , Periaqueductal Gray , Animals , Periaqueductal Gray/physiopathology , Periaqueductal Gray/physiology , Locus Coeruleus/physiopathology , Locus Coeruleus/pathology , Locus Coeruleus/physiology , Neurons/physiology , Male , Mice , Action Potentials/physiology , Neural Pathways/physiopathology , Neuralgia/physiopathology , Neuralgia/pathology , Peripheral Nerve Injuries/physiopathology , Peripheral Nerve Injuries/pathology , Proto-Oncogene Proteins c-fos/metabolism
3.
Transl Psychiatry ; 14(1): 269, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956048

ABSTRACT

Addiction is a complex behavioral disorder characterized by compulsive drug-seeking and drug use despite harmful consequences. The prefrontal cortex (PFC) plays a crucial role in cocaine addiction, involving decision-making, impulse control, memory, and emotional regulation. The PFC interacts with the brain's reward system, including the ventral tegmental area (VTA) and nucleus accumbens (NAc). The PFC also projects to the lateral habenula (LHb), a brain region critical for encoding negative reward and regulating the reward system. In the current study, we examined the role of PFC-LHb projections in regulating cocaine reward-related behaviors. We found that optogenetic stimulation of the PFC-LHb circuit during cocaine conditioning abolished cocaine preference without causing aversion. In addition, increased c-fos expression in LHb neurons was observed in animals that received optic stimulation during cocaine conditioning, supporting the circuit's involvement in cocaine preference regulation. Molecular analysis in animals that received optic stimulation revealed that cocaine-induced alterations in the expression of GluA1 subunit of AMPA receptor was normalized to saline levels in a region-specific manner. Moreover, GluA1 serine phosphorylation on S845 and S831 were differentially altered in LHb and VTA but not in the PFC. Together these findings highlight the critical role of the PFC-LHb circuit in controlling cocaine reward-related behaviors and shed light on the underlying mechanisms. Understanding this circuit's function may provide valuable insights into addiction and contribute to developing targeted treatments for substance use disorders.


Subject(s)
Cocaine , Habenula , Neurons , Optogenetics , Prefrontal Cortex , Receptors, AMPA , Reward , Animals , Prefrontal Cortex/metabolism , Cocaine/pharmacology , Male , Habenula/metabolism , Neurons/metabolism , Receptors, AMPA/metabolism , Cocaine-Related Disorders/physiopathology , Cocaine-Related Disorders/metabolism , Neural Pathways , Rats , Proto-Oncogene Proteins c-fos/metabolism , Phosphorylation , Ventral Tegmental Area/metabolism , Behavior, Animal
4.
Sci Rep ; 14(1): 15136, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956153

ABSTRACT

The potential long-term effects of anesthesia on cognitive development, especially in neonates and infants, have raised concerns. However, our understanding of its underlying mechanisms and effective treatments is still limited. In this study, we found that early exposure to isoflurane (ISO) impaired fear memory retrieval, which was reversed by dexmedetomidine (DEX) pre-treatment. Measurement of c-fos expression revealed that ISO exposure significantly increased neuronal activation in the zona incerta (ZI). Fiber photometry recording showed that ZI neurons from ISO mice displayed enhanced calcium activity during retrieval of fear memory compared to the control group, while DEX treatment reduced this enhanced calcium activity. Chemogenetic inhibition of ZI neurons effectively rescued the impairments caused by ISO exposure. These findings suggest that the ZI may play a pivotal role in mediating the cognitive effects of anesthetics, offering a potential therapeutic target for preventing anesthesia-related cognitive impairments.


Subject(s)
Fear , Isoflurane , Memory Disorders , Zona Incerta , Isoflurane/pharmacology , Isoflurane/adverse effects , Animals , Fear/drug effects , Mice , Memory Disorders/chemically induced , Zona Incerta/drug effects , Male , Anesthetics, Inhalation/adverse effects , Anesthetics, Inhalation/pharmacology , Neurons/drug effects , Neurons/metabolism , Mice, Inbred C57BL , Dexmedetomidine/pharmacology , Female , Proto-Oncogene Proteins c-fos/metabolism , Memory/drug effects
5.
Exp Dermatol ; 33(7): e15128, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38973249

ABSTRACT

Dry skin is common to many pruritic diseases and is difficult to improve with oral traditional antihistamines. Recently, increasing evidence indicated that histamine H4 receptor (H4R) plays an important role in the occurrence and development of pruritus. Extracellular signal-regulated kinase (ERK) phosphorylation activation in the spinal cord mediates histamine-induced acute and choric itch. However, whether the histamine H4 receptor regulates ERK activation in the dry skin itch remains unclear. In the study, we explore the role of the histamine H4 receptor and p-ERK in the spinal cord in a dry skin mouse model induced by acetone-ether-water (AEW). q-PCR, Western blot, pharmacology and immunofluorescence  were applied in the study. We established a dry skin itch model by repeated application of AEW on the nape of neck in mice. The AEW mice showed typically dry skin histological change and persistent spontaneous scratching behaviour. Histamine H4 receptor, instead of histamine H1 receptor, mediated spontaneous scratching behaviour in AEW mice. Moreover, c-Fos and p-ERK expression in the spinal cord neurons were increased and co-labelled with GRPR-positive neurons in AEW mice. Furthermore, H4R agonist 4-methyhistamine dihydrochloride (4-MH)induced itch. Both 4-MH-induced itch and the spontaneous itch in AEW mice were blocked by p-ERK inhibitor U0126. Finally, intrathecal H4R receptor antagonist JNJ7777120 inhibited spinal p-ERK expression in AEW mice. Our results indicated that spinal H4R mediates itch via ERK activation in the AEW-induced dry skin mice.


Subject(s)
Acetone , Extracellular Signal-Regulated MAP Kinases , Pruritus , Receptors, Histamine H4 , Spinal Cord , Animals , Pruritus/chemically induced , Pruritus/metabolism , Receptors, Histamine H4/metabolism , Mice , Spinal Cord/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Acetone/pharmacology , Water , Ether , Disease Models, Animal , Phosphorylation , Indoles/pharmacology , Butadienes/pharmacology , Piperazines/pharmacology , Nitriles/pharmacology , Skin/metabolism , Chronic Disease , Methylhistamines , Proto-Oncogene Proteins c-fos/metabolism , Mice, Inbred C57BL
6.
Adv Neurobiol ; 38: 81-109, 2024.
Article in English | MEDLINE | ID: mdl-39008012

ABSTRACT

When neurons are recruited to form the memory engram, they are driven to activate the expression of a series of immediate-early genes (IEGs). While these IEGs have been used relatively indiscriminately to identify the so-called engram neurons, recent research has demonstrated that different IEG ensembles can be physically and functionally distinct within the memory engram. This inherent heterogeneity of the memory engram is driven by the diversity in the functions and distributions of different IEGs. This process, which we call molecular sorting, is analogous to sorting the entire population of engram neurons into different sub-engrams molecularly defined by different IEGs. In this chapter, we will describe the molecular sorting process by systematically reviewing published work on engram ensemble cells defined by the following four major IEGs: Fos, Npas4, Arc, and Egr1. By comparing and contrasting these likely different components of the memory engram, we hope to gain a better understanding of the logic and significance behind the molecular sorting process for memory functions.


Subject(s)
Early Growth Response Protein 1 , Genes, Immediate-Early , Memory , Neurons , Memory/physiology , Neurons/metabolism , Animals , Humans , Early Growth Response Protein 1/metabolism , Nerve Tissue Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Cytoskeletal Proteins/metabolism
7.
PLoS Biol ; 22(7): e3002646, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39012916

ABSTRACT

Autism spectrum disorders (ASDs) are considered neural dysconnectivity syndromes. To better understand ASD and uncover potential treatments, it is imperative to know and dissect the connectivity deficits under conditions of autism. Here, we apply a whole-brain immunostaining and quantification platform to demonstrate impaired structural and functional connectivity and aberrant whole-brain synchronization in a Tbr1+/- autism mouse model. We express a channelrhodopsin variant oChIEF fused with Citrine at the basolateral amygdala (BLA) to outline the axonal projections of BLA neurons. By activating the BLA under blue light theta-burst stimulation (TBS), we then evaluate the effect of BLA activation on C-FOS expression at a whole brain level to represent neural activity. We show that Tbr1 haploinsufficiency almost completely disrupts contralateral BLA axonal projections and results in mistargeting in both ipsilateral and contralateral hemispheres, thereby globally altering BLA functional connectivity. Based on correlated C-FOS expression among brain regions, we further show that Tbr1 deficiency severely disrupts whole-brain synchronization in the absence of salient stimulation. Tbr1+/- and wild-type (WT) mice exhibit opposing responses to TBS-induced amygdalar activation, reducing synchronization in WT mice but enhancing it in Tbr1+/- mice. Whole-brain modular organization and intermodule connectivity are also affected by Tbr1 deficiency and amygdalar activation. Following BLA activation by TBS, the synchronizations of the whole brain and the default mode network, a specific subnetwork highly relevant to ASD, are enhanced in Tbr1+/- mice, implying a potential ameliorating effect of amygdalar stimulation on brain function. Indeed, TBS-mediated BLA activation increases nose-to-nose social interactions of Tbr1+/- mice, strengthening evidence for the role of amygdalar connectivity in social behaviors. Our high-resolution analytical platform reveals the inter- and intrahemispheric connectopathies arising from ASD. Our study emphasizes the defective synchronization at a whole-brain scale caused by Tbr1 deficiency and implies a potential beneficial effect of deep brain stimulation at the amygdala for TBR1-linked autism.


Subject(s)
Autism Spectrum Disorder , Basolateral Nuclear Complex , Deep Brain Stimulation , Disease Models, Animal , Social Behavior , T-Box Domain Proteins , Animals , Autism Spectrum Disorder/physiopathology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/genetics , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/physiopathology , Mice , Deep Brain Stimulation/methods , Male , Amygdala/metabolism , Amygdala/physiopathology , Brain/metabolism , Brain/physiopathology , Mice, Inbred C57BL , Neural Pathways/physiopathology , Neural Pathways/metabolism , Proto-Oncogene Proteins c-fos/metabolism
8.
Int J Mol Sci ; 25(14)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39062944

ABSTRACT

Polysulfides are endogenously produced in mammals and generally associated with protective functions. Our aim was to investigate the effect of dimethyl trisulfide (DMTS) in a mouse model of acute stress. DMTS activates transient receptor potential ankyrin 1 (TRPA1) channels and leads to neuropeptide release, potentially that of substance P (SP). We hypothesize that DMTS might inhibit the degrading enzymes of endocannabinoids, so this system was also investigated as another possible pathway for mediating the effects of DMTS. Trpa1 gene wild-type (WT) and knockout (KO) mice were used to confirm the role of the TRPA1 ion channel in mediating the effects of DMTS. C57BL/6J, NK1 gene KO, and Tac1 gene KO mice were used to evaluate the effect of DMTS on the release and expression of SP. Some C57BL/6J animals were treated with AM251, an inhibitor of the cannabinoid CB1 receptor, to elucidate the role of the endocannabinoid system in these processes. Open field test (OFT) and forced swim test (FST) were performed in each mouse strain. A tail suspension test (TST) was performed in Trpa1 WT and KO animals. C-FOS immunohistochemistry was carried out on Trpa1 WT and KO animals. The DMTS treatment increased the number of highly active periods and decreased immobility time in the FST in WT animals, but had no effect on the Trpa1 KO mice. The DMTS administration induced neuronal activation in the Trpa1 WT mice in the stress-related brain areas, such as the locus coeruleus, dorsal raphe nucleus, lateral septum, paraventricular nucleus of the thalamus, and paraventricular nucleus of the hypothalamus. DMTS may have a potential role in the regulation of stress-related processes, and the TRPA1 ion channel may also be involved in mediating the effects of DMTS. DMTS can be an ideal candidate for further study as a potential remedy for stress-related disorders.


Subject(s)
Disease Models, Animal , Mice, Inbred C57BL , Mice, Knockout , Sulfides , TRPA1 Cation Channel , Animals , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics , Mice , Sulfides/pharmacology , Male , Substance P/metabolism , Stress, Psychological/metabolism , Stress, Physiological/drug effects , Proto-Oncogene Proteins c-fos/metabolism
9.
CNS Neurosci Ther ; 30(6): e14782, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38828651

ABSTRACT

BACKGROUND: The thalamus system plays critical roles in the regulation of reversible unconsciousness induced by general anesthetics, especially the arousal stage of general anesthesia (GA). But the function of thalamus in GA-induced loss of consciousness (LOC) is little known. The thalamic reticular nucleus (TRN) is the only GABAergic neurons-composed nucleus in the thalamus, which is composed of parvalbumin (PV) and somatostatin (SST)-expressing GABAergic neurons. The anterior sector of TRN (aTRN) is indicated to participate in the induction of anesthesia, but the roles remain unclear. This study aimed to reveal the role of the aTRN in propofol and isoflurane anesthesia. METHODS: We first set up c-Fos straining to monitor the activity variation of aTRNPV and aTRNSST neurons during propofol and isoflurane anesthesia. Subsequently, optogenetic tools were utilized to activate aTRNPV and aTRNSST neurons to elucidate the roles of aTRNPV and aTRNSST neurons in propofol and isoflurane anesthesia. Electroencephalogram (EEG) recordings and behavioral tests were recorded and analyzed. Lastly, chemogenetic activation of the aTRNPV neurons was applied to confirm the function of the aTRN neurons in propofol and isoflurane anesthesia. RESULTS: c-Fos straining showed that both aTRNPV and aTRNSST neurons are activated during the LOC period of propofol and isoflurane anesthesia. Optogenetic activation of aTRNPV and aTRNSST neurons promoted isoflurane induction and delayed the recovery of consciousness (ROC) after propofol and isoflurane anesthesia, meanwhile chemogenetic activation of the aTRNPV neurons displayed the similar effects. Moreover, optogenetic and chemogenetic activation of the aTRN neurons resulted in the accumulated burst suppression ratio (BSR) during propofol and isoflurane GA, although they represented different effects on the power distribution of EEG frequency. CONCLUSION: Our findings reveal that the aTRN GABAergic neurons play a critical role in promoting the induction of propofol- and isoflurane-mediated GA.


Subject(s)
Anesthesia, General , Consciousness , GABAergic Neurons , Isoflurane , Propofol , Propofol/pharmacology , Isoflurane/pharmacology , Animals , GABAergic Neurons/drug effects , GABAergic Neurons/physiology , Mice , Consciousness/drug effects , Consciousness/physiology , Male , Electroencephalography , Anesthetics, Inhalation/pharmacology , Anterior Thalamic Nuclei/drug effects , Anterior Thalamic Nuclei/physiology , Mice, Inbred C57BL , Mice, Transgenic , Anesthetics, Intravenous/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Optogenetics
10.
Elife ; 122024 Jun 21.
Article in English | MEDLINE | ID: mdl-38904658

ABSTRACT

Maternal choline supplementation (MCS) improves cognition in Alzheimer's disease (AD) models. However, the effects of MCS on neuronal hyperexcitability in AD are unknown. We investigated the effects of MCS in a well-established mouse model of AD with hyperexcitability, the Tg2576 mouse. The most common type of hyperexcitability in Tg2576 mice are generalized EEG spikes (interictal spikes [IIS]). IIS also are common in other mouse models and occur in AD patients. In mouse models, hyperexcitability is also reflected by elevated expression of the transcription factor ∆FosB in the granule cells (GCs) of the dentate gyrus (DG), which are the principal cell type. Therefore, we studied ΔFosB expression in GCs. We also studied the neuronal marker NeuN within hilar neurons of the DG because reduced NeuN protein expression is a sign of oxidative stress or other pathology. This is potentially important because hilar neurons regulate GC excitability. Tg2576 breeding pairs received a diet with a relatively low, intermediate, or high concentration of choline. After weaning, all mice received the intermediate diet. In offspring of mice fed the high choline diet, IIS frequency declined, GC ∆FosB expression was reduced, and hilar NeuN expression was restored. Using the novel object location task, spatial memory improved. In contrast, offspring exposed to the relatively low choline diet had several adverse effects, such as increased mortality. They had the weakest hilar NeuN immunoreactivity and greatest GC ΔFosB protein expression. However, their IIS frequency was low, which was surprising. The results provide new evidence that a diet high in choline in early life can improve outcomes in a mouse model of AD, and relatively low choline can have mixed effects. This is the first study showing that dietary choline can regulate hyperexcitability, hilar neurons, ΔFosB, and spatial memory in an animal model of AD.


Subject(s)
Alzheimer Disease , Choline , Dietary Supplements , Disease Models, Animal , Animals , Alzheimer Disease/metabolism , Choline/administration & dosage , Choline/metabolism , Mice , Female , Mice, Transgenic , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , Neurons/metabolism , Neurons/drug effects , Male , Dentate Gyrus/metabolism , Dentate Gyrus/drug effects , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , DNA-Binding Proteins
11.
Cell Rep ; 43(6): 114350, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38870013

ABSTRACT

Renal cell carcinoma with sarcomatoid differentiation (sRCC) is associated with poor survival and a heightened response to immune checkpoint inhibitors (ICIs). Two major barriers to improving outcomes for sRCC are the limited understanding of its gene regulatory programs and the low diagnostic yield of tumor biopsies due to spatial heterogeneity. Herein, we characterized the epigenomic landscape of sRCC by profiling 107 epigenomic libraries from tissue and plasma samples from 50 patients with RCC and healthy volunteers. By profiling histone modifications and DNA methylation, we identified highly recurrent epigenomic reprogramming enriched in sRCC. Furthermore, CRISPRa experiments implicated the transcription factor FOSL1 in activating sRCC-associated gene regulatory programs, and FOSL1 expression was associated with the response to ICIs in RCC in two randomized clinical trials. Finally, we established a blood-based diagnostic approach using detectable sRCC epigenomic signatures in patient plasma, providing a framework for discovering epigenomic correlates of tumor histology via liquid biopsy.


Subject(s)
Carcinoma, Renal Cell , Epigenomics , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/metabolism , Epigenomics/methods , DNA Methylation/genetics , Cell Differentiation , Gene Expression Regulation, Neoplastic , Male , Female , Epigenesis, Genetic , Middle Aged , Proto-Oncogene Proteins c-fos
12.
Eur J Pharmacol ; 977: 176756, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38897021

ABSTRACT

Repeated exposure to propofol during early brain development is associated with anxiety disorders in adulthood, yet the mechanisms underlying propofol-induced susceptibility to anxiety disorders remain elusive. The lateral septum (LS), primarily composed of γ-aminobutyric acidergic (GABAergic) neurons, serves as a key brain region in the regulation of anxiety. However, it remains unclear whether LS GABAergic neurons are implicated in propofol-induced anxiety. Therefore, we conducted c-Fos immunostaining of whole-brain slices from mice exposed to propofol during early life. Our findings indicate that propofol exposure activates GABAergic neurons in the LS. Selective activation of LS GABAergic neurons resulted in increased anxiety-like behavior, while selective inhibition of these neurons reduced such behaviors. These results suggest that the LS is a critical brain region involved in propofol-induced anxiety. Furthermore, we investigated the molecular mechanism of propofol-induced anxiety in the LS. Microglia activation underlies the development of anxiety. Immunofluorescence staining and Western blot analysis of LS revealed activated microglia and significantly elevated levels of phospho-NF-κB p65 protein. Additionally, a decrease in the number of neuronal spines was observed. Our study highlights the crucial role of the LS in the development of anxiety-like behavior in adulthood following childhood propofol exposure, accompanied by the activation of inflammatory pathways.


Subject(s)
Anxiety , Behavior, Animal , GABAergic Neurons , Microglia , Propofol , Propofol/pharmacology , Animals , Anxiety/chemically induced , Mice , Male , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Behavior, Animal/drug effects , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Proto-Oncogene Proteins c-fos/metabolism , Mice, Inbred C57BL , Transcription Factor RelA/metabolism , Dendritic Spines/drug effects , Dendritic Spines/pathology , Dendritic Spines/metabolism
13.
Cell Biol Toxicol ; 40(1): 44, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862832

ABSTRACT

BACKGROUND: Vasculogenic mimicry (VM) is an enigmatic physiological feature that influences blood supply within glioblastoma (GBM) tumors for their sustained growth. Previous studies identify NFATC3, FOSL1 and HNRNPA2B1 as significant mediators of VEGFR2, a key player in vasculogenesis, and their molecular relationships may be crucial for VM in GBM. AIMS: The aim of this study was to understand how NFATC3, FOSL1 and HNRNPA2B1 collectively influence VM in GBM. METHODS: We have investigated the underlying gene regulatory mechanisms for VM in GBM cell lines U251 and U373 in vitro and in vivo. In vitro cell-based assays were performed to explore the role of NFATC3, FOSL1 and HNRNPA2B1 in GBM cell proliferation, VM and migration, in the context of RNA interference (RNAi)-mediated knockdown alongside corresponding controls. Western blotting and qRT-PCR assays were used to examine VEGFR2 expression levels. CO-IP was employed to detect protein-protein interactions, ChIP was used to detect DNA-protein complexes, and RIP was used to detect RNA-protein complexes. Histochemical staining was used to detect VM tube formation in vivo. RESULTS: Focusing on NFATC3, FOSL1 and HNRNPA2B1, we found each was significantly upregulated in GBM and positively correlated with VM-like cellular behaviors in U251 and U373 cell lines. Knockdown of NFATC3, FOSL1 or HNRNPA2B1 each resulted in decreased levels of VEGFR2, a key growth factor gene that drives VM, as well as the inhibition of proliferation, cell migration and extracorporeal VM activity. Chromatin immunoprecipitation (ChIP) studies and luciferase reporter gene assays revealed that NFATC3 binds to the promoter region of VEGFR2 to enhance VEGFR2 gene expression. Notably, FOSL1 interacts with NFATC3 as a co-factor to potentiate the DNA-binding capacity of NFATC3, resulting in enhanced VM-like cellular behaviors. Also, level of NFATC3 protein in cells was enhanced through HNRNPA2B1 binding of NFATC3 mRNA. Furthermore, RNAi-mediated silencing of NFATC3, FOSL1 and HNRNPA2B1 in GBM cells reduced their capacity for tumor formation and VM-like behaviors in vivo. CONCLUSION: Taken together, our findings identify NFATC3 as an important mediator of GBM tumor growth through its molecular and epistatic interactions with HNRNPA2B1 and FOSL1 to influence VEGFR2 expression and VM-like cellular behaviors.


Subject(s)
Cell Movement , Cell Proliferation , Glioblastoma , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , NFATC Transcription Factors , Neovascularization, Pathologic , Proto-Oncogene Proteins c-fos , Humans , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/blood supply , Cell Line, Tumor , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Animals , Cell Proliferation/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Cell Movement/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Gene Expression Regulation, Neoplastic , Mice , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/blood supply , Mice, Nude
14.
J Physiol Sci ; 74(1): 33, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38867187

ABSTRACT

Hibernation and torpor are not passive responses caused by external temperature drops and fasting but are active brain functions that lower body temperature. A population of neurons in the preoptic area was recently identified as such active torpor-regulating neurons. We hypothesized that the other hypothermia-inducing maneuvers would also activate these neurons. To test our hypothesis, we first refined the previous observations, examined the brain regions explicitly activated during the falling phase of body temperature using c-Fos expression, and confirmed the preoptic area. Next, we observed long-lasting hypothermia by reactivating torpor-tagged Gq-expressing neurons using the activity tagging and DREADD systems. Finally, we found that about 40-60% of torpor-tagged neurons were activated by succeeding isoflurane anesthesia and by icv administration of an adenosine A1 agonist. Isoflurane-induced and central adenosine-induced hypothermia is, at least in part, an active process mediated by the torpor-regulating neurons in the preoptic area.


Subject(s)
Adenosine , Isoflurane , Neurons , Preoptic Area , Animals , Preoptic Area/drug effects , Preoptic Area/metabolism , Isoflurane/pharmacology , Isoflurane/administration & dosage , Adenosine/administration & dosage , Adenosine/pharmacology , Adenosine/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Male , Anesthetics, Inhalation/pharmacology , Anesthetics, Inhalation/administration & dosage , Body Temperature/drug effects , Body Temperature/physiology , Hypothermia/chemically induced , Hypothermia/metabolism , Torpor/drug effects , Mice , Proto-Oncogene Proteins c-fos/metabolism
15.
eNeuro ; 11(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38839305

ABSTRACT

Social behavior is important for our well-being, and its dysfunctions impact several pathological conditions. Although the involvement of glutamate is undeniable, the relevance of vesicular glutamate transporter type 3 (VGluT3), a specific vesicular transporter, in the control of social behavior is not sufficiently explored. Since midbrain median raphe region (MRR) is implicated in social behavior and the nucleus contains high amount of VGluT3+ neurons, we compared the behavior of male VGluT3 knock-out (KO) and VGluT3-Cre mice, the latter after chemogenetic MRR-VGluT3 manipulation. Appropriate control groups were included. Behavioral test battery was used for social behavior (sociability, social discrimination, social interaction, resident intruder test) and possible confounding factors (open field, elevated plus maze, Y-maze tests). Neuronal activation was studied by c-Fos immunohistochemistry. Human relevance was confirmed by VGluT3 gene expression in relevant human brainstem areas. VGluT3 KO mice exhibited increased anxiety, social interest, but also aggressive behavior in anxiogenic environment and impaired social memory. For KO animals, social interaction induced lower cell activation in the anterior cingulate, infralimbic cortex, and medial septum. In turn, excitation of MRR-VGluT3+ neurons was anxiolytic. Inhibition increased social interest 24 h later but decreased mobility and social behavior in aggressive context. Chemogenetic activation increased the number of c-Fos+ neurons only in the MRR. We confirmed the increased anxiety-like behavior and impaired memory of VGluT3 KO strain and revealed increased, but inadequate, social behavior. MRR-VGluT3 neurons regulated mobility and social and anxiety-like behavior in a context-dependent manner. The presence of VGluT3 mRNA on corresponding human brain areas suggests clinical relevance.


Subject(s)
Anxiety , Mice, Knockout , Social Behavior , Animals , Male , Humans , Anxiety/metabolism , Raphe Nuclei/metabolism , Mice , Neurons/metabolism , Mice, Inbred C57BL , Behavior, Animal/physiology , Mice, Transgenic , Amino Acid Transport Systems, Acidic/metabolism , Amino Acid Transport Systems, Acidic/genetics , Proto-Oncogene Proteins c-fos/metabolism , Aggression/physiology
16.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891875

ABSTRACT

Transcranial focused ultrasound stimulation (tFUS) has emerged as a promising neuromodulation technique that delivers acoustic energy with high spatial resolution for inducing long-term potentiation (LTP)- or depression (LTD)-like plasticity. The variability in the primary effects of tFUS-induced plasticity could be due to different stimulation patterns, such as intermittent versus continuous, and is an aspect that requires further detailed exploration. In this study, we developed a platform to evaluate the neuromodulatory effects of intermittent and continuous tFUS on motor cortical plasticity before and after tFUS application. Three groups of rats were exposed to either intermittent, continuous, or sham tFUS. We analyzed the neuromodulatory effects on motor cortical excitability by examining changes in motor-evoked potentials (MEPs) elicited by transcranial magnetic stimulation (TMS). We also investigated the effects of different stimulation patterns on excitatory and inhibitory neural biomarkers, examining c-Fos and glutamic acid decarboxylase (GAD-65) expression using immunohistochemistry staining. Additionally, we evaluated the safety of tFUS by analyzing glial fibrillary acidic protein (GFAP) expression. The current results indicated that intermittent tFUS produced a facilitation effect on motor excitability, while continuous tFUS significantly inhibited motor excitability. Furthermore, neither tFUS approach caused injury to the stimulation sites in rats. Immunohistochemistry staining revealed increased c-Fos and decreased GAD-65 expression following intermittent tFUS. Conversely, continuous tFUS downregulated c-Fos and upregulated GAD-65 expression. In conclusion, our findings demonstrate that both intermittent and continuous tFUS effectively modulate cortical excitability. The neuromodulatory effects may result from the activation or deactivation of cortical neurons following tFUS intervention. These effects are considered safe and well-tolerated, highlighting the potential for using different patterns of tFUS in future clinical neuromodulatory applications.


Subject(s)
Evoked Potentials, Motor , Motor Cortex , Neuronal Plasticity , Transcranial Magnetic Stimulation , Animals , Motor Cortex/physiology , Rats , Male , Evoked Potentials, Motor/physiology , Transcranial Magnetic Stimulation/methods , Proto-Oncogene Proteins c-fos/metabolism , Ultrasonic Waves , Rats, Sprague-Dawley , Glial Fibrillary Acidic Protein/metabolism , Glutamate Decarboxylase/metabolism
17.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38937108

ABSTRACT

Ghrelin is a stomach-derived hormone that increases feeding and is elevated in response to chronic psychosocial stressors. The effects of ghrelin on feeding are mediated by the binding of ghrelin to the growth hormone secretagogue receptor (GHSR), a receptor located in hypothalamic and extrahypothalamic regions important for regulating food intake and metabolic rate. The ability of ghrelin to enter the brain, however, seems to be restricted to circumventricular organs like the median eminence and the brainstem area postrema, whereas ghrelin does not readily enter other GHSR-expressing regions like the ventral tegmental area (VTA). Interestingly, social stressors result in increased blood-brain barrier permeability, and this could therefore facilitate the entry of ghrelin into the brain. To investigate this, we exposed mice to social defeat stress for 21 d and then peripherally injected a Cy5-labelled biologically active ghrelin analog. The results demonstrate that chronically stressed mice exhibit higher Cy5-ghrelin fluorescence in several hypothalamic regions in addition to the ARC, including the hippocampus and midbrain. Furthermore, Cy5-ghrelin injections resulted in increased FOS expression in regions associated with the reward system in chronically stressed mice. Further histologic analyses identified a reduction in the branching of hypothalamic astrocytes in the ARC-median eminence junction, suggesting increased blood-brain barrier permeability. These data support the hypothesis that during metabolically challenging conditions like chronic stress, ghrelin may be more able to cross the blood-brain barrier and diffuse throughout the brain to target GHSR-expressing brain regions away from circumventricular organs.


Subject(s)
Blood-Brain Barrier , Brain , Ghrelin , Mice, Inbred C57BL , Social Defeat , Stress, Psychological , Animals , Ghrelin/metabolism , Male , Stress, Psychological/metabolism , Brain/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Mice , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Ghrelin/metabolism
18.
Pflugers Arch ; 476(8): 1235-1247, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38856775

ABSTRACT

To assess the possible interactions between the dorsolateral periaqueductal gray matter (dlPAG) and the different domains of the nucleus ambiguus (nA), we have examined the pattern of double-staining c-Fos/FoxP2 protein immunoreactivity (c-Fos-ir/FoxP2-ir) and tyrosine hydroxylase (TH) throughout the rostrocaudal extent of nA in spontaneously breathing anaesthetised male Sprague-Dawley rats during dlPAG electrical stimulation. Activation of the dlPAG elicited a selective increase in c-Fos-ir with an ipsilateral predominance in the somatas of the loose (p < 0.05) and compact formation (p < 0.01) within the nA and confirmed the expression of FoxP2 bilaterally in all the domains within the nA. A second group of experiments was made to examine the importance of the dlPAG in modulating the laryngeal response evoked after electrical or chemical (glutamate) dlPAG stimulations. Both electrical and chemical stimulations evoked a significant decrease in laryngeal resistance (subglottal pressure) (p < 0.001) accompanied with an increase in respiratory rate together with a pressor and tachycardic response. The results of our study contribute to new data on the role of the mesencephalic neuronal circuits in the control mechanisms of subglottic pressure and laryngeal activity.


Subject(s)
Electric Stimulation , Larynx , Periaqueductal Gray , Proto-Oncogene Proteins c-fos , Rats, Sprague-Dawley , Animals , Male , Rats , Periaqueductal Gray/metabolism , Periaqueductal Gray/physiology , Electric Stimulation/methods , Proto-Oncogene Proteins c-fos/metabolism , Larynx/physiology , Larynx/metabolism , Forkhead Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/metabolism , Pressure , Medulla Oblongata/metabolism , Medulla Oblongata/physiology , Glutamic Acid/metabolism
19.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928250

ABSTRACT

Posttraumatic stress disorder (PTSD) is a debilitating psychosomatic condition characterized by impairment of brain fear circuits and persistence of exceptionally strong associative memories resistant to extinction. In this study, we investigated the neural and behavioral consequences of inhibiting protein synthesis, a process known to suppress the formation of conventional aversive memories, in an established PTSD animal model based on contextual fear conditioning in mice. Control animals were subjected to the conventional fear conditioning task. Utilizing c-Fos neural activity mapping, we found that the retrieval of PTSD and normal aversive memories produced activation of an overlapping set of brain structures. However, several specific areas, such as the infralimbic cortex and the paraventricular thalamic nucleus, showed an increase in the PTSD group compared to the normal aversive memory group. Administration of protein synthesis inhibitor before PTSD induction disrupted the formation of traumatic memories, resulting in behavior that matched the behavior of mice with usual aversive memory. Concomitant with this behavioral shift was a normalization of brain c-Fos activation pattern matching the one observed in usual fear memory. Our findings demonstrate that inhibiting protein synthesis during traumatic experiences significantly impairs the development of PTSD in a mouse model. These data provide insights into the neural underpinnings of protein synthesis-dependent traumatic memory formation and open prospects for the development of new therapeutic strategies for PTSD prevention.


Subject(s)
Fear , Memory , Proto-Oncogene Proteins c-fos , Stress Disorders, Post-Traumatic , Animals , Male , Mice , Brain/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Protein Biosynthesis , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Stress Disorders, Post-Traumatic/metabolism
20.
Mol Biol (Mosk) ; 58(1): 78-87, 2024.
Article in Russian | MEDLINE | ID: mdl-38943581

ABSTRACT

Stress can play a significant role in arterial hypertension and many other complications of cardiovascular diseases. Considerable attention is paid to the study of the molecular mechanisms involved in the body response to stressful influences, but there are still many blank spots in understanding the details. ISIAH rats model the stress-sensitive form of arterial hypertension. ISIAH rats are characterized by genetically determined enhanced activities of the hypothalamic-pituitary-adrenocortical and sympathetic-adrenomedullary systems, suggesting a functional state of increased stress reactivity. For the first time, the temporal expression patterns of Fos and several related genes were studied in the hypothalamus of adult male hypertensive ISIAH rats after a single exposure to restraint stress for 30, 60, or 120 min. Fos transcription was activated and peaked 1 h after the start of restraint stress. The time course of Fos activation coincided with that of blood pressure increase after stress. Activation of hypothalamic neurons also alters the transcription levels of several transcription factor genes (Jun, Nr4a3, Jdp2, and Ppargc1a), which are associated with the development of cardiovascular diseases. Because Fos induction is a marker of brain neuron activation, activation of hypothalamic neurons and an increase in blood pressure were concluded to accompany increased stress reactivity of the hypothalamic-pituitary-adrenocortical and sympathoadrenal systems in hypertensive ISIAH rats during short-term restraint.


Subject(s)
Gene Expression Regulation , Hypertension , Hypothalamus , Animals , Hypertension/metabolism , Hypertension/genetics , Hypertension/pathology , Rats , Hypothalamus/metabolism , Male , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Restraint, Physical , Stress, Psychological/metabolism , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Blood Pressure/genetics , Stress, Physiological/genetics , Neurons/metabolism , Neurons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL