Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article in English | MEDLINE | ID: mdl-39000021

ABSTRACT

Retinoblastoma, a pediatric ocular malignancy, presents significant challenges in comprehending its molecular underpinnings and targeted therapeutic approaches. The dysregulated activity of histone deacetylases (HDACs) has been associated with retinoblastoma pathogenesis, influencing critical cellular processes like cell cycle regulation or retinal ganglion cell apoptosis. Through their deacetylase activity, HDACs exert control over key tumor suppressors and oncogenes, influencing the delicate equilibrium between proliferation and cell death. Furthermore, the interplay between HDACs and the retinoblastoma protein pathway, a pivotal aspect of retinoblastoma etiology, reveals a complex network of interactions influencing the tumor microenvironment. The examination of HDAC inhibitors, encompassing both established and novel compounds, offers insights into potential approaches to restore acetylation balance and impede retinoblastoma progression. Moreover, the identification of specific HDAC isoforms exhibiting varying expression in retinoblastoma provides avenues for personalized therapeutic strategies, allowing for interventions tailored to individual patient profiles. This review focuses on the intricate interrelationship between HDACs and retinoblastoma, shedding light on epigenetic mechanisms that control tumor development and progression. The exploration of HDAC-targeted therapies underscores the potential for innovative treatment modalities in the pursuit of more efficacious and personalized management strategies for this disease.


Subject(s)
Histone Deacetylase Inhibitors , Histone Deacetylases , Retinoblastoma , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retinoblastoma/pathology , Humans , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylase Inhibitors/pharmacology , Animals , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinal Neoplasms/pathology , Epigenesis, Genetic , Acetylation , Tumor Microenvironment , Gene Expression Regulation, Neoplastic , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/genetics
2.
Front Mol Neurosci ; 17: 1412407, 2024.
Article in English | MEDLINE | ID: mdl-38813437

ABSTRACT

The complex nature of the retina demands well-organized signaling to uphold signal accuracy and avoid interference, a critical aspect in handling a variety of visual stimuli. A-kinase anchoring proteins (AKAPs), known for binding protein kinase A (PKA), contribute to the specificity and efficiency of retinal signaling. They play multifaceted roles in various retinal cell types, influencing photoreceptor sensitivity, neurotransmitter release in bipolar cells, and the integration of visual information in ganglion cells. AKAPs like AKAP79/150 and AKAP95 exhibit distinct subcellular localizations, impacting synaptic transmission and receptor sensitivity in photoreceptors and bipolar cells. Furthermore, AKAPs are involved in neuroprotective mechanisms and axonal degeneration, particularly in retinal ganglion cells. In particular, AKAP6 coordinates stress-specific signaling and promotes neuroprotection following optic nerve injury. As our review underscores the therapeutic potential of targeting AKAP signaling complexes for retinal neuroprotection and enhancement, it acknowledges challenges in developing selective drugs that target complex protein-protein interactions. Overall, this exploration of AKAPs provides valuable insights into the intricacies of retinal signaling, offering a foundation for understanding and potentially addressing retinal disorders.

3.
Mol Neurobiol ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38639863

ABSTRACT

Retinal ganglion cells (RGCs), neurons transmitting visual information via the optic nerve, fail to regenerate their axons after injury. The progressive loss of RGC function underlies the pathophysiology of glaucoma and other optic neuropathies, often leading to irreversible blindness. Therefore, there is an urgent need to identify the regulators of RGC survival and the regenerative program. In this study, we investigated the role of the family of transcription factors known as nuclear factor of activated T cells (NFAT), which are expressed in the retina; however, their role in RGC survival after injury is unknown. Using the optic nerve crush (ONC) model, widely employed to study optic neuropathies and central nervous system axon injury, we found that NFATc4 is specifically but transiently up-regulated in response to mechanical injury. In the injured retina, NFATc4 immunolocalized primarily to the ganglionic cell layer. Utilizing NFATc4-/- and NFATc3-/- mice, we demonstrated that NFATc4, but not NFATc3, knockout increased RGC survival, improved retina function, and delayed axonal degeneration. Microarray screening data, along with decreased immunostaining of cleaved caspase-3, revealed that NFATc4 knockout was protective against ONC-induced degeneration by suppressing pro-apoptotic signaling. Finally, we used lentiviral-mediated NFATc4 delivery to the retina of NFATc4-/- mice and reversed the pro-survival effect of NFATc4 knockout, conclusively linking the enhanced survival of injured RGCs to NFATc4-dependent mechanisms. In summary, this study is the first to demonstrate that NFATc4 knockout may confer transient RGC neuroprotection and decelerate axonal degeneration after injury, providing a potent therapeutic strategy for optic neuropathies.

4.
Biomolecules ; 14(2)2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38397420

ABSTRACT

The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.


Subject(s)
Calcium , Neurons , Calcium/metabolism , Neurons/metabolism , Calcium Signaling/physiology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Nerve Regeneration
5.
Front Immunol ; 14: 1281882, 2023.
Article in English | MEDLINE | ID: mdl-38077352

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by a progressive loss of cognitive functions. While the exact causes of this debilitating disorder remain elusive, numerous investigations have characterized its two core pathologies: the presence of ß-amyloid plaques and tau tangles. Additionally, multiple studies of postmortem brain tissue, as well as results from AD preclinical models, have consistently demonstrated the presence of a sustained inflammatory response. As the persistent immune response is associated with neurodegeneration, it became clear that it may also exacerbate other AD pathologies, providing a link between the initial deposition of ß-amyloid plaques and the later development of neurofibrillary tangles. Initially discovered in T cells, the nuclear factor of activated T-cells (NFAT) is one of the main transcription factors driving the expression of inflammatory genes and thus regulating immune responses. NFAT-dependent production of inflammatory mediators is controlled by Ca2+-dependent protein phosphatase calcineurin (CaN), which dephosphorylates NFAT and promotes its transcriptional activity. A substantial body of evidence has demonstrated that aberrant CaN/NFAT signaling is linked to several pathologies observed in AD, including neuronal apoptosis, synaptic deficits, and glia activation. In view of this, the role of NFAT isoforms in AD has been linked to disease progression at different stages, some of which are paralleled to diminished cognitive status. The use of classical inhibitors of CaN/NFAT signaling, such as tacrolimus or cyclosporine, or adeno-associated viruses to specifically inhibit astrocytic NFAT activation, has alleviated some symptoms of AD by diminishing ß-amyloid neurotoxicity and neuroinflammation. In this article, we discuss the recent findings related to the contribution of CaN/NFAT signaling to the progression of AD and highlight the possible benefits of targeting this pathway in AD treatment.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/metabolism , Calcineurin/metabolism , Plaque, Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism
6.
Antioxidants (Basel) ; 12(2)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36829790

ABSTRACT

Vitamin C (ascorbic acid) is well known for its potent antioxidant properties, as it can neutralize ROS and free radicals, thereby protecting cellular elements from oxidative stress. It predominantly exists as an ascorbate anion and after oxidation to dehydroascorbic acid and further breakdown, is removed from the cells. In nervous tissue, a progressive decrease in vitamin C level or its prolonged deficiency have been associated with an increased risk of disturbances in neurotransmission, leading to dysregulation in brain function. Therefore, understanding the regulatory function of vitamin C in antioxidant defence and identification of its molecular targets deserves more attention. One of the key signalling ions is calcium and a transient rise in its concentration is crucial for all neuronal processes. Extracellular Ca2+ influx (through specific ion channels) or Ca2+ release from intracellular stores (endoplasmic reticulum, mitochondria) are precisely controlled. Ca2+ regulates the functioning of the CNS, including growth, development, myelin formation, synthesis of catecholamines, modulation of neurotransmission and antioxidant protection. A growing body of evidence indicates a unique role for vitamin C in these processes. In this short review, we focus on vitamin C in the regulation of calcium-involved pathways under physiological and stress conditions in the brain.

7.
Int J Mol Sci ; 24(4)2023 Feb 04.
Article in English | MEDLINE | ID: mdl-36834528

ABSTRACT

The family of myocyte enhancer factor 2 (MEF2) transcription factors comprises four highly conserved members that play an important role in the nervous system. They appear in precisely defined time frames in the developing brain to turn on and turn off genes affecting growth, pruning and survival of neurons. MEF2s are known to dictate neuronal development, synaptic plasticity and restrict the number of synapses in the hippocampus, thus affecting learning and memory formation. In primary neurons, negative regulation of MEF2 activity by external stimuli or stress conditions is known to induce apoptosis, albeit the pro or antiapoptotic action of MEF2 depends on the neuronal maturation stage. By contrast, enhancement of MEF2 transcriptional activity protects neurons from apoptotic death both in vitro and in preclinical models of neurodegenerative diseases. A growing body of evidence places this transcription factor in the center of many neuropathologies associated with age-dependent neuronal dysfunctions or gradual but irreversible neuron loss. In this work, we discuss how the altered function of MEF2s during development and in adulthood affecting neuronal survival may be linked to neuropsychiatric disorders.


Subject(s)
Apoptosis , Gene Expression Regulation , MEF2 Transcription Factors/genetics , Brain/metabolism , Neurons/metabolism
8.
Front Cell Neurosci ; 16: 890827, 2022.
Article in English | MEDLINE | ID: mdl-35677757

ABSTRACT

PMCA2 is not expressed until the late embryonic state when the control of subtle Ca2+ fluxes becomes important for neuronal specialization. During this period, immature neurons are especially vulnerable to degenerative insults induced by the N-methyl-D-aspartate (NMDA) receptor blocker, ketamine. As H19-7 hippocampal progenitor cells isolated from E17 do not express the PMCA2 isoform, they constitute a valuable model for studying its role in neuronal development. In this study, we demonstrated that heterologous expression of PMCA2b enhanced the differentiation of H19-7 cells and protected from ketamine-induced death. PMCA2b did not affect resting [Ca2+]c in the presence or absence of ketamine and had no effect on the rate of Ca2+ clearance following membrane depolarization in the presence of the drug. The upregulation of endogenous PMCA1 demonstrated in response to PMCA2b expression as well as ketamine-induced PMCA4 depletion were indifferent to the rate of Ca2+ clearance in the presence of ketamine. Yet, co-expression of PMCA4b and PMCA2b was able to partially restore Ca2+ extrusion diminished by ketamine. The profiling of NMDA receptor expression showed upregulation of the NMDAR1 subunit in PMCA2b-expressing cells and increased co-immunoprecipitation of both proteins following ketamine treatment. Further microarray screening demonstrated a significant influence of PMCA2b on GABA signaling in differentiating progenitor cells, manifested by the unique regulation of several genes key to the GABAergic transmission. The overall activity of glutamate decarboxylase remained unchanged, but Ca2+-induced GABA release was inhibited in the presence of ketamine. Interestingly, PMCA2b expression was able to reverse this effect. The mechanism of GABA secretion normalization in the presence of ketamine may involve PMCA2b-mediated inhibition of GABA transaminase, thus shifting GABA utilization from energetic purposes to neurosecretion. In this study, we show for the first time that developmentally controlled PMCA expression may dictate the pattern of differentiation of hippocampal progenitor cells. Moreover, the appearance of PMCA2 early in development has long-standing consequences for GABA metabolism with yet an unpredictable influence on GABAergic neurotransmission during later stages of brain maturation. In contrast, the presence of PMCA2b seems to be protective for differentiating progenitor cells from ketamine-induced apoptotic death.

9.
Chemosphere ; 287(Pt 3): 132284, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34563782

ABSTRACT

Among polychlorinated naphthalenes (PCNs), listed by the Stockholm convention as Persistent Organic Pollutants (POPs), hexachloronaphthalenes are considered the most toxic and raise the highest concern. Of these, 1,2,3,5,6,7-hexachloronaphthalanene (PCN67) is considered the main congener affecting human health due to its hepatotoxicity and its ability to disturb the reproductive, endocrine, and hematological systems. It is also prevalent in human serum/plasma, milk, and adipose tissue. However, little is known about its neurotoxicity, despite the fact that anorectic effects have been observed in workers occupationally exposed to PCNs and in animal research on PCN67. Since dopamine is involved in many aspects of food intake, the aim of this study was to confirm whether PCN67 affects dopamine synthesis in differentiated PC12 cells, a widely used model of neurosecretion. Our results show that exposure to PCN67 resulted in diminished dopamine content and release. Moreover, PCN67 also affected the expression of tyrosine hydroxylase and lowered the expression of vesicular monoamine transporter 1 (VMAT1). In addition, significantly lower expression of antioxidant enzymes, including catalase, glutathione peroxidase and copper/zinc superoxide dismutase, was observed in comparison to the vehicle. In conclusion, PCN67 appears to disturb dopaminergic transmission by altering tyrosine hydroxylation, reducing VMAT1 expression and impairing antioxidant protection. Our study provides a potential mechanism for how PCN67 may cause dopamine deficiency and contribute to neuronal death by affecting cellular antioxidant potency; however, this conclusion requires further research.


Subject(s)
Dopamine , Neurotoxicity Syndromes , Animals , Humans , Naphthalenes/toxicity , PC12 Cells , Rats
10.
Cells ; 10(5)2021 05 17.
Article in English | MEDLINE | ID: mdl-34067760

ABSTRACT

Schizophrenia is a common debilitating disease characterized by continuous or relapsing episodes of psychosis. Although the molecular mechanisms underlying this psychiatric illness remain incompletely understood, a growing body of clinical, pharmacological, and genetic evidence suggests that G protein-coupled receptors (GPCRs) play a critical role in disease development, progression, and treatment. This pivotal role is further highlighted by the fact that GPCRs are the most common targets for antipsychotic drugs. The GPCRs activation evokes slow synaptic transmission through several downstream pathways, many of them engaging intracellular Ca2+ mobilization. Dysfunctions of the neurotransmitter systems involving the action of GPCRs in the frontal and limbic-related regions are likely to underly the complex picture that includes the whole spectrum of positive and negative schizophrenia symptoms. Therefore, the progress in our understanding of GPCRs function in the control of brain cognitive functions is expected to open new avenues for selective drug development. In this paper, we review and synthesize the recent data regarding the contribution of neurotransmitter-GPCRs signaling to schizophrenia symptomology.


Subject(s)
Brain/metabolism , Calcium Signaling , Receptors, Chemokine/metabolism , Receptors, Neurotransmitter/metabolism , Schizophrenia/metabolism , Schizophrenic Psychology , Synaptic Transmission , Animals , Antipsychotic Agents/therapeutic use , Brain/drug effects , Brain/physiopathology , Calcium Signaling/drug effects , Humans , Neurotransmitter Agents/therapeutic use , Receptors, Chemokine/antagonists & inhibitors , Receptors, Neurotransmitter/antagonists & inhibitors , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Synaptic Transmission/drug effects
11.
Int J Mol Sci ; 22(6)2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33801794

ABSTRACT

Calcium in mammalian neurons is essential for developmental processes, neurotransmitter release, apoptosis, and signal transduction. Incorrectly processed Ca2+ signal is well-known to trigger a cascade of events leading to altered response to variety of stimuli and persistent accumulation of pathological changes at the molecular level. To counterbalance potentially detrimental consequences of Ca2+, neurons are equipped with sophisticated mechanisms that function to keep its concentration in a tightly regulated range. Calcium pumps belonging to the P-type family of ATPases: plasma membrane Ca2+-ATPase (PMCA), sarco/endoplasmic Ca2+-ATPase (SERCA) and secretory pathway Ca2+-ATPase (SPCA) are considered efficient line of defense against abnormal Ca2+ rises. However, their role is not limited only to Ca2+ transport, as they present tissue-specific functionality and unique sensitive to the regulation by the main calcium signal decoding protein-calmodulin (CaM). Based on the available literature, in this review we analyze the contribution of these three types of Ca2+-ATPases to neuropathology, with a special emphasis on mental diseases.


Subject(s)
Calcium-Transporting ATPases/metabolism , Mental Disorders/enzymology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Animals , Calcium-Transporting ATPases/chemistry , Humans , Models, Molecular , Nervous System Diseases/enzymology , Plasma Membrane Calcium-Transporting ATPases/chemistry , Protein Conformation , Sarcoplasmic Reticulum Calcium-Transporting ATPases/chemistry
12.
Int J Mol Sci ; 21(21)2020 Nov 09.
Article in English | MEDLINE | ID: mdl-33182497

ABSTRACT

Ketamine is a non-competitive antagonist of NMDA (N-methyl-D-aspartate) receptor, which has been in clinical practice for over a half century. Despite recent data suggesting its harmful side effects, such as neuronal loss, synapse dysfunction or disturbed neural network formation, the drug is still applied in veterinary medicine and specialist anesthesia. Several lines of evidence indicate that structural and functional abnormalities in the nervous system caused by ketamine are crosslinked with the imbalanced activity of multiple Ca2+-regulated signaling pathways. Due to its ubiquitous nature, Ca2+ is also frequently located in the center of ketamine action, although the precise mechanisms underlying drug's negative or therapeutic properties remain mysterious for the large part. This review seeks to delineate the relationship between ketamine-triggered imbalance in Ca2+ homeostasis and functional consequences for downstream processes regulating key aspects of neuronal function.


Subject(s)
Calcium Signaling/drug effects , Calcium/metabolism , Ketamine/adverse effects , Neurons/drug effects , Signal Transduction/drug effects , Animals , Humans , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/drug effects , Synapses/metabolism
13.
Oxid Med Cell Longev ; 2020: 2479234, 2020.
Article in English | MEDLINE | ID: mdl-32685088

ABSTRACT

Hexachloronaphthalene (PCN67) is one of the most toxic among polychlorinated naphthalenes. Despite the known high bioaccumulation and persistence of PCN67 in the environment, it is still unclear to what extent exposure to these substances may interfere with normal neuronal physiology and lead to neurotoxicity. Therefore, the primary goal of this study was to assess the effect of PCN67 in neuronal in vitro models. Neuronal death was assessed upon PCN67 treatment using differentiated PC12 cells and primary hippocampal neurons. At 72 h postexposure, cell viability assays showed an IC50 value of 0.35 µg/ml and dose-dependent damage of neurites and concomitant downregulation of neurofilaments L and M. Moreover, we found that younger primary neurons (DIV4) were much more sensitive to PCN67 toxicity than mature cultures (DIV14). Our comprehensive analysis indicated that the application of PCN67 at the IC50 concentration caused necrosis, which was reflected by an increase in LDH release, HMGB1 protein export to the cytosol, nuclear swelling, and loss of homeostatic control of energy balance. The blockage of mitochondrial calcium uniporter partially rescued the cell viability, loss of mitochondrial membrane potential (ΔΨ m), and the overproduction of reactive oxygen species, suggesting that the underlying mechanism of neurotoxicity involved mitochondrial calcium accumulation. Increased lipid peroxidation as a consequence of oxidative stress was additionally seen for 0.1 µg/ml of PCN67, while this concentration did not affect ΔΨ m and plasma membrane permeability. Our results show for the first time that neuronal mitochondria act as a target for PCN67 and indicate that exposure to this drug may result in neuron loss via mitochondrial-dependent mechanisms.


Subject(s)
Mitochondria/drug effects , Naphthalenes/adverse effects , Nerve Degeneration/chemically induced , Animals , Cell Culture Techniques , Cell Differentiation , Humans , PC12 Cells , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
14.
Neurosci Lett ; 663: 48-54, 2018 01 10.
Article in English | MEDLINE | ID: mdl-28780170

ABSTRACT

Although first mentions about calcium disturbances in psychiatric diseases appeared more than 30 years ago, the most recent genomic and proteomic findings confirmed a significant role of Ca2+ and Ca2+-regulated pathways in development of neuropathological processes, including bipolar disorder and schizophrenia. Moreover, last decades have shown that due to multifactorial nature of both diseases, impairment in neuronal calcium homeostasis may depend not only on disturbed Ca2+ entry system, but also on altered extrusion system. A pivotal role in Ca2+ clearance mechanism is played by plasma membrane Ca2+-ATPase (PMCA), the enzyme responsible for returning the elevated levels of cytosolic Ca2+ back to the resting state. In this paper we summarize the current knowledge about the role of PMCA in bipolar disorder and schizophrenia pathologies, as well as the contribution of several proteins that by interaction with PMCA modify signal transduction mechanisms.


Subject(s)
Bipolar Disorder/metabolism , Calcium Signaling/physiology , Calcium/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Schizophrenia/metabolism , Animals , Bipolar Disorder/pathology , Cell Membrane/metabolism , Cell Membrane/pathology , Humans , Schizophrenia/pathology , Signal Transduction/physiology
15.
Front Cell Neurosci ; 11: 181, 2017.
Article in English | MEDLINE | ID: mdl-28701926

ABSTRACT

Ketamine causes psychotic episodes and is often used as pharmacological model of psychotic-like behavior in animals. There is increasing evidence that molecular mechanism of its action is more complicated than just N-methyl-D-aspartic acid (NMDA) receptor antagonism and involves interaction with the components of calcium homeostatic machinery, in particular plasma membrane calcium pump (PMCA). Therefore, in this study we aimed to characterize brain region-specific effects of ketamine on PMCA activity, interaction with NMDA receptor through postsynaptic density protein 95 (PSD95) scaffolding proteins and glutamate release from nerve endings. In our study, ketamine induced behavioral changes in healthy male rats consistent with psychotic effects. In the same animals, we were able to demonstrate significant inhibition of plasma membrane calcium ATPase (PMCA) activity in cerebellum, hippocampus and striatum. The expression level and isoform composition of PMCAs were also affected in some of these brain compartments, with possible compensatory effects of PMCA1 substituting for decreased expression of PMCA3. Expression of the PDZ domain-containing scaffold protein PSD95 was induced and its association with PMCA4 was higher in most brain compartments upon ketamine treatment. Moreover, increased PSD95/NMDA receptor direct interaction was also reported, strongly suggesting the formation of multiprotein complexes potentially mediating the effect of ketamine on calcium signaling. We further support this molecular mechanism by showing brain region-specific changes in PSD95/PMCA4 spatial colocalization. We also show that ketamine significantly increases synaptic glutamate release in cortex and striatum (without affecting total tissue glutamate content), inducing the expression of vesicular glutamate transporters and decreasing the expression of membrane glutamate reuptake pump excitatory amino acid transporters 2 (EAAT2). Thus, ketamine-mediated PMCA inhibition, by decreasing total Ca2+ clearing potency, may locally raise cytosolic Ca2+ promoting excessive glutamate release. Regional alterations in glutamate secretion can be further driven by PSD95-mediated spatial recruitment of signaling complexes including glutamate receptors and calcium pumps, representing a novel mechanism of psychogenic action of ketamine.

16.
Biochim Biophys Acta Gene Regul Mech ; 1860(4): 502-515, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28153703

ABSTRACT

Brain aging is characterized by progressive loss of plasma membrane calcium pump (PMCA) and its activator - calmodulin (CaM), but the mechanism of this phenomenon remains unresolved. CaM encoded by three genes Calm1, Calm2, Calm3, works to translate Ca2+ signal into changes in frequently opposite cellular activities. This unique function allows CaM to affect gene expression via stimulation of calcineurin (CaN) and its downstream target - nuclear factor of activated T-cells (NFAT) and to terminate Ca2+ signal by stimulation of its extrusion. PMCA, which exists in four isoforms PMCA1-4, may in turn shape the pattern of Ca2+ transients and control CaN activity by its direct binding. Therefore, the interplay between PMCA, CaM and CaN/NFAT is highly plausible. To verify that, we used differentiated PC12 cells with reduced expression of PMCA2 or PMCA3 to mimic the potential changes in aged brain. Manipulation in PMCAs level decreased CaM protein in PMCA2 or PMCA3-reduced lines that was accompanied by down-regulation of Calm1 and Calm2 in both lines, but Calm3 only in PMCA2-reduced cells. Further studies showed substantially higher NFATc2 nuclear accumulation and increased NFAT transcriptional activity. Blocking of CaN/NFAT signalling resulted in almost full CaM recovery, mainly due to up-regulation of Calm2 and Calm3 genes. Moreover, higher occupancy of Calm2 and Calm3 promoters by NFATc2 and increased expression of these genes in response to NFATc2 silencing were demonstrated in PMCA2 and PMCA3-reduced lines. Our results indicate that decrease in CaM level in response to PMCAs downregulation can be driven by CaN/NFAT pathway.


Subject(s)
Calcineurin/metabolism , Calmodulin/genetics , Cell Differentiation/genetics , Gene Expression Regulation , NFATC Transcription Factors/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Animals , Calmodulin/metabolism , Chromatin Immunoprecipitation , Down-Regulation/genetics , Gene Knockdown Techniques , Isoenzymes/metabolism , Models, Biological , Oligonucleotide Array Sequence Analysis , PC12 Cells , RNA, Small Interfering/metabolism , Rats , Transcription, Genetic
18.
Cell Tissue Res ; 363(3): 609-20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26685921

ABSTRACT

Chronic N-methyl-D-aspartate receptor (NMDAR) antagonist treatment can provide valuable neurochemical and neuroanatomical models of experimental psychosis. One such antagonist, ketamine, with its short half-time and well-documented psychotomimetic action, has cognitive effects resembling various aspects of schizophrenia-like symptoms. In order to obtain insights into possible relationships between Ca(2+) homeostasis and schizophrenia-related symptoms, we investigate the effects of chronic ketamine administration on intracellular Ca(2+) levels in various brain regions and on the expression level of key members of the neuronal Ca(2+)-handling system in rats. We show increased intracellular [Ca(2+)] in all of the examined brain regions following ketamine treatment but an altered cytosolic Ca(2+) level correlated with hyperlocomotor activity was only established for the cortex and striatum. Our findings also suggest that an imbalance in the expression between the calcium "on" and "off" systems contributes to the deregulation of brain Ca(2+) homeostasis in our ketamine-induced model of experimental psychosis. Identification of the genes whose expression is affected by ketamine treatment indicates their involvement as putative etiological factors in schizophrenia.


Subject(s)
Calcium/metabolism , Psychotic Disorders/metabolism , Psychotic Disorders/pathology , Animals , Behavior, Animal , Brain/metabolism , Brain/pathology , Cell Separation , Cell Survival , Disease Models, Animal , Gene Expression Regulation , Homeostasis , Intracellular Space/metabolism , Ketamine , Male , Motor Activity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Wistar , Real-Time Polymerase Chain Reaction
19.
Neurochem Int ; 91: 13-25, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26492822

ABSTRACT

A growing body of evidence indicates that clinical use of ketamine as a promising antidepressant can be accompanied by psychotic-like side effects. Although, the generation of such effects is thought to be attributed to dysfunction of prefrontal GABAergic interneurons, the mechanism underlying ketamine's propsychotic-like action is not fully understood. Due to wide spectrum of behavioral abnormalities, it is hypothesized that ketamine action is not limited to only cortical GABA metabolism but may also involve alterations in other functional brain areas. To test it, we treated rats with ketamine (30 mg/kg, i.p.) for 5 days, and next we analyzed GABA metabolizing enzymes in cortex, cerebellum, hippocampus and striatum. Our results demonstrated that diminished GAD67 expression in cortex, cerebellum (by ∼60%) and in hippocampus (by ∼40%) correlated with lowered protein level in these areas. The expression of GAD65 isoform decreased by ∼45% in striatum, but pronounced increase by ∼90% was observed in hippocampus. Consecutively, reduction in glutamate decarboxylase activity and GABA concentration were detected in cortex, cerebellum and striatum, but not in hippocampus. Ketamine administration decreased GABA transaminase protein in cortex and striatum (by ∼50% and 30%, respectively), which was reflected in diminished activity of the enzyme. Also, a significant drop in succinic semialdehyde dehydrogenase activity in cortex, cerebellum and striatum was present. These data suggest a reduced utilization of GABA for energetic purposes. In addition, we observed synaptic GABA release to be reduced by ∼30% from striatal terminals. It correlated with lowered KCl-induced Ca(2+) influx and decreased amount of L-type voltage-dependent calcium channel. Our results indicate that unique changes in GABA metabolism triggered by chronic ketamine treatment in functionally distinct brain regions may be involved in propsychotic-like effects of this drug.


Subject(s)
Brain Chemistry/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Receptors, Presynaptic/drug effects , gamma-Aminobutyric Acid/physiology , Animals , Calcium Channels, L-Type/biosynthesis , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/genetics , GABA Plasma Membrane Transport Proteins/metabolism , Glutamate Decarboxylase/metabolism , Male , Rats , Rats, Wistar , Succinate Dehydrogenase/metabolism , Synapses/drug effects , Synapses/metabolism , Synaptosomes/metabolism , Synaptosomes/ultrastructure , gamma-Aminobutyric Acid/metabolism
20.
Biochem Biophys Res Commun ; 465(2): 312-7, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26278817

ABSTRACT

Ketamine, a high affinity uncompetitive antagonist of voltage-dependent NMDA receptor, has been used for years as a dissociative anesthetic. Although the drug is considered as safe and well-tolerable, it is now evident that it can exert dose-dependent multidirectional effects acting on different cellular targets and pathways. The latest clinical studies also demonstrated its promising antidepressant action. However, the widespread use of this drug in humans is largely limited by a broad range of cognitive adverse effects that resemble some core symptoms of schizophrenia. In line with the hypothesis of unifying role of calcium in schizophrenia symptomology, we used ketamine-induced rat model of experimental psychosis to study the effect of 5-day ketamine treatment (30 mg/kg, ip) on the activity of plasma membrane Ca(2+)-ATPase. Whereas no change in a total amount of the enzyme in cortical synaptosomal membranes was observed, a decrease by ∼50% in hydrolytic activity, as well as lowered phosphointermediate formation were detected. Moreover, ketamine action appeared to be isoform-independent. The experiments on intact Ca(2+)-ATPase purified from vehicle-treated rat cortex revealed dose-dependent inhibition of enzymatic activity. Furthermore, ketamine decreased, but not eliminated, the stimulation by calmodulin. The inhibitory effect, although much weaker, was also evident for truncated form of calcium pump obtained following digestion by trypsin. Our results indicate that plasma membrane Ca(2+)-ATPase is a novel target for ketamine and putative interaction sites may involve central catalytic loop and calmodulin-binding domain.


Subject(s)
Calcium/metabolism , Cell Membrane/drug effects , Cerebral Cortex/drug effects , Plasma Membrane Calcium-Transporting ATPases/metabolism , Schizophrenia/metabolism , Synaptosomes/drug effects , Anesthetics, Dissociative , Animals , Calmodulin/metabolism , Calmodulin/pharmacology , Catalytic Domain , Cell Membrane/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Ion Transport , Isoenzymes/chemistry , Isoenzymes/metabolism , Ketamine , Male , Plasma Membrane Calcium-Transporting ATPases/chemistry , Protein Binding , Protein Structure, Secondary , Rats , Rats, Wistar , Schizophrenia/chemically induced , Schizophrenia/pathology , Synaptosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...