Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.149
Filter
Add more filters

Publication year range
1.
Mol Psychiatry ; 29(3): 704-717, 2024 03.
Article in English | MEDLINE | ID: mdl-38123724

ABSTRACT

The Shank3 gene encodes the major postsynaptic scaffolding protein SHANK3. Its mutation causes a syndromic form of autism spectrum disorder (ASD): Phelan-McDermid Syndrome (PMDS). It is characterized by global developmental delay, intellectual disorders (ID), ASD behavior, affective symptoms, as well as extra-cerebral symptoms. Although Shank3 deficiency causes a variety of molecular alterations, they do not suffice to explain all clinical aspects of this heterogenic syndrome. Since global gene expression alterations in Shank3 deficiency remain inadequately studied, we explored the transcriptome in vitro in primary hippocampal cells from Shank3∆11(-/-) mice, under control and lithium (Li) treatment conditions, and confirmed the findings in vivo. The Shank3∆11(-/-) genotype affected the overall transcriptome. Remarkably, extracellular matrix (ECM) and cell cycle transcriptional programs were disrupted. Accordingly, in the hippocampi of adolescent Shank3∆11(-/-) mice we found proteins of the collagen family and core cell cycle proteins downregulated. In vitro Li treatment of Shank3∆11(-/-) cells had a rescue-like effect on the ECM and cell cycle gene sets. Reversed ECM gene sets were part of a network, regulated by common transcription factors (TF) such as cAMP responsive element binding protein 1 (CREB1) and ß-Catenin (CTNNB1), which are known downstream effectors of synaptic activity and targets of Li. These TFs were less abundant and/or hypo-phosphorylated in hippocampi of Shank3∆11(-/-) mice and could be rescued with Li in vitro and in vivo. Our investigations suggest the ECM compartment and cell cycle genes as new players in the pathophysiology of Shank3 deficiency, and imply involvement of transcriptional regulators, which can be modulated by Li. This work supports Li as potential drug in the management of PMDS symptoms, where a Phase III study is ongoing.


Subject(s)
Extracellular Matrix , Hippocampus , Mice, Knockout , Nerve Tissue Proteins , beta Catenin , Animals , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Hippocampus/metabolism , Extracellular Matrix/metabolism , Mice , beta Catenin/metabolism , beta Catenin/genetics , Chromosome Disorders/genetics , Chromosome Disorders/metabolism , Chromosome Deletion , Cell Cycle/drug effects , Cell Cycle/genetics , Autistic Disorder/genetics , Autistic Disorder/metabolism , Chromosomes, Human, Pair 22/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Male , Transcriptome/genetics , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/drug therapy , Mice, Inbred C57BL , Lithium/pharmacology , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Cells, Cultured
2.
J Cell Mol Med ; 28(10): e18385, 2024 May.
Article in English | MEDLINE | ID: mdl-38801405

ABSTRACT

Autophagy may play an important role in the occurrence and development of glucocorticoid-induced osteonecrosis of the femoral head (GC-ONFH). Lithium is a classical autophagy regulator, and lithium can also activate osteogenic pathways, making it a highly promising therapeutic agent for GC-ONFH. We aimed to evaluate the potential therapeutic effect of lithium on GC-ONFH. For in vitro experiments, primary osteoblasts of rats were used for investigating the underlying mechanism of lithium's protective effect on GC-induced autophagy levels and osteogenic activity dysfunction. For in vivo experiments, a rat model of GC-ONFH was used for evaluating the therapeutic effect of oral lithium on GC-ONFH and underlying mechanism. Findings demonstrated that GC over-activated the autophagy of osteoblasts and reduced their osteogenic activity. Lithium reduced the over-activated autophagy of GC-treated osteoblasts through PI3K/AKT/mTOR signalling pathway and increased their osteogenic activity. Oral lithium reduced the osteonecrosis rates in a rat model of GC-ONFH, and restrained the increased expression of autophagy related proteins in bone tissues through PI3K/AKT/mTOR signalling pathway. In conclusion, lithium can restrain over-activated autophagy by activating PI3K/AKT/mTOR signalling pathway and up-regulate the expression of genes for bone formation both in GC induced osteoblasts and in a rat model of GC-ONFH. Lithium may be a promising therapeutic agent for GC-ONFH. However, the role of autophagy in the pathogenesis of GC-ONFH remains controversial. Studies are still needed to further explore the role of autophagy in the pathogenesis of GC-ONFH, and the efficacy of lithium in the treatment of GC-ONFH and its underlying mechanisms.


Subject(s)
Autophagy , Femur Head Necrosis , Glucocorticoids , Lithium , Osteoblasts , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Autophagy/drug effects , Glucocorticoids/pharmacology , Glucocorticoids/adverse effects , Rats , Femur Head Necrosis/chemically induced , Femur Head Necrosis/pathology , Femur Head Necrosis/drug therapy , Femur Head Necrosis/metabolism , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Lithium/pharmacology , Osteoblasts/drug effects , Osteoblasts/metabolism , Male , Osteogenesis/drug effects , Rats, Sprague-Dawley , Proto-Oncogene Proteins c-akt/metabolism , Disease Models, Animal , Phosphatidylinositol 3-Kinases/metabolism , Femur Head/pathology , Femur Head/drug effects , Femur Head/metabolism , Osteonecrosis/chemically induced , Osteonecrosis/pathology , Osteonecrosis/drug therapy , Osteonecrosis/metabolism , Osteonecrosis/prevention & control
3.
Antimicrob Agents Chemother ; 68(3): e0075623, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38259086

ABSTRACT

Pneumocystis cyst life forms contain abundant ß-glucan carbohydrates, synthesized using ß-1,3 and ß-1,6 glucan synthase enzymes and the donor uridine diphosphate (UDP)-glucose. In yeast, phosphoglucomutase (PGM) plays a crucial role in carbohydrate metabolism by interconverting glucose 1-phosphate and glucose 6-phosphate, a vital step in UDP pools for ß-glucan cell wall formation. This pathway has not yet been defined in Pneumocystis. Herein, we surveyed the Pneumocystis jirovecii and Pneumocystis murina genomes, which predicted a homolog of the Saccharomyces cerevisiae major PGM enzyme. Furthermore, we show that PjPgm2p and PmPgm2p function similarly to the yeast counterpart. When both Pneumocystis pgm2 homologs are heterologously expressed in S. cerevisiae pgm2Δ cells, both genes can restore growth and sedimentation rates to wild-type levels. Additionally, we demonstrate that yeast pgm2Δ cell lysates expressing the two Pneumocystis pgm2 transcripts individually can restore PGM activities significantly altered in the yeast pgm2Δ strain. The addition of lithium, a competitive inhibitor of yeast PGM activity, significantly reduces PGM activity. Next, we tested the effects of lithium on P. murina viability ex vivo and found the compound displays significant anti-Pneumocystis activity. Finally, we demonstrate that a para-aryl derivative (ISFP10) with known inhibitory activity against the Aspergillus fumigatus PGM protein and exhibiting 50-fold selectivity over the human PGM enzyme homolog can also significantly reduce Pmpgm2 activity in vitro. Collectively, our data genetically and functionally validate phosphoglucomutases in both P. jirovecii and P. murina and suggest the potential of this protein as a selective therapeutic target for individuals with Pneumocystis pneumonia.


Subject(s)
Pneumocystis carinii , Pneumocystis , Pneumonia, Pneumocystis , beta-Glucans , Humans , Pneumocystis carinii/genetics , Pneumonia, Pneumocystis/drug therapy , Phosphoglucomutase/genetics , Phosphoglucomutase/metabolism , Phosphoglucomutase/pharmacology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Lithium/metabolism , Lithium/pharmacology , Pneumocystis/genetics , beta-Glucans/metabolism , Phosphates/pharmacology , Glucose/metabolism , Uridine Diphosphate/metabolism , Uridine Diphosphate/pharmacology
4.
Mol Genet Metab ; 141(3): 108140, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38262289

ABSTRACT

Tay-Sachs disease is a rare lysosomal storage disorder (LSD) caused by a mutation in the HexA gene coding ß-hexosaminidase A enzyme. The disruption of the HexA gene causes the accumulation of GM2 ganglioside resulting in progressive neurodegeneration in humans. Surprisingly, Hexa-/- mice did not show neurological phenotypes. Our group recently generated a murine model of Tay-Sachs disease exhibiting excessive GM2 accumulation and severe neuropathological abnormalities mimicking Tay-Sachs patients. Previously, we reported impaired autophagic flux in the brain of Hexa/-Neu3-/- mice. However, regulation of autophagic flux using inducers has not been clarified in Tay-Sachs disease cells. Here, we evaluated the effects of lithium treatment on dysfunctional autophagic flux using LC3 and p62 in the fibroblast and neuroglia of Hexa-/-Neu3-/- mice and Tay-Sachs patients. We discovered the clearance of accumulating autophagosomes, aggregate-prone metabolites, and GM2 ganglioside under lithium-induced conditions. Our data suggest that targeting autophagic flux with an autophagy inducer might be a rational therapeutic strategy for the treatment of Tay-Sachs disease.


Subject(s)
Tay-Sachs Disease , Humans , Mice , Animals , Tay-Sachs Disease/drug therapy , Tay-Sachs Disease/genetics , Lithium/pharmacology , Lithium/therapeutic use , G(M2) Ganglioside , Autophagy , Lithium Compounds/therapeutic use , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism , beta-N-Acetylhexosaminidases/therapeutic use
5.
Mol Psychiatry ; 28(4): 1667-1678, 2023 04.
Article in English | MEDLINE | ID: mdl-36690794

ABSTRACT

Bipolar-disorder's pathophysiology and the mechanism by which medications exert their beneficial effect is yet unknown, but others' and our data implicate patients' brain mitochondrial-dysfunction and its amendment by mood-stabilizers. We recently designed a novel mouse bipolar-disorder-like model using chronic administration of a low-dose of the oxidative-phosphorylation complex I inhibitor, rotenone. Four and eight weeks rotenone treatment induced manic- and depressive-like behavior, respectively, accompanied by mood-related neurochemical changes. Here we aimed to investigate whether each of the autophagy-enhancers lithium (a mood-stabilizer), trehalose and resveratrol and/or each of the reactive oxygen species (ROS)-scavengers, resveratrol and N-acetylcystein and/or the combinations lithium+resveratrol or trehalose+N-acetylcystein, can ameliorate behavioral and neurochemical consequences of neuronal mild mitochondrial-dysfunction. We observed that lithium, trehalose and N-acetylcystein reversed rotenone-induced manic-like behavior as well as deviations in protein levels of mitochondrial complexes and the autophagy marker LC3-II. This raises the possibility that mild mitochondrial-dysfunction accompanied by impaired autophagy and a very mild increase in ROS levels are related to predisposition to manic-like behavior. On the other hand, although, as expected, most of the drugs tested eliminated the eight weeks rotenone-induced increase in protein levels of all hippocampal mitochondrial complexes, only lithium ubiquitously ameliorated the depressive-like behaviors. We cautiously deduce that aberrant autophagy and/or elevated ROS levels are not involved in predisposition to the depressive phase of bipolar-like behavior. Rather, that amending the depressive-like characteristics requires different mitochondria-related interventions. The latter might be antagonizing N-methyl-D-aspartate receptors (NMDARs), thus protecting from disruption of mitochondrial calcium homeostasis and its detrimental consequences. In conclusion, our findings suggest that by-and-large, among the autophagy-enhancers and ROS-scavengers tested, lithium is the most effective in counteracting rotenone-induced changes. Trehalose and N-acetylcystein may also be effective in attenuating manic-like behavior.


Subject(s)
Brain Diseases , Lithium , Animals , Mice , Lithium/pharmacology , Reactive Oxygen Species , Resveratrol , Rotenone , Trehalose , Autophagy , Mitochondria
6.
Mol Psychiatry ; 28(7): 3033-3043, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36653674

ABSTRACT

Lithium (Li) is recommended for long-term treatment of bipolar disorder (BD). However, its mechanism of action is still poorly understood. Induced pluripotent stem cell (iPSC)-derived brain organoids have emerged as a powerful tool for modeling BD-related disease mechanisms. We studied the effects of 1 mM Li treatment for 1 month in iPSC-derived human cortical spheroids (hCS) from 10 healthy controls (CTRL) and 11 BD patients (6 Li-responders, Li-R, and 5 Li non-treated, Li-N). At day 180 of differentiation, BD hCS showed smaller size, reduced proportion of neurons, decreased neuronal excitability and reduced neural network activity compared to CTRL hCS. Li rescued excitability of BD hCS neurons by exerting an opposite effect in the two diagnostic groups, increasing excitability in BD hCS and decreasing it in CTRL hCS. We identified 132 Li-associated differentially expressed genes (DEGs), which were overrepresented in sodium ion homeostasis and kidney-related pathways. Moreover, Li regulated secretion of pro-inflammatory cytokines and increased mitochondrial reserve capacity in BD hCS. Through long-term Li treatment of a human 3D brain model, this study partly elucidates the functional and transcriptional mechanisms underlying the clinical effects of Li, such as rescue of neuronal excitability and neuroprotection. Our results also underscore the substantial influence of treatment duration in Li studies. Lastly, this study illustrates the potential of patient iPSC-derived 3D brain models for precision medicine in psychiatry.


Subject(s)
Bipolar Disorder , Induced Pluripotent Stem Cells , Humans , Lithium/pharmacology , Lithium/therapeutic use , Lithium/metabolism , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Induced Pluripotent Stem Cells/metabolism , Lithium Compounds/therapeutic use , Neurons/metabolism
7.
Int J Med Sci ; 21(2): 284-298, 2024.
Article in English | MEDLINE | ID: mdl-38169754

ABSTRACT

Ischemic stroke ranks among the foremost clinical causes of mortality and disability, instigating neuronal degeneration, fatalities, and various sequelae. While standard treatments, such as intravenous thrombolysis and endovascular thrombectomy, prove effective, they come with limitations. Hence, there is a compelling need to develop neuroprotective agents capable of improving the functional outcomes of the nervous system. Numerous preclinical studies have demonstrated that lithium can act in multiple molecular pathways, including glycogen synthase kinase 3(GSK-3), the Wnt signaling pathway, the mitogen-activated protein kinase (MAPK)/ extracellular signal-regulated kinase (ERK) signaling pathway, brain-derived neurotrophic factor (BDNF), mammalian target of rapamycin (mTOR), and glutamate receptors. Through these pathways, lithium has been shown to affect inflammation, autophagy, apoptosis, ferroptosis, excitotoxicity, and other pathological processes, thereby improving central nervous system (CNS) damage caused by ischemic stroke. Despite these promising preclinical findings, the number of clinical trials exploring lithium's efficacy remains limited. Additional trials are imperative to thoroughly ascertain the effectiveness and safety of lithium in clinical settings. This review delineates the mechanisms underpinning lithium's neuroprotective capabilities in the context of ischemic stroke. It elucidates the intricate interplay between these mechanisms and sheds light on the involvement of mitochondrial dysfunction and inflammatory markers in the pathophysiology of ischemic stroke. Furthermore, the review offers directions for future research, thereby advancing the understanding of the potential therapeutic utility of lithium and establishing a theoretical foundation for its clinical application.


Subject(s)
Ischemic Stroke , Neuroprotective Agents , Humans , Lithium/pharmacology , Lithium/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Ischemic Stroke/drug therapy , Glycogen Synthase Kinase 3 , Apoptosis
8.
PLoS Genet ; 17(4): e1009484, 2021 04.
Article in English | MEDLINE | ID: mdl-33886537

ABSTRACT

Primary cilia are microtubule-based organelles present on most cells that regulate many physiological processes, ranging from maintaining energy homeostasis to renal function. However, the role of these structures in the regulation of behavior remains unknown. To study the role of cilia in behavior, we employ mouse models of the human ciliopathy, Bardet-Biedl Syndrome (BBS). Here, we demonstrate that BBS mice have significant impairments in context fear conditioning, a form of associative learning. Moreover, we show that postnatal deletion of BBS gene function, as well as congenital deletion, specifically in the forebrain, impairs context fear conditioning. Analyses indicated that these behavioral impairments are not the result of impaired hippocampal long-term potentiation. However, our results indicate that these behavioral impairments are the result of impaired hippocampal neurogenesis. Two-week treatment with lithium chloride partially restores the proliferation of hippocampal neurons which leads to a rescue of context fear conditioning. Overall, our results identify a novel role of cilia genes in hippocampal neurogenesis and long-term context fear conditioning.


Subject(s)
Bardet-Biedl Syndrome/genetics , Fear/drug effects , Neurogenesis/drug effects , Neurons/metabolism , Animals , Bardet-Biedl Syndrome/drug therapy , Bardet-Biedl Syndrome/pathology , Cell Proliferation/drug effects , Cilia/genetics , Cilia/metabolism , Cilia/pathology , Disease Models, Animal , Fear/physiology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Lithium/pharmacology , Memory Disorders/drug therapy , Memory Disorders/genetics , Memory Disorders/pathology , Mice , Microtubule-Associated Proteins/genetics , Neurogenesis/genetics , Neurons/pathology
9.
Gen Physiol Biophys ; 43(3): 263-271, 2024 May.
Article in English | MEDLINE | ID: mdl-38774925

ABSTRACT

Lithium (Li) is a mood-stabilizing drug. Although one of the potential mechanisms underlying the neuroprotective effects of lithium is related to its antioxidative effect, its mechanisms of action are not fully understood. Herein we aimed to investigate the impact of varied dosages of long-term lithium therapy on oxidative stress parameters in the brains of healthy rats, and on anxiety-like behaviors, and whether any changes in behavior can be attributed to modifications in oxidative stress levels within the brain. Thirty-two adult Wistar albino male rats were randomly assigned to four treatment groups. While the control (C) group was fed with a standard diet, low Li (1.4 g/kg/diet), moderate Li (1.8 g/kg/diet), and high Li (2.2 g/kg/diet) groups were fed with lithium bicarbonate (Li2CO3) for 30 days. Malondialdehyde increased, while superoxide dismutase and catalase levels decreased in the brains of the high Li group animals. In addition, anxiety-like behaviors of animals increased in the high Li group considering fewer entries to and less time spent in the open arms of the elevated plus maze test. Our findings underscore the potential adverse effects of prolonged lithium treatment, especially at doses approaching the upper therapeutic range. The induction of toxicity, manifested through heightened oxidative stress, appears to be a key mechanism contributing to the observed increase in anxiety-like behaviors. Consequently, caution is warranted when considering extended lithium therapy at higher doses, emphasizing the need for further research to delineate the precise mechanisms underlying these effects and to inform safer therapeutic practices.


Subject(s)
Anxiety , Brain , Dose-Response Relationship, Drug , Oxidative Stress , Rats, Wistar , Animals , Oxidative Stress/drug effects , Male , Rats , Anxiety/chemically induced , Anxiety/drug therapy , Brain/drug effects , Brain/metabolism , Lithium/pharmacology , Lithium/administration & dosage , Behavior, Animal/drug effects , Drug Administration Schedule , Lithium Compounds/pharmacology , Lithium Compounds/administration & dosage
10.
Arch Pharm (Weinheim) ; 357(8): e2400063, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38704748

ABSTRACT

Lithium induces nephrogenic diabetes insipidus (NDI) and microcystic chronic kidney disease (CKD). As previous clinical studies suggest that NDI is dose-dependent and CKD is time-dependent, we investigated the effect of low exposition to lithium in a long-term experimental rat model. Rats were fed with a normal diet (control group), with the addition of lithium (Li+ group), or with lithium and amiloride (Li+/Ami group) for 6 months, allowing obtaining low plasma lithium concentrations (0.25 ± 0.06 and 0.43 ± 0.16 mmol/L, respectively). Exposition to low concentrations of plasma lithium levels prevented NDI but not microcystic dilations of kidney tubules, which were identified as collecting ducts (CDs) on immunofluorescent staining. Both hypertrophy, characterized by an increase in the ratio of nuclei per tubular area, and microcystic dilations were observed. The ratio between principal cells and intercalated cells was higher in microcystic than in hypertrophied tubules. There was no correlation between AQP2 messenger RNA levels and cellular remodeling of the CD. Additional amiloride treatment in the Li+/Ami group did not allow consistent morphometric and cellular composition changes compared to the Li+ group. Low exposition to lithium prevented overt NDI but not microcystic dilations of the CD, with differential cellular composition in hypertrophied and microcystic CDs, suggesting different underlying cellular mechanisms.


Subject(s)
Amiloride , Aquaporin 2 , Diabetes Insipidus, Nephrogenic , Disease Models, Animal , Kidney Tubules, Collecting , Animals , Diabetes Insipidus, Nephrogenic/chemically induced , Diabetes Insipidus, Nephrogenic/prevention & control , Kidney Tubules, Collecting/drug effects , Kidney Tubules, Collecting/pathology , Kidney Tubules, Collecting/metabolism , Male , Rats , Aquaporin 2/metabolism , Amiloride/pharmacology , Rats, Wistar , Time Factors , Renal Insufficiency, Chronic/prevention & control , Renal Insufficiency, Chronic/chemically induced , Lithium/pharmacology , Dose-Response Relationship, Drug
11.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062811

ABSTRACT

Epilepsy is known to cause alterations in neural networks. However, many details of these changes remain poorly understood. The objective of this study was to investigate changes in the properties of hippocampal CA1 pyramidal neurons and their synaptic inputs in a rat lithium-pilocarpine model of epilepsy. In the chronic phase of the model, we found a marked loss of pyramidal neurons in the CA1 area. However, the membrane properties of the neurons remained essentially unaltered. The results of the electrophysiological and morphological studies indicate that the direct pathway from the entorhinal cortex to CA1 neurons is reinforced in epileptic animals, whereas the inputs to them from CA3 are either unaltered or even diminished. In particular, the dendritic spine density in the str. lacunosum moleculare, where the direct pathway from the entorhinal cortex terminates, was found to be 2.5 times higher in epileptic rats than in control rats. Furthermore, the summation of responses upon stimulation of the temporoammonic pathway was enhanced by approximately twofold in epileptic rats. This enhancement is believed to be a significant contributing factor to the heightened epileptic activity observed in the entorhinal cortex of epileptic rats using an ex vivo 4-aminopyridine model.


Subject(s)
CA1 Region, Hippocampal , Disease Models, Animal , Epilepsy , Lithium , Pilocarpine , Pyramidal Cells , Animals , Pyramidal Cells/pathology , Pyramidal Cells/metabolism , Rats , Epilepsy/chemically induced , Epilepsy/pathology , Epilepsy/physiopathology , Male , CA1 Region, Hippocampal/pathology , Lithium/toxicity , Lithium/pharmacology , Entorhinal Cortex/pathology , Rats, Wistar
12.
Hum Genomics ; 16(1): 45, 2022 10 17.
Article in English | MEDLINE | ID: mdl-36253798

ABSTRACT

BACKGROUND: It has been suggested that bipolar disorder (BD) is associated with clinical and biological features of accelerated aging. In our previous studies, we showed that long-term lithium treatment was correlated with longer leukocyte telomere length (LTL) in BD patients. A recent study explored the role of TL in BD using patients-derived lymphoblastoid cell lines (LCLs), showing that baseline TL was shorter in BD compared to controls and that lithium in vitro increased TL but only in BD. Here, we used the same cell system (LCLs) to explore if a 7-day treatment protocol with lithium chloride (LiCl) 1 mM was able to highlight differences in TL between BD patients clinically responders (Li-R; n = 15) or non-responders (Li-NR; n = 15) to lithium, and if BD differed from non-psychiatric controls (HC; n = 15). RESULTS: There was no difference in TL between BD patients and HC. Moreover, LiCl did not influence TL in the overall sample, and there was no difference between diagnostic or clinical response groups. Likewise, LiCl did not affect TL in neural precursor cells from healthy donors. CONCLUSIONS: Our findings suggest that a 7-day lithium treatment protocol and the use of LCLs might not represent a suitable approach to deepen our understanding on the role of altered telomere dynamics in BD as previously suggested by studies in vivo.


Subject(s)
Bipolar Disorder , Neural Stem Cells , Bipolar Disorder/diagnosis , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Cell Line , Humans , Lithium/pharmacology , Lithium/therapeutic use , Lithium Chloride/pharmacology , Lithium Chloride/therapeutic use , Lithium Compounds/pharmacology , Lithium Compounds/therapeutic use , Neural Stem Cells/metabolism , Telomere/genetics
13.
Neurochem Res ; 48(12): 3538-3559, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37526866

ABSTRACT

Chronic exposure to stress is a non-adaptive situation that is associated with mitochondrial dysfunction and the accumulation of reactive oxygen species (ROS), especially superoxide anion (SA). This accumulation of ROS produces damage-associated molecular patterns (DAMPs), which activate chronic inflammatory states and behavioral changes found in several mood disorders. In a previous study, we observed that an imbalance of SA triggered by rotenone (Ro) exposure caused evolutionarily conserved oxi-inflammatory disturbances and behavioral changes in Eisenia fetida earthworms. These results supported our hypothesis that SA imbalance triggered by Ro exposure could be attenuated by lithium carbonate (LC), which has anti-inflammatory properties. The initial protocol exposed earthworms to Ro (30 nM) and four different LC concentrations. LC at a concentration of 12.85 mg/L decreased SA and nitric oxide (NO) levels and was chosen to perform complementary assays: (1) neuromuscular damage evaluated by optical and scanning electron microscopy (SEM), (2) innate immune inefficiency by analysis of Eisenia spp. extracellular neutrophil traps (eNETs), and (3) behavioral changes. Gene expression was also evaluated involving mitochondrial (COII, ND1), inflammatory (EaTLR, AMP), and neuronal transmission (nAchR α5). LC attenuated the high melanized deposits in the circular musculature, fiber disarrangement, destruction of secretory glands, immune inefficiency, and impulsive behavior pattern triggered by Ro exposure. However, the effects of LC and Ro on gene expression were more heterogeneous. In summary, SA imbalance, potentially associated with mitochondrial dysfunction, appears to be an evolutionary component triggering oxidative, inflammatory, and behavioral changes observed in psychiatric disorders that are inhibited by LC exposure.


Subject(s)
Oligochaeta , Oxidative Stress , Humans , Animals , Reactive Oxygen Species/metabolism , Oligochaeta/genetics , Oligochaeta/metabolism , Lithium/pharmacology , Rotenone/toxicity , Superoxides/metabolism , Brain/metabolism , Superoxide Dismutase/metabolism , Catalase/metabolism
14.
Eur Biophys J ; 52(1-2): 91-100, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36929427

ABSTRACT

Lithium has been the treatment of choice for patients with bipolar disorder. However, lithium overdose happens more frequently since it has a very narrow therapeutic range in blood, necessitating investigation of its adverse effects on blood cells. The possible changes that lithium exposure may have on functional and morphological characteristics of human red blood cells (RBCs) have been studied ex vivo using single-cell Raman spectroscopy, optical trapping, and membrane fluorescent probe. The Raman spectroscopy was performed with excitation at 532 nm light, which also results in simultaneous photoreduction of intracellular hemoglobin (Hb). The level of photoreduction of lithium-exposed RBCs was observed to decline with lithium concentration, indicating irreversible oxygenation of intracellular Hb from lithium exposure. The lithium exposure may also have an effect on RBC membrane, which was investigated via optical stretching in a laser trap and the results suggest lower membrane fluidity for the lithium-exposed RBCs. The membrane fluidity of RBCs was further studied using the Prodan generalized polarization method and the results verify the reduction of membrane fluidity upon lithium exposure.


Subject(s)
Erythrocytes , Lithium , Humans , Lithium/pharmacology , Lithium/analysis , Erythrocytes/chemistry , Hemoglobins , Lasers , Spectrum Analysis, Raman
15.
Mol Biol Rep ; 50(2): 1617-1625, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36526850

ABSTRACT

BACKGROUND: The application of neuroprotective agents in combination with stem cells is considered a potential effective treatment for multiple sclerosis (MS). Therefore, the effects of lithium chloride as a neuroprotective agent and a GSK3-ß inhibitor were evaluated in combination with human adipose derived stem cells on re-myelination, oligodendrocyte differentiation, and functional recovery. METHODS: After inducing a mouse model of MS and proving it by the hanging wire test, the mice were randomly assigned to five experimental groups: Cup, Sham, Li, hADSC, and Li + hADSC. Additionally, a control group with normal feeding was considered. Finally, toluidine blue staining was carried out to estimate the level of myelination. Furthermore, immunofluorescent staining was used to evaluate the mean of OLIG2 and MOG positive cells. The mRNA levels of ß-Catenin, myelin and oligodendrocyte specific genes were determined via the Real-Time PCR. RESULTS: The results of the hanging wire test and toluidine blue staining showed a significant increase in myelin density and improvements in motor function in groups, which received lithium and stem cells, particularly in the Li + hADSC group compared with the untreated groups (P < 0.01). Moreover, immunostaining results indicated that the mean percentages of MOG and OLIG2 positive cells were significantly higher in the Li + hADSC group than in the other groups (P < 0.01). Finally, gene expression studies indicated that the use of lithium could increase the expression of ß-Catenin, myelin and oligodendrocyte specific genes. CONCLUSION: The use of Lithium Chloride can increase stem cells differentiation into oligodendrocytes and improve re-myelination in MS.


Subject(s)
Multiple Sclerosis , Animals , Humans , Mice , beta Catenin/metabolism , Cell Differentiation , Disease Models, Animal , Glycogen Synthase Kinase 3/metabolism , Lithium/pharmacology , Lithium Chloride/pharmacology , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Oligodendroglia/metabolism , Stem Cells/metabolism , Tolonium Chloride/metabolism , Tolonium Chloride/pharmacology , Enzyme Inhibitors/pharmacology
16.
Biochemistry (Mosc) ; 88(3): 353-363, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37076282

ABSTRACT

Status epilepticus (SE) triggers many not yet fully understood pathological changes in the nervous system that can lead to the development of epilepsy. In this work, we studied the effects of SE on the properties of excitatory glutamatergic transmission in the hippocampus in the lithium-pilocarpine model of temporal lobe epilepsy in rats. The studies were performed 1 day (acute phase), 3 and 7 days (latent phase), and 30 to 80 days (chronic phase) after SE. According to RT-qPCR data, expression of the genes coding for the AMPA receptor subunits GluA1 and GluA2 was downregulated in the latent phase, which may lead to the increased proportion of calcium-permeable AMPA receptors that play an essential role in the pathogenesis of many CNS diseases. The efficiency of excitatory synaptic neurotransmission in acute brain slices was decreased in all phases of the model, as determined by recording field responses in the CA1 region of the hippocampus in response to the stimulation of Schaffer collaterals by electric current of different strengths. However, the frequency of spontaneous excitatory postsynaptic potentials increased in the chronic phase, indicating an increased background activity of the glutamatergic system in epilepsy. This was also evidenced by a decrease in the threshold current causing hindlimb extension in the maximal electroshock seizure threshold test in rats with temporal lobe epilepsy compared to the control animals. The results suggest a series of functional changes in the properties of glutamatergic system associated with the epilepsy development and can be used to develop the antiepileptogenic therapy.


Subject(s)
Epilepsy, Temporal Lobe , Epilepsy , Status Epilepticus , Rats , Animals , Pilocarpine/toxicity , Pilocarpine/metabolism , Epilepsy, Temporal Lobe/metabolism , Lithium/pharmacology , Lithium/metabolism , Hippocampus/metabolism , Epilepsy/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Disease Models, Animal
17.
Int J Mol Sci ; 24(17)2023 Sep 02.
Article in English | MEDLINE | ID: mdl-37686419

ABSTRACT

Hydrogels have gained significant attention as biomaterials due to their remarkable properties resembling those of the extracellular matrix (ECM). In the present investigation, we successfully synthesized interpenetrating polymer network (IPN) hydrogels using gelatin methacryloyl (GelMA) and sodium alginate (SA), incorporating various concentrations of lithium chloride (LiCl; 0, 5, and 10 mM), aiming to develop a hydrogel scaffold for bone regeneration. Notably, the compressive modulus of the IPN hydrogels remained largely unaffected upon the inclusion of LiCl. However, the hydrogel with the high concentration of LiCl exhibited reduced fragmentation after compression testing. Intriguingly, we observed a significant improvement in cellular biocompatibility, primarily attributed to activation of the Wnt/ß-catenin signaling pathway induced by LiCl. Subsequently, we evaluated the efficacy of the newly developed IPN-Li hydrogels in a rat cranial defect model and found that they substantially enhanced bone regeneration. Nevertheless, it is important to note that the introduction of high concentrations of LiCl did not significantly promote osteogenesis. This outcome can be attributed to the excessive release of Li+ ions into the extracellular matrix, hindering the desired effect. Overall, the IPN-Li hydrogel developed in this study holds great promise as a biodegradable material for bone regeneration applications.


Subject(s)
Lithium , Wnt Signaling Pathway , Animals , Rats , Alginates/pharmacology , Bone Regeneration , Hydrogels/pharmacology , Lithium/metabolism , Lithium/pharmacology , Polymers
18.
Int J Mol Sci ; 24(21)2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37958600

ABSTRACT

Serotonin 1A (5-HT1A) autoreceptors located on serotonin neurons inhibit their activity, and their upregulation has been implicated in depression, suicide and resistance to antidepressant treatment. Conversely, post-synaptic 5-HT1A heteroreceptors are important for antidepressant response. The transcription factor deformed epidermal autoregulatory factor 1 (Deaf1) acts as a presynaptic repressor and postsynaptic enhancer of 5-HT1A transcription, but the mechanism is unclear. Because Deaf1 interacts with and is phosphorylated by glycogen synthase kinase 3ß (GSK3ß)-a constitutively active protein kinase that is inhibited by the mood stabilizer lithium at therapeutic concentrations-we investigated the role of GSK3ß in Deaf1 regulation of human 5-HT1A transcription. In 5-HT1A promoter-reporter assays, human HEK293 kidney and 5-HT1A-expressing SKN-SH neuroblastoma cells, transfection of Deaf1 reduced 5-HT1A promoter activity by ~45%. To identify potential GSK3ß site(s) on Deaf1, point mutations of known and predicted phosphorylation sites on Deaf1 were tested. Deaf1 repressor function was not affected by any of the mutants tested except the Y300F mutant, which augmented Deaf1 repression. Both lithium and the selective GSK3 inhibitors CHIR-99021 and AR-014418 attenuated and reversed Deaf1 repression compared to vector. This inhibition was at concentrations that maximally inhibit GSK3ß activity as detected by the GSK3ß-sensitive TCF/LEF reporter construct. Our results support the hypothesis that GSK3ß regulates the activity of Deaf1 to repress 5-HT1A transcription and provide a potential mechanism for actions of GSK3 inhibitors on behavior.


Subject(s)
Glycogen Synthase Kinase 3 beta , Lithium , Receptor, Serotonin, 5-HT1A , Serotonin , Humans , Antidepressive Agents , Glycogen Synthase Kinase 3 beta/genetics , HEK293 Cells , Lithium/pharmacology , Receptor, Serotonin, 5-HT1A/genetics , Serotonin/pharmacology
19.
Int J Mol Sci ; 25(1)2023 Dec 24.
Article in English | MEDLINE | ID: mdl-38203457

ABSTRACT

The structure, antioxidant and neuroprotective properties of lithium comenate (lithium 5-hydroxy-4-oxo-4H-pyran-2-carboxylate) were studied. Lithium comenate was obtained by reacting comenic acid (H2Com) with lithium hydroxide in an aqueous solution. The structure of lithium comenate was confirmed via thermal analysis, mass spectrometry, IR, NMR and UV spectroscopy. The crystal structure was studied in detail via X-ray diffraction. The compound crystallized in a non-centrosymmetric space group of symmetry of the orthorhombic system Pna21 in the form of a hydrate, with three water molecules entering the first coordination sphere of the cation Li+ and one molecule forming a second environment through non-valent contacts. The gross formula of the complex compound was established [Li(HCom)(H2O)3]·H2O. It has been established that lithium comenate has a pronounced neuroprotective activity under the excitotoxic effect of glutamate, increasing the survival rate of cultured rat cerebellar neurons more than two-fold. It has also been found that the pre-stress use of lithium comenate at doses of 1 and 2 mg/kg has an antioxidant effect, which is manifested in a decrease in oxidative damage to the brain tissues of mice subjected to immobilization stress. Based on the data available in the literature, we believe that the high neuroprotective and antioxidant efficacy of lithium comenate is a consequence of the mutual potentiation of the pharmacological effects of lithium and comenic acid.


Subject(s)
Antioxidants , Carboxylic Acids , Lithium , Pyrones , Radioisotopes , Animals , Mice , Rats , Lithium/pharmacology , Antioxidants/pharmacology , Glutamic Acid , Water
20.
Int J Mol Sci ; 24(9)2023 May 08.
Article in English | MEDLINE | ID: mdl-37176158

ABSTRACT

Epilepsy is a challenging brain disorder that is often difficult to treat with conventional therapies. The gut microbiota has been shown to play an important role in the development of neuropsychiatric disorders, including epilepsy. In this study, the effects of Bifidobacterium longum, a probiotic, on inflammation, neuronal degeneration, and behavior are evaluated in a lithium-pilocarpine model of temporal lobe epilepsy (TLE) induced in young adult rats. B. longum was administered orally at a dose of 109 CFU/rat for 30 days after pilocarpine injection. The results show that B. longum treatment has beneficial effects on the TLE-induced changes in anxiety levels, neuronal death in the amygdala, and body weight recovery. In addition, B. longum increased the expression of anti-inflammatory and neuroprotective genes, such as Il1rn and Pparg. However, the probiotic had little effect on TLE-induced astrogliosis and microgliosis and did not reduce neuronal death in the hippocampus and temporal cortex. The study suggests that B. longum may have a beneficial effect on TLE and may provide valuable insights into the role of gut bacteria in epileptogenesis. In addition, the results show that B. longum may be a promising drug for the comprehensive treatment of epilepsy.


Subject(s)
Bifidobacterium longum , Epilepsy, Temporal Lobe , Epilepsy , Probiotics , Rats , Animals , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/metabolism , Pilocarpine/adverse effects , Lithium/pharmacology , Hippocampus/metabolism , Epilepsy/metabolism , Probiotics/pharmacology , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL