Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.156
Filtrar
1.
J Vet Sci ; 25(5): e69, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39363657

RESUMEN

IMPORTANCE: Guillain-Barré syndrome (GBS)-like neuropathy mimics the leading cause of sporadic acute nontraumatic limb paralysis in individuals from developed countries. Experimental autoimmune neuritis (EAN) is an animal model of GBS and of syndromes such as acute canine polyradiculoneuritis, seen in dogs and cats. OBJECTIVE: The involvement of glycogen synthase kinase (GSK)-3ß, a pro-inflammatory molecule, in rat EAN is not fully understood. This study evaluated the potential role of GSK-3ß in EAN through its inhibition by lithium. METHODS: Lewis rats were injected with SP26 antigen to induce EAN. Lithium was administered from 1 day before immunization to day 14 post-immunization (PI). Then the rats were euthanized and their neural tissues were prepared for histological and Western blotting analyses. RESULTS: Lithium, an inhibitor of GSK-3, significantly ameliorated EAN paralysis in rats, when administered from day 1 to day 14 PI. This corresponded with reduced inflammation in the sciatic nerves of EAN rats, where phosphorylation of GSK-3ß was also upregulated, indicating suppression of GSK-3. CONCLUSIONS AND RELEVANCE: These findings suggest that lithium, an inhibitor of GSK-3ß, plays a significant role in ameliorating rat EAN paralysis, by suppressing GSK-3ß and its related signals in EAN-affected sciatic nerves.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta , Neuritis Autoinmune Experimental , Ratas Endogámicas Lew , Animales , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Neuritis Autoinmune Experimental/tratamiento farmacológico , Ratas , Masculino , Parálisis/tratamiento farmacológico , Parálisis/veterinaria , Litio/uso terapéutico , Litio/farmacología , Nervio Ciático/efectos de los fármacos
2.
ACS Biomater Sci Eng ; 10(10): 6377-6396, 2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39259706

RESUMEN

Clinical oncology is currently experiencing a technology bottleneck due to the expeditious evolution of therapy defiance in tumors. Although drugs used in chemotherapy work for a sort of cell death with potential clinical application, the effectiveness of chemotherapy-inducing drugs is subject to several endogenous conditions when used alone, necessitating the urgent need for controlled mechanisms. A tumor-targeted drug delivery therapy using Li-Al (M+/M3+)-based layered double hydroxide (LDHs) family has been proposed with the general chemical formula [M+1-x M3+x (OH)]2x+[(Am-)2x/m. n(H2O)]2x-, which is fully biodegradable and works in connection with the therapeutic interaction between LDH nanocarriers and anticancerous doxorubicin (DOX). Compositional variation of Li and Al in LDHs has been used as a nanoplatform, which provides a functional balance between circulation lifetime, drug loading capacity, encapsulation efficiency, and tumor-specific uptake to act as self-regulatory therapeutic cargo to be released intracellularly. First-principle analyses based on DFT have been employed to investigate the interaction of bonding and electronic structure of LDH with DOX and assess its capability and potential for a superior drug carrier. Following the internalization into cancer cells, nanoformulations are carried to the nucleus via lysosomes, and the mechanistic pathways have been revealed. Additionally, in vitro along with in vivo therapeutic assessments on melanoma-bearing mice show a dimensional effect of nanoformulation for better biocompatibility and excellent synergetic anticancer activity. Further, the severe toxic consequences associated with traditional chemotherapy have been eradicated by using injectable hydrogel placed just beneath the tumor site, and regulated release of the drug has been confirmed through protein expression applying various markers. However, Li-Al-based LDH nanocarriers open up new design options for multifunctional nanomedicine, which has intriguing potential for use in cancer treatment through sustained drug delivery.


Asunto(s)
Doxorrubicina , Hidróxidos , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Doxorrubicina/química , Doxorrubicina/administración & dosificación , Animales , Concentración de Iones de Hidrógeno , Hidróxidos/química , Humanos , Ratones , Litio/química , Litio/farmacología , Litio/uso terapéutico , Portadores de Fármacos/química , Nanopartículas/química , Nanopartículas/uso terapéutico , Sistemas de Liberación de Medicamentos , Línea Celular Tumoral , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/uso terapéutico , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antineoplásicos/administración & dosificación
3.
Biomolecules ; 14(8)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39199293

RESUMEN

Lithium is one of the lightest elements on Earth and it has been in the environment since the formation of the galaxy. While a common element, it has not been found to be an essential element in biological processes, ranging from single cell organisms to Homo sapiens. Instead, at an early stage of evolution, organisms committed to a range of elements such as sodium, potassium, calcium, magnesium, zinc, and iron to serve essential functions. Such ions serve critical functions in ion channels, as co-factors in enzymes, as a cofactor in oxygen transport, in DNA replication, as a storage molecule in bone and liver, and in a variety of other roles in biological processes. While seemingly excluded from a major essential role in such processes, lithium ions appear to be able to modulate a variety of biological processes and "correct" deviation from normal activity, as a deficiency of lithium can have biological consequences. Lithium salts are found in low levels in many foods and water supplies, but the effectiveness of Li salts to affect biological systems came to recent prominence with the work of Cade, who reported that administrating Li salts calmed guinea pigs and was subsequently effective at relatively high doses to "normalize" a subset of patients with bipolar disorders. Because of its ability to modulate many biological pathways and processes (e.g., cyclic AMP, GSK-3beta, inositol metabolism, NaK ATPases, neuro processes and centers, immune-related events, respectively) both in vitro and in vivo and during development and adult life, Li salts have become both a useful tool to better understand the molecular regulation of such processes and to also provide insights into altered biological processes in vivo during aging and in disease states. While the range of targets for lithium action supports its possible role as a modulator of biological dysregulation, it presents a conundrum for researchers attempting to elucidate its specific primary target in different tissues in vivo. This review will discuss aspects of the state of knowledge regarding some of the systems that can be influenced, focusing on those involving neural and autoimmunity as examples, some of the mechanisms involved, examples of how Li salts can be used to study model systems, as well as suggesting areas where the use of Li salts could lead to additional insights into both disease mechanisms and natural processes at the molecular and cell levels. In addition, caveats regarding lithium doses used, the strengths and weaknesses of rodent models, the background genetics of the strain of mice or rats employed, and the sex of the animals or the cells used, are discussed. Low-dose lithium may have excellent potential, alone or in combination with other interventions to prevent or alleviate aging-associated conditions and disease progression.


Asunto(s)
Envejecimiento , Litio , Humanos , Animales , Envejecimiento/efectos de los fármacos , Envejecimiento/metabolismo , Litio/farmacología
4.
ACS Appl Bio Mater ; 7(9): 6101-6113, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39121349

RESUMEN

Bacterial infections and biofilm growth are common mishaps associated with medical devices, and they contribute significantly to ill health and mortality. Removal of bacterial deposition from these devices is a major challenge, resulting in an immediate necessity for developing antibacterial coatings on the surfaces of medical implants. In this context, we developed an innovative coating strategy that can operate at low temperatures (80 °C) and preserve the devices' integrity and functionality. An innovative Ag-TiO2 based coating was developed by ion exchange between silver nitrate (AgNO3) and lithium titanate (Li4Ti5O12) on glass substrates for different periods, ranging from 10 to 60 min. The differently coated samples were tested for their antibacterial and antibiofilm efficacy.


Asunto(s)
Antibacterianos , Biopelículas , Materiales Biocompatibles Revestidos , Litio , Ensayo de Materiales , Pruebas de Sensibilidad Microbiana , Tamaño de la Partícula , Plata , Titanio , Titanio/química , Titanio/farmacología , Biopelículas/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Litio/química , Litio/farmacología , Plata/química , Plata/farmacología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Propiedades de Superficie , Nanopartículas del Metal/química , Staphylococcus aureus/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Nanopartículas/química
5.
J Mater Chem B ; 12(38): 9575-9591, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39210776

RESUMEN

In this study, we have developed unique bioresorbable lithiated nanoparticles (LiCP, d50 = 20 nm), demonstrating a versatile material for bone repair and regeneration applications. The LiCPs are biocompatible even at the highest concentration tested (1000 µg mL-1) where bone marrow derived mesenchymal stem cells (BM-MSCs) maintained over 90% viability compared to the control. Notably, LiCP significantly enhanced the expression of osteogenic and angiogenic markers in vitro; collagen I, Runx2, angiogenin, and EGF increased by 8-fold, 8-fold, 9-fold, and 7.5-fold, respectively. Additionally, LiCP facilitated a marked improvement in tubulogenesis in endothelial cells across all tested concentrations. Remarkably, in an ectopic mouse model, LiCP induced mature bone formation, outperforming both the control group and non-lithiated nanoparticles. These findings establish lithiated nanoparticles as a highly promising material for advancing bone repair and regeneration therapies, offering dual benefits in osteogenesis and angiogenesis. The results lay the groundwork for future studies and potential clinical applications, where precise modulation of lithium release could tailor therapeutic outcomes to meet specific patient needs in bone and vascular tissue engineering.


Asunto(s)
Litio , Nanopartículas , Neovascularización Fisiológica , Osteogénesis , Osteogénesis/efectos de los fármacos , Animales , Litio/química , Litio/farmacología , Nanopartículas/química , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Humanos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Tamaño de la Partícula , Células Cultivadas
6.
Plant Physiol Biochem ; 215: 109062, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39178803

RESUMEN

With progress in technology, soaring demand for lithium (Li) has led to its release into the environment. This study demonstrated the mitigation of the adverse effects of Li stress on tomato (Solanum lycopersicum L.) by the application of waste materials, namely coconut shell biochar (CBC) and steel slag (SS). To explore the impact of Li treatment on tomato plants different morphological, biochemical parameters and plant defense system were analyzed. Tomato plants exposed to Li had shorter roots and shoots, lower biomass and relative water contents, and showed decreases in physiological variables, as well as increases in electrolyte leakage and lipid peroxidation. However, the application of CBC and SS as passivators, either singly or in combination, increased growth variables of tomato and relieved Li-induced oxidative stress responses. The combined CBC and SS amendments reduced Li accumulation 82 and 90% in tomato roots and shoots, respectively, thereby minimizing the negative impacts of Li. Antioxidant enzymes SOD, CAT, APX and GR reflected 4, 5, 30, and 52% and glyoxalase enzymes I and II 7 and 250% enhancement in presence of both CBC and SS in Li treated soil, with a concurrent decrease in methylglyoxal content. Lithium treatment triggered oxidative stress, increased enzymatic and non-enzymatic antioxidant levels, and induced the synthesis of thiols and phytochelatins in roots and shoots. Hence, co-amendment with CBC and SS protected tomato plants from Li-induced oxidative damage by increasing antioxidant defenses and glyoxalase system activity. Both CBC, generated from agricultural waste, and SS, an industrial waste, are environmentally benign, safe, economical, and non-hazardous materials that can be easily applied on a large scale for crop production in Li-polluted soils. The present findings highlight the novel reutilization of waste materials as renewable assets to overcome soil Li problems and emphasize the conversion of waste into wealth and its potential for practical applications.


Asunto(s)
Antioxidantes , Carbón Orgánico , Litio , Piruvaldehído , Solanum lycopersicum , Solanum lycopersicum/efectos de los fármacos , Solanum lycopersicum/metabolismo , Piruvaldehído/metabolismo , Antioxidantes/metabolismo , Carbón Orgánico/farmacología , Litio/metabolismo , Litio/farmacología , Acero , Estrés Oxidativo/efectos de los fármacos
7.
Biomed Pharmacother ; 178: 117287, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39137652

RESUMEN

This study investigates the effects of inositol (INO) supplementation on cardiac changes caused by Li in mice. The study involved 4 groups of C57BL6 mice (n=10 each): (i) mice orally administered with Li2CO3 for 8 weeks, then 4 additional weeks without (Li_group) or (ii) with INO supplementation (Li_INOdelayed_group) (total of 12 weeks); (iii) mice given Li2CO3 and INO supplementation concurrently for 12 weeks (Li+INO_group); (iv) one group left untreated (C-group). The INO was administered as a mixture of myo-inositol and d-chiro-inositol (80:1) in drinking water. The mice were characterised for heart morphology, function, electrical activity, arrhythmogenic susceptibility, and multiorgan histopathology (heart, liver and kidney). Cardiomyocyte size, protein expression of key signalling pathways related to hypertrophy, and transcription levels of ion channel subunits and hypertrophy markers were evaluated in the ventricle tissue. The study found that INO supplementation reduced the Li-induced cardiac adverse effects, including systolic impairment and increased susceptibility to arrhythmias. The positive effect on arrhythmias might be attributed to the restored expression levels of the potassium channel subunit Kv 1.5. Additionally, INO improved cardiomyocyte hypertrophy, possibly by inhibiting the Li-induced activation of the ERK1/2 signalling pathway and by restoring the normal expression level of BNP, and alleviated injury in the liver and kidney. The effect was preventive if INO supplementation was taken concurrently with Li and therapeutic if INO was administered after Li-induced cardiac impairments were established. These results provide new insights into the cardioprotective effect of INO and suggest a potential treatment approach for Li-induced cardiac disease.


Asunto(s)
Suplementos Dietéticos , Inositol , Ratones Endogámicos C57BL , Animales , Masculino , Ratones , Administración Oral , Inositol/farmacología , Inositol/administración & dosificación , Litio/administración & dosificación , Litio/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Miocitos Cardíacos/metabolismo , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/prevención & control , Arritmias Cardíacas/tratamiento farmacológico , Cardiopatías/inducido químicamente , Cardiopatías/prevención & control , Cardiopatías/patología , Cardiopatías/tratamiento farmacológico
8.
Int J Mol Sci ; 25(14)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-39062811

RESUMEN

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.


Asunto(s)
Región CA1 Hipocampal , Modelos Animales de Enfermedad , Epilepsia , Litio , Pilocarpina , Células Piramidales , Animales , Células Piramidales/patología , Células Piramidales/metabolismo , Ratas , Epilepsia/inducido químicamente , Epilepsia/patología , Epilepsia/fisiopatología , Masculino , Región CA1 Hipocampal/patología , Litio/toxicidad , Litio/farmacología , Corteza Entorrinal/patología , Ratas Wistar
9.
Life Sci ; 352: 122917, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39019341

RESUMEN

BACKGROUND: Status epilepticus (SE) as a severe neurodegenerative disease, greatly negatively affects people's health, and there is an urgent need for innovative treatments. The valuable neuroprotective effects of glucagon-like peptide-1 (GLP-1) in several neurodegenerative diseases have raised motivation to investigate the dipeptidyl peptidase-4 (DPP-4) inhibitor; alogliptin (ALO), an oral antidiabetic drug as a potential treatment for SE. ALO has shown promising neuroprotective effects in Alzheimer's and Parkinson's diseases, but its impact on SE has not yet been studied. AIM: The present study aimed to explore the repurposing potential for ALO in a lithium/pilocarpine (Li/Pil)-induced SE model in rats. MAIN METHODS: ALO (30 mg/kg/day) was administered via gavage for 14 days, and SE was subsequently induced in the rats using a single dose of Li/Pil (127/60 mg/kg), while levetiracetam was used as a standard antiepileptic drug. KEY FINDINGS: The results showed that ALO reduced seizure severity and associated hippocampal neurodegeneration. ALO also increased γ-aminobutyric acid (GABA) levels, diminished glutamate spikes, and corrected glial fibrillary acidic protein (GFAP) changes. At the molecular level, ALO increased GLP-1 levels and activated its downstream signaling pathway, AMP-activated protein kinase (AMPK)/sirtuin-1 (SIRT1). ALO also dampened the brain's pro-oxidant response, curbed neuroinflammation, and counteracted hippocampal apoptosis affording neuroprotection. In addition, it activated autophagy as indicated by Beclin1 elevation. SIGNIFICANCE: This study suggested that the neuroprotective properties and autophagy-enhancing effects of ALO make it a promising treatment for SE and can potentially be used as a management approach for this condition.


Asunto(s)
Autofagia , Factor 2 Relacionado con NF-E2 , Fármacos Neuroprotectores , Pilocarpina , Piperidinas , Convulsiones , Sirtuina 1 , Animales , Ratas , Sirtuina 1/metabolismo , Fármacos Neuroprotectores/farmacología , Masculino , Autofagia/efectos de los fármacos , Piperidinas/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Convulsiones/inducido químicamente , Convulsiones/tratamiento farmacológico , Convulsiones/metabolismo , Litio/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Uracilo/análogos & derivados , Uracilo/farmacología , Ratas Sprague-Dawley , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Aging (Albany NY) ; 16(11): 9309-9333, 2024 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-38862239

RESUMEN

The amount of dietary sugars and the administration of lithium both impact the lifespan of the fruit fly Drosophila melanogaster. It is noteworthy that lithium is attributed with insulin-like activity as it stimulates protein kinase B/Akt and suppresses the activity of glycogen synthase kinase-3 (GSK-3). However, its interaction with dietary sugar has largely remained unexplored. Therefore, we investigated the effects of lithium supplementation on known lithium-sensitive parameters in fruit flies, such as lifespan, body composition, GSK-3 phosphorylation, and the transcriptome, while varying the dietary sugar concentration. For all these parameters, we observed that the efficacy of lithium was significantly influenced by the sucrose content in the diet. Overall, we found that lithium was most effective in enhancing longevity and altering body composition when added to a low-sucrose diet. Whole-body RNA sequencing revealed a remarkably similar transcriptional response when either increasing dietary sucrose from 1% to 10% or adding 1 mM LiCl to a 1% sucrose diet, characterized by a substantial overlap of nearly 500 differentially expressed genes. Hence, dietary sugar supply is suggested as a key factor in understanding lithium bioactivity, which could hold relevance for its therapeutic applications.


Asunto(s)
Sacarosa en la Dieta , Drosophila melanogaster , Longevidad , Animales , Drosophila melanogaster/genética , Drosophila melanogaster/efectos de los fármacos , Longevidad/efectos de los fármacos , Longevidad/genética , Regulación de la Expresión Génica/efectos de los fármacos , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Litio/farmacología , Cloruro de Litio/farmacología , Fosforilación/efectos de los fármacos , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
11.
Brain Behav ; 14(6): e3595, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38874089

RESUMEN

INTRODUCTION: Traumatic brain injury (TBI) refers to damage to brain tissue by mechanical or blunt force via trauma. TBI is often associated with impaired cognitive abilities, like difficulties in memory, learning, attention, and other higher brain functions, that typically remain for years after the injury. Lithium is an elementary light metal that is only utilized in salt form due to its high intrinsic reactivity. This current review discusses the molecular mechanisms and therapeutic and neuroprotective effects of lithium in TBI. METHOD: The "Boolean logic" was used to search for articles on the subject matter in PubMed and PubMed Central, as well as Google Scholar. RESULTS: Lithium's therapeutic action is extremely complex, involving multiple effects on gene secretion, neurotransmitter or receptor-mediated signaling, signal transduction processes, circadian modulation, as well as ion transport. Lithium is able to normalize multiple short- as well as long-term modifications in neuronal circuits that ultimately result in disparity in cortical excitation and inhibition activated by TBI. Also, lithium levels are more distinct in the hippocampus, thalamus, neo-cortex, olfactory bulb, amygdala as well as the gray matter of the cerebellum following treatment of TBI. CONCLUSION: Lithium attenuates neuroinflammation and neuronal toxicity as well as protects the brain from edema, hippocampal neurodegeneration, loss of hemispheric tissues, and enhanced memory as well as spatial learning after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Fármacos Neuroprotectores , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Humanos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/administración & dosificación , Animales , Litio/farmacología , Litio/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Compuestos de Litio/farmacología
12.
ACS Appl Mater Interfaces ; 16(24): 30793-30809, 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38833412

RESUMEN

Both bone mesenchymal stem cells (BMSCs) and their exosomes suggest promising therapeutic tools for bone regeneration. Lithium has been reported to regulate BMSC function and engineer exosomes to improve bone regeneration in patients with glucocorticoid-induced osteonecrosis of the femoral head. However, the mechanisms by which lithium promotes osteogenesis have not been elucidated. Here, we demonstrated that lithium promotes the osteogenesis of BMSCs via lithium-induced increases in the secretion of exosomal Wnt10a to activate Wnt/ß-catenin signaling, whose secretion is correlated with enhanced MARK2 activation to increase the trafficking of the Rab11a and Rab11FIP1 complexes together with exosomal Wnt10a to the plasma membrane. Then, we compared the proosteogenic effects of exosomes derived from lithium-treated or untreated BMSCs (Li-Exo or Con-Exo) both in vitro and in vivo. We found that, compared with Con-Exo, Li-Exo had superior abilities to promote the uptake and osteogenic differentiation of BMSCs. To optimize the in vivo application of these hydrogels, we fabricated Li-Exo-functionalized gelatin methacrylate (GelMA) hydrogels, which are more effective at promoting osteogenesis and bone repair than Con-Exo. Collectively, these findings demonstrate the mechanism by which lithium promotes osteogenesis and the great promise of lithium for engineering BMSCs and their exosomes for bone regeneration, warranting further exploration in clinical practice.


Asunto(s)
Exosomas , Litio , Células Madre Mesenquimatosas , Osteogénesis , beta Catenina , Proteínas de Unión al GTP rab , Osteogénesis/efectos de los fármacos , Exosomas/metabolismo , Exosomas/efectos de los fármacos , Exosomas/química , Animales , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Proteínas de Unión al GTP rab/metabolismo , beta Catenina/metabolismo , Litio/química , Litio/farmacología , Proteínas Wnt/metabolismo , Ratones , Diferenciación Celular/efectos de los fármacos , Ratas , Hidrogeles/química , Hidrogeles/farmacología , Ratas Sprague-Dawley , Vía de Señalización Wnt/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Humanos , Masculino
13.
Arch Pharm (Weinheim) ; 357(8): e2400063, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38704748

RESUMEN

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.


Asunto(s)
Amilorida , Acuaporina 2 , Diabetes Insípida Nefrogénica , Modelos Animales de Enfermedad , Túbulos Renales Colectores , Animales , Diabetes Insípida Nefrogénica/inducido químicamente , Diabetes Insípida Nefrogénica/prevención & control , Túbulos Renales Colectores/efectos de los fármacos , Túbulos Renales Colectores/patología , Túbulos Renales Colectores/metabolismo , Masculino , Ratas , Acuaporina 2/metabolismo , Amilorida/farmacología , Ratas Wistar , Factores de Tiempo , Insuficiencia Renal Crónica/prevención & control , Insuficiencia Renal Crónica/inducido químicamente , Litio/farmacología , Relación Dosis-Respuesta a Droga
14.
Gen Physiol Biophys ; 43(3): 263-271, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38774925

RESUMEN

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.


Asunto(s)
Ansiedad , Encéfalo , Relación Dosis-Respuesta a Droga , Estrés Oxidativo , Ratas Wistar , Animales , Estrés Oxidativo/efectos de los fármacos , Masculino , Ratas , Ansiedad/inducido químicamente , Ansiedad/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Litio/farmacología , Litio/administración & dosificación , Conducta Animal/efectos de los fármacos , Esquema de Medicación , Compuestos de Litio/farmacología , Compuestos de Litio/administración & dosificación
15.
J Cell Mol Med ; 28(10): e18385, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38801405

RESUMEN

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.


Asunto(s)
Autofagia , Necrosis de la Cabeza Femoral , Glucocorticoides , Litio , Osteoblastos , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Autofagia/efectos de los fármacos , Glucocorticoides/farmacología , Glucocorticoides/efectos adversos , Ratas , Necrosis de la Cabeza Femoral/inducido químicamente , Necrosis de la Cabeza Femoral/patología , Necrosis de la Cabeza Femoral/tratamiento farmacológico , Necrosis de la Cabeza Femoral/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/efectos de los fármacos , Litio/farmacología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Masculino , Osteogénesis/efectos de los fármacos , Ratas Sprague-Dawley , Proteínas Proto-Oncogénicas c-akt/metabolismo , Modelos Animales de Enfermedad , Fosfatidilinositol 3-Quinasas/metabolismo , Cabeza Femoral/patología , Cabeza Femoral/efectos de los fármacos , Cabeza Femoral/metabolismo , Osteonecrosis/inducido químicamente , Osteonecrosis/patología , Osteonecrosis/tratamiento farmacológico , Osteonecrosis/metabolismo , Osteonecrosis/prevención & control
16.
J Affect Disord ; 358: 416-421, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38735581

RESUMEN

BACKGROUND: The therapeutic response to lithium in patients with bipolar disorder is highly variable and has a polygenic basis. Genome-wide association studies investigating lithium response have identified several relevant loci, though the precise mechanisms driving these associations are poorly understood. We aimed to prioritise the most likely effector gene and determine the mechanisms underlying an intergenic lithium response locus on chromosome 21 identified by the International Consortium on Lithium Genetics (ConLi+Gen). METHODS: We conducted in-silico functional analyses by integrating and synthesising information from several publicly available functional genetic datasets and databases including the Genotype-Tissue Expression (GTEx) project and HaploReg. RESULTS: The findings from this study highlighted TMPRSS15 as the most likely effector gene at the ConLi+Gen lithium response locus. TMPRSS15 encodes enterokinase, a gastrointestinal enzyme responsible for converting trypsinogen into trypsin and thus aiding digestion. Convergent findings from gene-based lookups in human and mouse databases as well as co-expression network analyses of small intestinal RNA-seq data (GTEx) implicated TMPRSS15 in the regulation of intestinal nutrient absorption, including ions like sodium and potassium, which may extend to lithium. LIMITATIONS: Although the findings from this study indicated that TMPRSS15 was the most likely effector gene at the ConLi+Gen lithium response locus, the evidence was circumstantial. Thus, the conclusions from this study need to be validated in appropriately designed wet-lab studies. CONCLUSIONS: The findings from this study are consistent with a model whereby TMPRSS15 impacts the efficacy of lithium treatment in patients with bipolar disorder by modulating intestinal lithium absorption.


Asunto(s)
Trastorno Bipolar , Simulación por Computador , Absorción Intestinal , Serina Endopeptidasas , Trastorno Bipolar/tratamiento farmacológico , Trastorno Bipolar/genética , Trastorno Bipolar/metabolismo , Humanos , Absorción Intestinal/efectos de los fármacos , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Ratones , Animales , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Litio/uso terapéutico , Litio/farmacología , Antimaníacos/farmacología , Antimaníacos/uso terapéutico , Estudio de Asociación del Genoma Completo , Compuestos de Litio/farmacología , Compuestos de Litio/uso terapéutico , Compuestos de Litio/farmacocinética
17.
EBioMedicine ; 104: 105161, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38772282

RESUMEN

BACKGROUND: Bipolar disorder (BD) is a multifactorial psychiatric illness affecting ∼1% of the global adult population. Lithium (Li), is the most effective mood stabilizer for BD but works only for a subset of patients and its mechanism of action remains largely elusive. METHODS: In the present study, we used iPSC-derived neurons from patients with BD who are responsive (LR) or not (LNR) to lithium. Combined electrophysiology, calcium imaging, biochemistry, transcriptomics, and phosphoproteomics were employed to provide mechanistic insights into neuronal hyperactivity in BD, investigate Li's mode of action, and identify alternative treatment strategies. FINDINGS: We show a selective rescue of the neuronal hyperactivity phenotype by Li in LR neurons, correlated with changes to Na+ conductance. Whole transcriptome sequencing in BD neurons revealed altered gene expression pathways related to glutamate transmission, alterations in cell signalling and ion transport/channel activity. We found altered Akt signalling as a potential therapeutic effect of Li in LR neurons from patients with BD, and that Akt activation mimics Li effect in LR neurons. Furthermore, the increased neural network activity observed in both LR & LNR neurons from patients with BD were reversed by AMP-activated protein kinase (AMPK) activation. INTERPRETATION: These results suggest potential for new treatment strategies in BD, such as Akt activators in LR cases, and the use of AMPK activators for LNR patients with BD. FUNDING: Supported by funding from ERA PerMed, Bell Brain Canada Mental Research Program and Brain & Behavior Research Foundation.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Trastorno Bipolar , Células Madre Pluripotentes Inducidas , Neuronas , Proteínas Proto-Oncogénicas c-akt , Trastorno Bipolar/metabolismo , Trastorno Bipolar/tratamiento farmacológico , Humanos , Neuronas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Litio/farmacología , Litio/uso terapéutico , Transducción de Señal , Perfilación de la Expresión Génica , Transcriptoma
18.
Naunyn Schmiedebergs Arch Pharmacol ; 397(9): 6989-6999, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38625552

RESUMEN

Epilepsy is a condition marked by sudden, self-sustained, and recurring brain events, showcasing unique electro-clinical and neuropathological phenomena that can alter the structure and functioning of the brain, resulting in diverse manifestations. Antiepileptic drugs (AEDs) can be very effective in 30% of patients in controlling seizures. Several factors contribute to this: drug resistance, individual variability, side effects, complexity of epilepsy, incomplete understanding, comorbidities, drug interactions, and no adherence to treatment. Therefore, research into new AEDs is important for several reasons such as improved efficacy, reduced side effects, expanded treatment options, treatment for drug-resistant epilepsy, improved safety profiles, targeted therapies, and innovation and progress. Animal models serve as crucial biological tools for comprehending neuronal damage and aiding in the discovery of more effective new AEDs. The utilization of antioxidant agents that act on the central nervous system may serve as a supplementary approach in the secondary prevention of epilepsy, both in laboratory animals and potentially in humans. Chlorogenic acid (CGA) is a significant compound, widely prevalent in numerous medicinal and food plants, exhibiting an extensive spectrum of biological activities such as neuroprotection, antioxidant, anti-inflammatory, and analgesic effects, among others. In this research, we assessed the neuroprotective effects of commercially available CGA in Wistar rats submitted to lithium-pilocarpine-induced status epilepticus (SE) model. After 72-h induction of SE, rats received thiopental and were treated for three consecutive days (1st, 2nd, and 3rd doses). Next, brains were collected and studied histologically for viable cells in the hippocampus with staining for cresyl-violet (Nissl staining) and for degenerating cells with Fluoro-Jade C (FJC) staining. Moreover, to evaluate oxidative stress, the presence of malondialdehyde (MDA) and superoxide dismutase (SOD) was quantified. Rats administered with CGA (30 mg/kg) demonstrated a significant decrease of 59% in the number of hippocampal cell loss in the CA3, and of 48% in the hilus layers after SE. A significant reduction of 75% in the cell loss in the CA3, shown by FJC+ staining, was also observed with the administration of CGA (30 mg/kg). Furthermore, significant decreases of 49% in MDA production and 72% in the activity of SOD were seen, when compared to animals subjected to SE that received vehicle. This study introduces a novel finding: the administration of CGA at a dosage of 30 mg/kg effectively reduced oxidative stress induced by lithium-pilocarpine, with its effects lasting until the peak of neural damage 72 h following the onset of SE. Overall, the research and development of new AEDs are essential for advancing epilepsy treatment, improving patient outcomes, and ultimately enhancing the quality of life for individuals living with epilepsy.


Asunto(s)
Ácido Clorogénico , Fármacos Neuroprotectores , Estrés Oxidativo , Pilocarpina , Ratas Wistar , Estado Epiléptico , Animales , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Pilocarpina/toxicidad , Estrés Oxidativo/efectos de los fármacos , Ácido Clorogénico/farmacología , Ácido Clorogénico/uso terapéutico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Masculino , Ratas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Modelos Animales de Enfermedad , Anticonvulsivantes/farmacología , Anticonvulsivantes/uso terapéutico , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Litio/farmacología
19.
Exp Gerontol ; 191: 112442, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38663491

RESUMEN

In this study we investigated the potential synergistic effects of moderate interval training (MIT) and lithium on spatial learning and memory. Forty-two male Wistar males were classified into six groups including I: Control, II: 10 mg/kg/day IP lithium (Li10), III: MIT, IV: Li10 + MIT, V: 40 mg/kg/day IP lithium (Li40), and VI: Li40 + MIT. Then, the rats underwent Morris Water Maze (MWM) test to assess their spatial memory and learning ability. Brain-derived neurotrophic factor (BDNF) density was measured by enzyme-linked immunosorbent assay (ELISA), and the expression of PGC1 and SIRT3 were assessed via qRT-PCR. The results show that MIT improves both memory and spatial learning; but lithium alone, does not cause this. Additionally, those exposed to a combination of exercise and lithium also had improved spatial learning and memory. Finally, we observed a positive role of BDNF protein, and PGC1 gene on the effects of exercise and lithium.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Condicionamiento Físico Animal , Sirtuina 3 , Memoria Espacial , Animales , Masculino , Ratas , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Litio/farmacología , Aprendizaje por Laberinto/efectos de los fármacos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Condicionamiento Físico Animal/fisiología , Ratas Wistar , Sirtuina 3/metabolismo , Sirtuina 3/genética , Sirtuinas , Aprendizaje Espacial/efectos de los fármacos , Memoria Espacial/efectos de los fármacos
20.
PLoS One ; 19(4): e0299534, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38574297

RESUMEN

Alzheimer's disease (AD) is the most common neuronal disorder that leads to the development of dementia. Until nowadays, some therapies may alleviate the symptoms, but there is no pharmacological treatment. Microdosing lithium has been used to modify the pathological characteristics of the disease, with effects in both experimental and clinical conditions. The present work aimed to analyze the effects of this treatment on spatial memory, anxiety, and molecular mechanisms related to long-term memory formation during the aging process of a mouse model of accelerated aging (SAMP-8). Female SAMP-8 showed learning and memory impairments together with disruption of memory mechanisms, neuronal loss, and increased density of senile plaques compared to their natural control strain, the senescence-accelerated mouse resistant (SAMR-1). Chronic treatment with lithium promoted memory maintenance, reduction in anxiety, and maintenance of proteins related to memory formation and neuronal density. The density of senile plaques was also reduced. An increase in the density of gamma-aminobutyric acid A (GABAA) and α7 nicotinic cholinergic receptors was also observed and related to neuroprotection and anxiety reduction. In addition, this microdose of lithium inhibited the activation of glycogen synthase kinase-3beta (GSK-3ß), the classical mechanism of lithium cell effects, which could contribute to the preservation of the memory mechanism and reduction in senile plaque formation. This work shows that lithium effects in neuroprotection along the aging process are not related to a unique cellular mechanism but produce multiple effects that slowly protect the brain along the aging process.


Asunto(s)
Enfermedad de Alzheimer , Litio , Compuestos de Fenilmercurio , Ratones , Femenino , Animales , Litio/farmacología , Litio/uso terapéutico , Placa Amiloide/patología , Glucógeno Sintasa Quinasa 3 beta , Enfermedad de Alzheimer/patología , Envejecimiento/metabolismo , Modelos Animales de Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA