Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.139
Filtrer
1.
Front Endocrinol (Lausanne) ; 15: 1339191, 2024.
Article de Anglais | MEDLINE | ID: mdl-38974575

RÉSUMÉ

Background and purpose: Thyroid papillary carcinoma (PTC) had a high possibility of recurrence after surgery, and thyroid stimulating hormone (TSH) suppression and radioactive iodine (131I) were used for postoperative therapy. This study explored the potential mechanism of lymph node metastasis (LNM) and aimed to develop differentiated treatments for PTC. Method: This study explored the risk factors of lymph node metastasis in PTC by analyzing the clinical information of 2073 cases. The Cancer Genome Atlas Thyroid Cancer (TCGA-THCA) and the Gene Expression Omnibus (GEO) databases of gene expression were analyzed to identify the interrelationships between gene expression to phenotype. Results: Analyzing clinical data, we found that male gender, younger age, larger tumor size, and extra-thyroidal extension (ETE) were risk significant risk factors for lymph node metastasis(P<0.05). Conversely, thyroid function parameters such as TSH, FT3, FT4, TSH/FT3, and TSH/FT4 didn't correlate with LNM(P>0.05), and TSH levels were observed to be higher in females(P<0.05). Gene expression analysis revealed that SLC5A5 was down-regulated in males, younger individuals, and those with lymph node metastasis, and a lower level of SLC5A5 was associated with a worse disease-free survival(P<0.05). Additionally, our examination of single-cell RNA sequencing (scRNA-seq) data indicated that SLC5A5 expression was reduced in tumors and lymph node metastasis samples, correlating positively with the expression of TSHR. Conclusion: The impact of TSH on PTC behavior remained unclear, while the capacity for absorbing 131I in dependence on SLC5A5 showed variations across different genders and ages. We conclude that postoperative treatment of PTC should take into account the differences caused by gender and age.


Sujet(s)
Métastase lymphatique , Cancer papillaire de la thyroïde , Tumeurs de la thyroïde , Humains , Mâle , Femelle , Cancer papillaire de la thyroïde/anatomopathologie , Cancer papillaire de la thyroïde/génétique , Cancer papillaire de la thyroïde/chirurgie , Cancer papillaire de la thyroïde/thérapie , Tumeurs de la thyroïde/anatomopathologie , Tumeurs de la thyroïde/chirurgie , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/thérapie , Tumeurs de la thyroïde/métabolisme , Adulte d'âge moyen , Adulte , Radio-isotopes de l'iode/usage thérapeutique , Facteurs sexuels , Facteurs âges , Symporteurs/génétique , Symporteurs/métabolisme , Thyroïdectomie , Facteurs de risque , Thyréostimuline/sang , Récidive tumorale locale/génétique , Récidive tumorale locale/anatomopathologie , Sujet âgé , Pronostic
2.
J Control Release ; 372: 885-900, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38971425

RÉSUMÉ

Statins are widely used to treat hyperlipidemia; however, their mechanism-inhibiting cholesterol production without promoting its utilization-causes problems, such as inducing diabetes. In our research, we develop, for the first time, a chemically engineered statin conjugate that not only inhibits cholesterol production but also enhances its consumption through its multifunctional properties. The novel rosuvastatin (RO) and ursodeoxycholic acid (UDCA) conjugate (ROUA) is designed to bind to and inhibit the core of the apical sodium-dependent bile acid transporter (ASBT), effectively blocking ASBT's function in the small intestine, maintaining the effect of rosuvastatin. Consequently, ROUA not only preserves the cholesterol-lowering function of statins but also prevents the reabsorption of bile acids, thereby increasing cholesterol consumption. Additionally, ROUA's ability to self-assemble into nanoparticles in saline-attributable to its multiple hydroxyl groups and hydrophobic nature-suggests its potential for a prolonged presence in the body. The oral administration of ROUA nanoparticles in animal models using a high-fat or high-fat/high-fructose diet shows remarkable therapeutic efficacy in fatty liver, with low systemic toxicity. This innovative self-assembling multifunctional molecule design approach, which boosts a variety of therapeutic effects while minimizing toxicity, offers a significant contribution to the advancement of drug development.


Sujet(s)
Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Nanoparticules , Transporteurs d'anions organiques sodium-dépendants , Rosuvastatine de calcium , Symporteurs , Animaux , Nanoparticules/composition chimique , Transporteurs d'anions organiques sodium-dépendants/antagonistes et inhibiteurs , Transporteurs d'anions organiques sodium-dépendants/métabolisme , Symporteurs/antagonistes et inhibiteurs , Symporteurs/métabolisme , Mâle , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/administration et posologie , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/composition chimique , Rosuvastatine de calcium/administration et posologie , Humains , Souris de lignée C57BL , Acides et sels biliaires/métabolisme , Acides et sels biliaires/composition chimique , Cholestérol/composition chimique , Rat Sprague-Dawley , Souris
3.
Genes (Basel) ; 15(7)2024 Jul 14.
Article de Anglais | MEDLINE | ID: mdl-39062697

RÉSUMÉ

BACKGROUND: Polymorphism rs1049434 characterizes the nonsynonymous exchange of adenosine (A) by thymidine (T) in the gene for monocarboxylate transporter 1 (MCT1). We tested whether T-allele carriers of rs1049434 demonstrate increased accumulation of markers of metabolic strain. METHODS: Physically active, healthy, young male subjects (n = 22) conducted a power-matched one-legged cycling exercise to exhaustion. Metabolic substrates in capillary blood, selected metabolic compounds, and indices for the slow oxidative phenotype of vastus lateralis muscle were quantified in samples collected before and after exercise. The genotypes of the rs1049434 polymorphism were determined with polymerase chain reactions. RESULTS: One-legged exercise affected the concentration of muscle metabolites entering the tricarboxylic acid cycle, such as acetyl-co-enzyme A (+448%) and acetyl-L-carnitine (+548%), muscle glycogen (-59%), and adenosine monophosphate (-39%), 30 min post-exercise. Exercise-related variability in the muscular concentration of glycogen, long-chain acyl co-enzyme As and a triglyceride, nicotinamide adenine dinucleotide (NADH), and adenosine monophosphate (AMP) interacted with rs1049434. T-allele carriers demonstrated a 39% lesser reduction in glycogen after exercise than non-carriers when NADH increased only in the non-carriers. Muscle lactate concentration was 150% higher, blood triacyl-glyceride concentration was 53% lower, and slow fiber percentage was 20% lower in T-allele carriers. DISCUSSION: The observations suggest a higher anaerobic glycolytic strain during exhaustive exercise and a lowered lipid handling in T-allele non-carriers.


Sujet(s)
Allèles , Exercice physique , Transporteurs d'acides monocarboxyliques , Polymorphisme de nucléotide simple , Symporteurs , Humains , Mâle , Transporteurs d'acides monocarboxyliques/génétique , Transporteurs d'acides monocarboxyliques/métabolisme , Adulte , Symporteurs/génétique , Symporteurs/métabolisme , Jeune adulte , Muscles squelettiques/métabolisme , Glycogène/métabolisme , Génotype
4.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39062808

RÉSUMÉ

The melanocortin-4 receptor (MC4R) is a key player in the hypothalamic leptin-melanocortin pathway that regulates satiety and hunger. MC4R belongs to the G protein-coupled receptors (GPCRs), which are known to form heterodimers with other membrane proteins, potentially modulating receptor function or characteristics. Like MC4R, thyroid hormones (TH) are also essential for energy homeostasis control. TH transport across membranes is facilitated by the monocarboxylate transporter 8 (MCT8), which is also known to form heterodimers with GPCRs. Based on the finding in single-cell RNA-sequencing data that both proteins are simultaneously expressed in hypothalamic neurons, we investigated a putative interplay between MC4R and MCT8. We developed a novel staining protocol utilizing a fluorophore-labeled MC4R ligand and demonstrated a co-localization of MC4R and MCT8 in human brain tissue. Using in vitro assays such as BRET, IP1, and cAMP determination, we found that MCT8 modulates MC4R-mediated phospholipase C activation but not cAMP formation via a direct interaction, an effect that does not require a functional MCT8 as it was not altered by a specific MCT8 inhibitor. This suggests an extended functional spectrum of MCT8 as a GPCR signaling modulator and argues for the investigation of further GPCR-protein interactions with hitherto underrepresented physiological functions.


Sujet(s)
Transporteurs d'acides monocarboxyliques , Récepteur de la mélanocortine de type 4 , Type C Phospholipases , Humains , Récepteur de la mélanocortine de type 4/métabolisme , Récepteur de la mélanocortine de type 4/génétique , Transporteurs d'acides monocarboxyliques/métabolisme , Transporteurs d'acides monocarboxyliques/génétique , Type C Phospholipases/métabolisme , Cellules HEK293 , Transduction du signal , AMP cyclique/métabolisme , Symporteurs/métabolisme , Symporteurs/génétique , Liaison aux protéines , Animaux
5.
Proc Natl Acad Sci U S A ; 121(30): e2402560121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39018199

RÉSUMÉ

The key role of a thyroid hormone receptor in determining the maturation and diversity of cone photoreceptors reflects a profound influence of endocrine signaling on the cells that mediate color vision. However, the route by which hormone reaches cones remains enigmatic as cones reside in the retinal photoreceptor layer, shielded by the blood-retina barrier. Using genetic approaches, we report that cone differentiation is regulated by a membrane transporter for thyroid hormone, MCT8 (SLC16A2), in the retinal pigment epithelium (RPE), which forms the outer blood-retina barrier. Mct8-deficient mice display hypothyroid-like cone gene expression and compromised electroretinogram responses. Mammalian color vision is typically facilitated by cone types that detect medium-long (M) and short (S) wavelengths of light but Mct8-deficient mice have a partial shift of M to S cone identity, resembling the phenotype of thyroid hormone receptor deficiency. RPE-specific ablation of Mct8 results in similar shifts in cone identity and hypothyroid-like gene expression whereas reexpression of MCT8 in the RPE in Mct8-deficient mice partly restores M cone identity, consistent with paracrine-like control of thyroid hormone signaling by the RPE. Our findings suggest that in addition to transport of essential solutes and homeostatic support for photoreceptors, the RPE regulates the thyroid hormone signal that promotes cone-mediated vision.


Sujet(s)
Différenciation cellulaire , Souris knockout , Transporteurs d'acides monocarboxyliques , Cellules photoréceptrices en cône de la rétine , Épithélium pigmentaire de la rétine , Symporteurs , Animaux , Cellules photoréceptrices en cône de la rétine/métabolisme , Transporteurs d'acides monocarboxyliques/métabolisme , Transporteurs d'acides monocarboxyliques/génétique , Symporteurs/métabolisme , Symporteurs/génétique , Épithélium pigmentaire de la rétine/métabolisme , Souris , Hormones thyroïdiennes/métabolisme , Électrorétinographie
7.
Acta Med Okayama ; 78(3): 227-235, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38902210

RÉSUMÉ

Zolpidem, a non-benzodiazepine hypnotic, is primarily used to treat insomnia. In a previous study, pior treatment with non-benzodiazepine receptor agonists was associated with inflammation. The present study aimed to clarify the association between the effects of zolpidem and inflammation in mice treated with lipopolysaccharide (LPS), a known model of inflammation. We assessed the zolpidem-induced loss of righting reflex (LORR) duration 24 h after LPS treatment in mice. Additionally, the expressions of γ-aminobutyric acid (GABA)A receptor subunit and K+-Cl- cotransporter isoform 2 (KCC2) mRNA in the hippocampus and frontal cortex were examined in LPS-treated mice. Pretreatment with LPS was associated with significantly prolonged duration of zolpidem-induced LORR compared to control mice. This effect was significantly attenuated by administering bicuculline, a GABAA receptor antagonist, or flumazenil, a benzodiazepine receptor antagonist, in LPS-treated mice. Compared to controls, LPS-treated mice showed no significant change in the expression of GABAA receptor subunits in the hippocampus or frontal cortex. Bumetanide, an Na+-K+-2Cl- cotransporter isoform 1 blocker, attenuated the extended duration of zolpidem-induced LORR observed in LPS-treated mice. LPS significantly decreased Kcc2 mRNA expression in the hippocampus and the frontal cortex. These findings suggest that inflammation increases zolpidem-induced LORR, possibly through a reduction in KCC2 expression.


Sujet(s)
Lipopolysaccharides , Pyridines , Récepteurs GABA-A , Réflexe de redressement , Symporteurs , Zolpidem , Animaux , Zolpidem/pharmacologie , Souris , Pyridines/pharmacologie , Mâle , Récepteurs GABA-A/métabolisme , Récepteurs GABA-A/effets des médicaments et des substances chimiques , Symporteurs/génétique , Symporteurs/métabolisme , Réflexe de redressement/effets des médicaments et des substances chimiques , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , , Hypnotiques et sédatifs/pharmacologie , Inflammation/induit chimiquement , Lobe frontal/effets des médicaments et des substances chimiques , Lobe frontal/métabolisme
8.
PLoS One ; 19(6): e0304512, 2024.
Article de Anglais | MEDLINE | ID: mdl-38829838

RÉSUMÉ

The Organic Cation Transporter Novel 1 (OCTN1), also known as SLC22A4, is widely expressed in various human tissues, and involved in numerous physiological and pathological processes remains. It facilitates the transport of organic cations, zwitterions, with selectivity for positively charged solutes. Ergothioneine, an antioxidant compound, and acetylcholine (Ach) are among its substrates. Given the lack of experimentally solved structures of this protein, this study aimed at generating a reliable 3D model of OCTN1 to shed light on its substrate-binding preferences and the role of sodium in substrate recognition and transport. A chimeric model was built by grafting the large extracellular loop 1 (EL1) from an AlphaFold-generated model onto a homology model. Molecular dynamics simulations revealed domain-specific mobility, with EL1 exhibiting the highest impact on overall stability. Molecular docking simulations identified cytarabine and verapamil as highest affinity ligands, consistent with their known inhibitory effects on OCTN1. Furthermore, MM/GBSA analysis allowed the categorization of substrates into weak, good, and strong binders, with molecular weight strongly correlating with binding affinity to the recognition site. Key recognition residues, including Tyr211, Glu381, and Arg469, were identified through interaction analysis. Ach demonstrated a low interaction energy, supporting the hypothesis of its one-directional transport towards to outside of the membrane. Regarding the role of sodium, our model suggested the involvement of Glu381 in sodium binding. Molecular dynamics simulations of systems at increasing levels of Na+ concentrations revealed increased sodium occupancy around Glu381, supporting experimental data associating Na+ concentration to molecule transport. In conclusion, this study provides valuable insights into the 3D structure of OCTN1, its substrate-binding preferences, and the role of sodium in the recognition. These findings contribute to the understanding of OCTN1 involvement in various physiological and pathological processes and may have implications for drug development and disease management.


Sujet(s)
Simulation de docking moléculaire , Simulation de dynamique moléculaire , Transporteurs de cations organiques , Humains , Transporteurs de cations organiques/composition chimique , Transporteurs de cations organiques/métabolisme , Transporteurs de cations organiques/génétique , Symporteurs/composition chimique , Symporteurs/métabolisme , Sites de fixation , Liaison aux protéines , Ergothionéine/composition chimique , Ergothionéine/métabolisme , Sodium/métabolisme , Sodium/composition chimique , Simulation numérique , Acétylcholine/métabolisme , Acétylcholine/composition chimique , Ligands
9.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38928280

RÉSUMÉ

The present study examined how P2X7 receptor knockout (KO) modulates central post-stroke pain (CPSP) induced by lesions of the ventrobasal complex (VBC) of the thalamus in behaviors, molecular levels, and electrical recording tests. Following the experimental procedure, the wild-type and P2X7 receptor KO mice were injected with 10 mU/0.2 µL type IV collagenase in the VBC of the thalamus to induce an animal model of stroke-like thalamic hemorrhage. Behavioral data showed that the CPSP group induced thermal and mechanical pain. The P2X7 receptor KO group showed reduced thermal and mechanical pain responses compared to the CPSP group. Molecular assessments revealed that the CPSP group had lower expression of NeuN and KCC2 and higher expression of GFAP, IBA1, and BDNF. The P2X7 KO group showed lower expression of GFAP, IBA1, and BDNF but nonsignificant differences in KCC2 expression than the CPSP group. The expression of NKCC1, GABAa receptor, and TrkB did not differ significantly between the control, CPSP, and P2X7 receptor KO groups. Muscimol, a GABAa agonist, application increased multiunit numbers for monitoring many neurons and [Cl-] outflux in the cytosol in the CPSP group, while P2X7 receptor KO reduced multiunit activity and increased [Cl-] influx compared to the CPSP group. P2X4 receptor expression was significantly decreased in the 100 kDa but not the 50 kDa site in the P2X7 receptor KO group. Altogether, the P2X7 hypothesis of CPSP was proposed, wherein P2X7 receptor KO altered the CPSP pain responses, numbers of astrocytes and microglia, CSD amplitude of the anterior cingulate cortex and the medial dorsal thalamus, BDNF expression, [Cl-] influx, and P2X4 expression in 100 kDa with P2X7 receptors. The present findings have implications for the clinical treatment of CPSP symptoms.


Sujet(s)
, Souris knockout , Récepteurs purinergiques P2X7 , Accident vasculaire cérébral , Animaux , Récepteurs purinergiques P2X7/métabolisme , Récepteurs purinergiques P2X7/génétique , Souris , Accident vasculaire cérébral/métabolisme , Accident vasculaire cérébral/complications , Mâle , Douleur/métabolisme , Douleur/étiologie , Modèles animaux de maladie humaine , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/génétique , Symporteurs/métabolisme , Symporteurs/génétique , Souris de lignée C57BL , Neurones/métabolisme , Muscimol/pharmacologie , Protéine gliofibrillaire acide/métabolisme , Thalamus/métabolisme
10.
BMC Anesthesiol ; 24(1): 200, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38840092

RÉSUMÉ

BACKGROUND: The inhalational anesthetic isoflurane is commonly utilized in clinical practice, particularly in the field of pediatric anesthesia. Research has demonstrated its capacity to induce neuroinflammation and long-term behavioral disorders; however, the underlying mechanism remains unclear [1]. The cation-chloride cotransporters Na+-K+-2Cl--1 (NKCC1) and K+-2Cl--2 (KCC2) play a pivotal role in regulating neuronal responses to gamma-aminobutyric acid (GABA) [2]. Imbalances in NKCC1/KCC2 can disrupt GABA neurotransmission, potentially leading to neural circuit hyperexcitability and reduced inhibition following neonatal exposure to anesthesia [3]. Therefore, this study postulates that anesthetics have the potential to dysregulate NKCC1 and/or KCC2 during brain development. METHODS: We administered 1.5% isoflurane anesthesia to neonatal rats for a duration of 4 h at postnatal day 7 (PND7). Anxiety levels were assessed using the open field test at PND28, while cognitive function was evaluated using the Morris water maze test between PND31 and PND34. Protein levels of NKCC1, KCC2, BDNF, and phosphorylated ERK (P-ERK) in the hippocampus were measured through Western blotting analysis. Pro-inflammatory cytokines IL-1ß, IL-6, and TNF-α were quantified using ELISA. RESULTS: We observed a decrease in locomotion trajectories within the central region and a significantly shorter total distance in the ISO group compared to CON pups, indicating that isoflurane induces anxiety-like behavior. In the Morris water maze (MWM) test, rats exposed to isoflurane exhibited prolonged escape latency onto the platform. Additionally, isoflurane administration resulted in reduced time spent crossing in the MWM experiment at PND34, suggesting long-term impairment of memory function. Furthermore, we found that isoflurane triggered activation of pro-inflammatory cytokines IL-1ß, IL-6, and TNF-α; downregulated KCC2/BDNF/P-ERK expression; and increased the NKCC1/KCC2 ratio in the hippocampus of PND7 rats. Bumetadine (NKCC1 specific inhibitors) reversed cognitive damage and effective disorder induced by isoflurane in neonatal rats by inhibiting TNF-α activation, normalizing IL-6 and IL-1ß levels, restoring KCC2 expression levels as well as BDNF and ERK signaling pathways. Based on these findings, it can be speculated that BDNF, P-ERK, IL-1ß, IL-6 and TNF - α may act downstream of the NKCC1/KCC2 pathway. CONCLUSIONS: Our findings provide evidence that isoflurane administration in neonatal rats leads to persistent cognitive deficits through dysregulation of the Cation-Chloride Cotransporters NKCC1 and KCC2, BDNF, p-ERK proteins, as well as neuroinflammatory processes.


Sujet(s)
Anesthésiques par inhalation , Animaux nouveau-nés , Isoflurane , , Membre-2 de la famille-12 des transporteurs de solutés , Symporteurs , Animaux , Isoflurane/pharmacologie , Membre-2 de la famille-12 des transporteurs de solutés/métabolisme , Symporteurs/métabolisme , Anesthésiques par inhalation/pharmacologie , Anesthésiques par inhalation/effets indésirables , Rats , Souris , Rat Sprague-Dawley , Mâle , Maladies neuro-inflammatoires/induit chimiquement , Maladies neuro-inflammatoires/métabolisme , Femelle , Dysfonctionnement cognitif/induit chimiquement , Dysfonctionnement cognitif/métabolisme
11.
Biochem Biophys Res Commun ; 727: 150312, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38924962

RÉSUMÉ

A hallmark of Alzheimer's disease (AD) is amyloid-ß (Aß) plaque deposition in the brain, causing deficits in cognitive function. Amyloid-beta oligomers (AßOs), the soluble precursor peptides producing Aß plaques, also produce neurotoxicity and microgliosis together with glycolytic reprogramming. Recently, monocarboxylate transporter 1 (MCT1), a key glycolysis regulator, and its ancillary protein, CD147, are found to play an important role in the secretion of exosomes, 30-200 nm vesicles in size, which are considered as toxic molecule carriers in AD. However, the effect of low-concentration AßOs (1 nM) on microglia MCT1 and CD147 expression as well as 1 nM AßOs-treated microglia-derived exosomes on neuronal toxicity remain largely elusive. In this study, 1 nM AßOs induce significant axonopathy and microgliosis. Furthermore, 1 nM AßOs-treated neurons- or microglia-derived exosomes produce axonopathy through their autologous or heterologous uptake by neurons, supporting the role of exosomes as neurotoxicity mediators in AD. Interestingly, MCT1 and CD147 are enhanced in microglia by treatment with 1 nM AßOs or exosomes from 1 nM AßOs-treated- microglia or neurons, suggesting the implication of AßOs-induced enhanced MCT1 and CD147 in microglia with AD neuropathogenesis, which is consistent with the in-silico analysis of the single cell RNA sequencing data from microglia in mouse models of AD and AD patients.


Sujet(s)
Peptides bêta-amyloïdes , Exosomes , Microglie , Neurones , Exosomes/métabolisme , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/toxicité , Microglie/métabolisme , Microglie/anatomopathologie , Microglie/effets des médicaments et des substances chimiques , Animaux , Neurones/métabolisme , Neurones/anatomopathologie , Neurones/effets des médicaments et des substances chimiques , Souris , Antigènes CD147/métabolisme , Antigènes CD147/génétique , Transporteurs d'acides monocarboxyliques/métabolisme , Transporteurs d'acides monocarboxyliques/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Cellules cultivées , Symporteurs/métabolisme , Symporteurs/génétique , Souris de lignée C57BL , Humains
12.
Hear Res ; 450: 109048, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-38852535

RÉSUMÉ

The Blood-Labyrinth Barrier (BLB) is pivotal for the maintenance of lymphatic homeostasis within the inner ear, yet the intricacies of its development and function are inadequately understood. The present investigation delves into the contribution of the Mfsd2a molecule, integral to the structural and functional integrity of the Blood-Brain Barrier (BBB), to the ontogeny and sustenance of the BLB. Our empirical findings delineate that the maturation of the BLB in murine models is not realized until approximately two weeks post-birth, with preceding stages characterized by notable permeability. Transcriptomic analysis elucidates a marked augmentation in Mfsd2a expression within the lateral wall of the cochlea in specimens exhibiting an intact BLB. Moreover, both in vitro and in vivo assays substantiate that a diminution in Mfsd2a expression detrimentally impacts BLB permeability and structural integrity, principally via the attenuation of tight junction protein expression and the enhancement of endothelial cell transcytosis. These insights underscore the indispensable role of Mfsd2a in ensuring BLB integrity and propose it as a viable target for therapeutic interventions aimed at the amelioration of hearing loss.


Sujet(s)
Barrière hémato-encéphalique , Oreille interne , Symporteurs , Jonctions serrées , Transcytose , Animaux , Jonctions serrées/métabolisme , Barrière hémato-encéphalique/métabolisme , Oreille interne/métabolisme , Symporteurs/métabolisme , Symporteurs/génétique , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Cochlée/métabolisme , Souris de lignée C57BL , Perméabilité capillaire , Protéine-2 à domaine MARVEL/métabolisme , Protéine-2 à domaine MARVEL/génétique , Souris knockout , Protéines de la jonction serrée/métabolisme , Protéines de la jonction serrée/génétique , Régulation de l'expression des gènes au cours du développement , Souris , Perméabilité
13.
Int J Mol Sci ; 25(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892438

RÉSUMÉ

The strength of inhibitory neurotransmission depends on intracellular neuronal chloride concentration, primarily regulated by the activity of cation-chloride cotransporters NKCC1 (Sodium-Potassium-Chloride Cotransporter 1) and KCC2 (Potassium-Chloride Cotransporter 2). Brain-derived neurotrophic factor (BDNF) influences the functioning of these co-transporters. BDNF is synthesized from precursor proteins (proBDNF), which undergo proteolytic cleavage to yield mature BDNF (mBDNF). While previous studies have indicated the involvement of BDNF signaling in the activity of KCC2, its specific mechanisms are unclear. We investigated the interplay between both forms of BDNF and chloride homeostasis in rat hippocampal neurons and in utero electroporated cortices of rat pups, spanning the behavioral, cellular, and molecular levels. We found that both pro- and mBDNF play a comparable role in immature neurons by inhibiting the capacity of neurons to extrude chloride. Additionally, proBDNF increases the endocytosis of KCC2 while maintaining a depolarizing shift of EGABA in maturing neurons. Behaviorally, proBDNF-electroporated rat pups in the somatosensory cortex exhibit sensory deficits, delayed huddling, and cliff avoidance. These findings emphasize the role of BDNF signaling in regulating chloride transport through the modulation of KCC2. In summary, this study provides valuable insights into the intricate interplay between BDNF, chloride homeostasis, and inhibitory synaptic transmission, shedding light on the underlying cellular mechanisms involved.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Chlorures , , Neurones , Membre-2 de la famille-12 des transporteurs de solutés , Animaux , Femelle , Rats , Facteur neurotrophique dérivé du cerveau/métabolisme , Cellules cultivées , Chlorures/métabolisme , Hippocampe/métabolisme , Homéostasie , Neurones/métabolisme , Précurseurs de protéines/métabolisme , Symporteurs/métabolisme
14.
eNeuro ; 11(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38914464

RÉSUMÉ

Epilepsy is often comorbid with psychiatric illnesses, including anxiety and depression. Despite the high incidence of psychiatric comorbidities in people with epilepsy, few studies address the underlying mechanisms. Stress can trigger epilepsy and depression. Evidence from human and animal studies supports that hypothalamic-pituitary-adrenal (HPA) axis dysfunction may contribute to both disorders and their comorbidity ( Kanner, 2003). Here, we investigate if HPA axis dysfunction may influence epilepsy outcomes and psychiatric comorbidities. We generated a novel mouse model (Kcc2/Crh KO mice) lacking the K+/Cl- cotransporter, KCC2, in corticotropin-releasing hormone (CRH) neurons, which exhibit stress- and seizure-induced HPA axis hyperactivation ( Melon et al., 2018). We used the Kcc2/Crh KO mice to examine the impact on epilepsy outcomes, including seizure frequency/burden, comorbid behavioral deficits, and sudden unexpected death in epilepsy (SUDEP) risk. We found sex differences in HPA axis dysfunction's effect on chronically epileptic KCC2/Crh KO mice seizure burden, vulnerability to comorbid behavioral deficits, and SUDEP. Suppressing HPA axis hyperexcitability in this model using pharmacological or chemogenetic approaches decreased SUDEP incidence, suggesting that HPA axis dysfunction may contribute to SUDEP. Altered neuroendocrine markers were present in SUDEP cases compared with people with epilepsy or individuals without epilepsy. Together, these findings implicate HPA axis dysfunction in the pathophysiological mechanisms contributing to psychiatric comorbidities in epilepsy and SUDEP.


Sujet(s)
Corticolibérine , Axe hypothalamohypophysaire , Souris knockout , Axe hypophyso-surrénalien , Mort subite et inexpliquée en épilepsie , Animaux , Axe hypothalamohypophysaire/métabolisme , Axe hypophyso-surrénalien/métabolisme , Mâle , Femelle , Souris , Corticolibérine/métabolisme , Caractères sexuels , Épilepsie/métabolisme , Épilepsie/physiopathologie , , Symporteurs/métabolisme , Modèles animaux de maladie humaine , Souris de lignée C57BL , Humains , Facteurs sexuels
15.
Biochem Biophys Res Commun ; 726: 150269, 2024 Sep 24.
Article de Anglais | MEDLINE | ID: mdl-38909533

RÉSUMÉ

Mitochondrial dysfunction is implicated in a wide range of human disorders including many neurodegenerative and cardiovascular diseases, metabolic diseases, cancers, and respiratory disorders. Studies have suggested the potential of l-ergothioneine (ET), a unique dietary thione, to prevent mitochondrial damage and improve disease outcome. Despite this, no studies have definitively demonstrated uptake of ET into mitochondria. Moreover, the expression of the known ET transporter, OCTN1, on the mitochondria remains controversial. In this study, we utilise mass spectrometry to demonstrate direct ET uptake in isolated mitochondria as well as its presence in mitochondria isolated from ET-treated cells and animals. Mitochondria isolated from OCTN1 knockout mice tissues, have impaired but still detectable ET uptake, raising the possibility of alternative transporter(s) which may facilitate ET uptake into the mitochondria. Our data confirm that ET can enter mitochondria, providing a basis for further work on ET in the prevention of mitochondrial dysfunction in human disease.


Sujet(s)
Ergothionéine , Souris knockout , Mitochondries , Ergothionéine/métabolisme , Ergothionéine/pharmacologie , Animaux , Mitochondries/métabolisme , Humains , Souris , Transporteurs de cations organiques/métabolisme , Transporteurs de cations organiques/génétique , Symporteurs/métabolisme , Symporteurs/génétique
16.
Sci Adv ; 10(26): eadn4508, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38924407

RÉSUMÉ

Once considered as a "metabolic waste," lactate is now recognized as a major fuel for tricarboxylic acid (TCA) cycle. Our metabolic flux analysis reveals that skeletal muscle mainly uses lactate to fuel TCA cycle. Lactate is transported through the cell membrane via monocarboxylate transporters (MCTs) in which MCT1 is highly expressed in the muscle. We analyzed how MCT1 affects muscle functions using mice with specific deletion of MCT1 in skeletal muscle. MCT1 deletion enhances running performance, increases oxidative fibers while decreasing glycolytic fibers, and enhances flux of glucose to TCA cycle. MCT1 deficiency increases the expression of mitochondrial proteins, augments cell respiration rate, and elevates mitochondrial activity in the muscle. Mechanistically, the protein level of PGC-1α, a master regulator of mitochondrial biogenesis, is elevated upon loss of MCT1 via increases in cellular NAD+ level and SIRT1 activity. Collectively, these results demonstrate that MCT1-mediated lactate shuttle plays a key role in regulating muscle functions by modulating mitochondrial biogenesis and TCA flux.


Sujet(s)
Cycle citrique , Acide lactique , Transporteurs d'acides monocarboxyliques , Muscles squelettiques , Biogenèse des organelles , Symporteurs , Animaux , Transporteurs d'acides monocarboxyliques/métabolisme , Transporteurs d'acides monocarboxyliques/génétique , Muscles squelettiques/métabolisme , Symporteurs/métabolisme , Symporteurs/génétique , Acide lactique/métabolisme , Souris , Mitochondries/métabolisme , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Souris knockout , Glycolyse
17.
Nat Cell Biol ; 26(7): 1047-1061, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38839979

RÉSUMÉ

The lysosomal degradation of macromolecules produces diverse small metabolites exported by specific transporters for reuse in biosynthetic pathways. Here we deorphanized the major facilitator superfamily domain containing 1 (MFSD1) protein, which forms a tight complex with the glycosylated lysosomal membrane protein (GLMP) in the lysosomal membrane. Untargeted metabolomics analysis of MFSD1-deficient mouse lysosomes revealed an increase in cationic dipeptides. Purified MFSD1 selectively bound diverse dipeptides, while electrophysiological, isotope tracer and fluorescence-based studies in Xenopus oocytes and proteoliposomes showed that MFSD1-GLMP acts as a uniporter for cationic, neutral and anionic dipeptides. Cryoelectron microscopy structure of the dipeptide-bound MFSD1-GLMP complex in outward-open conformation characterized the heterodimer interface and, in combination with molecular dynamics simulations, provided a structural basis for its selectivity towards diverse dipeptides. Together, our data identify MFSD1 as a general lysosomal dipeptide uniporter, providing an alternative route to recycle lysosomal proteolysis products when lysosomal amino acid exporters are overloaded.


Sujet(s)
Dipeptides , Lysosomes , Lysosomes/métabolisme , Animaux , Dipeptides/métabolisme , Ovocytes/métabolisme , Cryomicroscopie électronique , Souris , Xenopus laevis , Humains , Souris knockout , Simulation de dynamique moléculaire , Symporteurs/métabolisme , Symporteurs/génétique , Symporteurs/composition chimique , Femelle , Canaux cationiques TRP
18.
J Biol Chem ; 300(7): 107427, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38823641

RÉSUMÉ

Salmonella enterica serovar Typhimurium melibiose permease (MelBSt) is a prototype of the major facilitator superfamily (MFS) transporters, which play important roles in human health and diseases. MelBSt catalyzed the symport of galactosides with Na+, Li+, or H+ but prefers the coupling with Na+. Previously, we determined the structures of the inward- and outward-facing conformation of MelBSt and the molecular recognition for galactoside and Na+. However, the molecular mechanisms for H+- and Na+-coupled symport remain poorly understood. In this study, we solved two x-ray crystal structures of MelBSt, the cation-binding site mutants D59C at an unliganded apo-state and D55C at a ligand-bound state, and both structures display the outward-facing conformations virtually identical as published. We determined the energetic contributions of three major Na+-binding residues for the selection of Na+ and H+ by free energy simulations. Transport assays showed that the D55C mutant converted MelBSt to a solely H+-coupled symporter, and together with the free-energy perturbation calculation, Asp59 is affirmed to be the sole protonation site of MelBSt. Unexpectedly, the H+-coupled melibiose transport exhibited poor activities at greater bulky ΔpH and better activities at reversal ΔpH, supporting the novel theory of transmembrane-electrostatically localized protons and the associated membrane potential as the primary driving force for the H+-coupled symport mediated by MelBSt. This integrated study of crystal structure, bioenergetics, and free energy simulations, demonstrated the distinct roles of the major binding residues in the cation-binding pocket of MelBSt.


Sujet(s)
Sodium , Symporteurs , Symporteurs/composition chimique , Symporteurs/métabolisme , Symporteurs/génétique , Sites de fixation , Cristallographie aux rayons X , Sodium/métabolisme , Sodium/composition chimique , Protéines bactériennes/composition chimique , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique , Salmonella typhimurium/métabolisme , Salmonella typhimurium/génétique , Salmonella typhimurium/composition chimique , Melibiose/métabolisme , Melibiose/composition chimique , Cations/métabolisme , Cations/composition chimique , Conformation des protéines
19.
Mol Med ; 30(1): 83, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867145

RÉSUMÉ

BACKGROUND: The terminal stage of ischemic heart disease develops into heart failure (HF), which is characterized by hypoxia and metabolic disturbances in cardiomyocytes. The hypoxic failing heart triggers hypoxia-inducible factor-1α (HIF-1α) actions in the cells sensitized to hypoxia and induces metabolic adaptation by accumulating HIF-1α. Furthermore, soluble monocarboxylic acid transporter protein 1 (MCT1) and mitochondrial pyruvate carrier 1 (MPC1), as key nodes of metabolic adaptation, affect metabolic homeostasis in the failing rat heart. Aerobic exercise training has been reported to retard the progression of HF due to enhancing HIF-1α levels as well as MCT1 expressions, whereas the effects of exercise on MCT1 and MPC1 in HF (hypoxia) remain elusive. This research aimed to investigate the action of exercise associated with MCT1 and MPC1 on HF under hypoxia. METHODS: The experimental rat models are composed of four study groups: sham stented (SHAM), HF sedentary (HF), HF short-term exercise trained (HF-E1), HF long-term exercise trained (HF-E2). HF was initiated via left anterior descending coronary artery ligation, the effects of exercise on the progression of HF were analyzed by ventricular ultrasound (ejection fraction, fractional shortening) and histological staining. The regulatory effects of HIF-1α on cell growth, MCT1 and MPC1 protein expression in hypoxic H9c2 cells were evaluated by HIF-1α activatort/inhibitor treatment and plasmid transfection. RESULTS: Our results indicate the presence of severe pathological remodelling (as evidenced by deep myocardial fibrosis, increased infarct size and abnormal hypertrophy of the myocardium, etc.) and reduced cardiac function in the failing hearts of rats in the HF group compared to the SHAM group. Treadmill exercise training ameliorated myocardial infarction (MI)-induced cardiac pathological remodelling and enhanced cardiac function in HF exercise group rats, and significantly increased the expression of HIF-1α (p < 0.05), MCT1 (p < 0.01) and MPC1 (p < 0.05) proteins compared to HF group rats. Moreover, pharmacological inhibition of HIF-1α in hypoxic H9c2 cells dramatically downregulated MCT1 and MPC1 protein expression. This phenomenon is consistent with knockdown of HIF-1α at the gene level. CONCLUSION: The findings propose that long-term aerobic exercise training, as a non- pharmacological treatment, is efficient enough to debilitate the disease process, improve the pathological phenotype, and reinstate cardiac function in HF rats. This benefit is most likely due to activation of myocardial HIF-1α and upregulation of MCT1 and MPC1.


Sujet(s)
Défaillance cardiaque , Sous-unité alpha du facteur-1 induit par l'hypoxie , Transporteurs d'acides monocarboxyliques , Conditionnement physique d'animal , Symporteurs , Animaux , Mâle , Rats , Modèles animaux de maladie humaine , Régulation de l'expression des gènes , Défaillance cardiaque/métabolisme , Défaillance cardiaque/génétique , Défaillance cardiaque/étiologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Protéines de transport de la membrane mitochondriale/métabolisme , Protéines de transport de la membrane mitochondriale/génétique , Transporteurs d'acides monocarboxyliques/métabolisme , Transporteurs d'acides monocarboxyliques/génétique , Myocytes cardiaques/métabolisme , Rat Sprague-Dawley , Symporteurs/métabolisme , Symporteurs/génétique , Régulation positive
20.
Neurobiol Dis ; 199: 106572, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38901782

RÉSUMÉ

Within the adult mouse subventricular zone (SVZ), neural stem cells (NSCs) produce neuroblasts and oligodendrocyte precursor cells (OPCs). T3, the active thyroid hormone, influences renewal and commitment of SVZ progenitors. However, how regulators of T3 availability affect these processes is less understood. Using Mct8/Dio2 knockout mice, we investigated the role of MCT8, a TH transporter, and DIO2, the T3-generating enzyme, in regulating adult SVZ-neurogliogenesis. Single-cell RNA-Seq revealed Mct8 expression in various SVZ cell types in WT mice, while Dio2 was enriched in neurons, astrocytes, and quiescent NSCs. The absence of both regulators in the knockout model dysregulated gene expression, increased the neuroblast/OPC ratio and hindered OPC differentiation. Immunostainings demonstrated compromised neuroblast migration reducing their supply to the olfactory bulbs, impairing interneuron differentiation and odor discrimination. These findings underscore the pivotal roles of MCT8 and DIO2 in neuro- and oligodendrogenesis, offering targets for therapeutic avenues in neurodegenerative and demyelinating diseases.


Sujet(s)
Ventricules latéraux , Souris knockout , Transporteurs d'acides monocarboxyliques , Cellules souches neurales , Neurogenèse , Animaux , Neurogenèse/physiologie , Souris , Transporteurs d'acides monocarboxyliques/génétique , Transporteurs d'acides monocarboxyliques/métabolisme , Cellules souches neurales/métabolisme , Ventricules latéraux/métabolisme , , Iodide peroxidase/génétique , Iodide peroxidase/métabolisme , Différenciation cellulaire/physiologie , Symporteurs/génétique , Symporteurs/métabolisme , Bulbe olfactif/métabolisme , Souris de lignée C57BL , Précurseurs des oligodendrocytes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE