Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Biochem Soc Trans ; 52(3): 1293-1304, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38716884

RÉSUMÉ

ATP has recently been reconsidered as a molecule with functional properties which go beyond its recognized role of the energetic driver of the cell. ATP has been described as an allosteric modulator as well as a biological hydrotrope with anti-aggregation properties in the crowded cellular environment. The role of ATP as a modulator of the homeostasis of the neurotrophins (NTs), a growth factor protein family whose most known member is the nerve growth factor (NGF), has been investigated. The modulation of NTs by small endogenous ligands is still a scarcely described area, with few papers reporting on the topic, and very few reports on the molecular determinants of these interactions. However, a detailed atomistic description of the NTs interaction landscape is of urgent need, aiming at the identification of novel molecules as potential therapeutics and considering the wide range of potential pharmacological applications for NGF and its family members. This mini-review will focus on the unique cartography casting the interactions of the endogenous ligand ATP, in the interaction with NGF as well as with its precursor proNGF. These interactions revealed interesting features of the ATP binding and distinct differences in the binding mode between the highly structured mature NGF and its precursor, proNGF, which is characterized by an intrinsically unstructured domain. The overview on the recent available data will be presented, together with the future perspectives on the field.


Sujet(s)
Adénosine triphosphate , Facteur de croissance nerveuse , Liaison aux protéines , Facteur de croissance nerveuse/métabolisme , Adénosine triphosphate/métabolisme , Humains , Animaux , Précurseurs de protéines/métabolisme , Facteurs de croissance nerveuse/métabolisme , Facteurs de croissance nerveuse/composition chimique , Ligands , Sites de fixation
2.
Heliyon ; 10(7): e28818, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38590871

RÉSUMÉ

G protein-coupled receptors (GPCRs), which form the largest family of membrane protein receptors in humans, are highly complex signaling systems with intricate structures and dynamic conformations and locations. Among these receptors, a specific subset is referred to as orphan GPCRs (oGPCRs) and has garnered significant interest in pain research due to their role in both central and peripheral nervous system function. The diversity of GPCR functions is attributed to multiple factors, including allosteric modulators, signaling bias, oligomerization, constitutive signaling, and compartmentalized signaling. This review primarily focuses on the recent advances in oGPCR research on pain mechanisms, discussing the role of specific oGPCRs including GPR34, GPR37, GPR65, GPR83, GPR84, GPR85, GPR132, GPR151, GPR160, GPR171, GPR177, and GPR183. The orphan receptors among these receptors associated with central nervous system diseases are also briefly described. Understanding the functions of these oGPCRs can contribute not only to a deeper understanding of pain mechanisms but also offer a reference for discovering new targets for pain treatment.

3.
Biol Pharm Bull ; 47(3): 580-590, 2024.
Article de Anglais | MEDLINE | ID: mdl-38432913

RÉSUMÉ

There are 48 nuclear receptors in the human genome, and many members of this superfamily have been implicated in human diseases. The NR4A nuclear receptor family consisting of three members, NR4A1, NR4A2, and NR4A3 (formerly annotated as Nur77, Nurr1, and NOR1, respectively), are still orphan receptors but exert pathological effects on immune-related and neurological diseases. We previously reported that prostaglandin A1 (PGA1) and prostaglandin A2 (PGA2) are potent activators of NR4A3, which bind directly to the ligand-binding domain (LBD) of the receptor. Recently, the co-crystallographic structures of NR4A2-LBD bound to PGA1 and PGA2 were reported, followed by reports of the neuroprotective effects of these possible endogenous ligands in mouse models of Parkinson's disease. Based on these structures, we modeled the binding structures of the other two members (NR4A1 and NR4A3) with these potential endogenous ligands using a template-based modeling method, and reviewed the similarity and diversity of ligand-binding mechanisms in the nuclear receptor family.


Sujet(s)
Maladie de Parkinson , Humains , Animaux , Souris , Ligands , Modèles animaux de maladie humaine , Domaines protéiques , Prostaglandines
4.
Yakugaku Zasshi ; 142(3): 195-203, 2022.
Article de Japonais | MEDLINE | ID: mdl-35228371

RÉSUMÉ

Chronic inflammation plays an important role in the pathogenesis of obesity and metabolic disorders. In obesity, pattern-recognition receptors in innate immune system, such as Toll-like receptor 4 (TLR4), cause chronic inflammation through prolonged activation by various endogenous ligands, including fatty acids and its metabolites. Gangliosides and other glycosphingolipids are important metabolites of fatty acids and saccharides. GM3, the simplest ganglioside comprising α2,3-sialyllactose, is expressed in insulin-sensitive peripheral tissues such as liver and adipose tissue, and furthermore secreted abundantly into serum. It has been shown that GM3 regulates the signal transduction of insulin receptor in adipose tissue as a component of membrane microdomains, and elevation in GM3 level causes insulin resistance. However, the homeostatic and pathophysiological functions of extracellularly secreted GM3 are poorly understood. We recently reported that GM3 species with differing fatty acid structures act as pro- and anti-inflammatory endogenous TLR4 ligands. GM3 with very long-chain fatty acid (VLCFA) and α-hydroxyl VLCFA strongly enhanced TLR4 activation. Conversely, GM3 with long-chain fatty acid (LCFA) and ω-9 unsaturated VLCFA inhibited TLR4 activation, counteracting the VLCFA species. GM3 interacted with the extracellular complex of TLR4 and promoted dimerization/oligomerization. In obesity and metabolic disorders, VLCFA species were increased in serum and adipose tissue, whereas LCFA species was relatively decreased; their imbalances were correlated to disease progression. Our findings suggest that GM3 species are disease-related endogenous TLR4 ligands, and "glycosphingolipid sensing" by TLR4 controls the homeostatic and pathological roles of innate immune signaling.


Sujet(s)
Ganglioside GM3/physiologie , Régulation de l'expression des gènes/génétique , Régulation de l'expression des gènes/physiologie , Homéostasie/génétique , Homéostasie/physiologie , Maladies métaboliques/étiologie , Obésité/étiologie , Transduction du signal/génétique , Transduction du signal/physiologie , Récepteur de type Toll-4/génétique , Récepteur de type Toll-4/métabolisme , Acides gras/métabolisme , Ganglioside GM3/composition chimique , Ganglioside GM3/métabolisme , Humains , Immunité innée , Inflammation , Ligands , Maladies métaboliques/génétique , Obésité/génétique , Récepteur à l'insuline/métabolisme
5.
Eur J Med Chem ; 231: 114154, 2022 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-35124532

RÉSUMÉ

Large numbers of diverse biologically active molecules are produced from phospholipids, the constituents of biological membranes. Indeed, many lipid-derived ligands, which can undergo inter-transformation between one and another by certain kinases or enzymes, bind to protein receptors such as G-protein-coupled receptors, and serve to regulate multiple biological processes through a variety of signaling pathways. Thus, lipid mediators are likely involved in a synergistic regulatory network, and dysfunction of this network may result in diseases. Here, we reviewed recent progress in the drug development targeting related receptors, focusing on the identification of common structural features which can both come from endogenous ligands or artificial ligands. We also discussed how these features have been utilized in drug design and relevant issues such as potency, selectivity, metabolic stability, and toxicity.


Sujet(s)
Récepteurs artificiels , Ligands , Lipides , Récepteurs couplés aux protéines G/métabolisme , Transduction du signal
6.
Proc Natl Acad Sci U S A ; 118(30)2021 07 27.
Article de Anglais | MEDLINE | ID: mdl-34290146

RÉSUMÉ

Many endogenous molecules, mostly proteins, purportedly activate the Toll-like receptor 4 (TLR4)-myeloid differentiation factor-2 (MD-2) complex, the innate immune receptor for lipopolysaccharide (LPS) derived from gram-negative bacteria. However, there is no structural evidence supporting direct TLR4-MD-2 activation by endogenous ligands. Sulfatides (3-O-sulfogalactosylceramides) are natural, abundant sulfated glycolipids that have variously been shown to initiate or suppress inflammatory responses. We show here that short fatty acid (FA) chain sulfatides directly activate mouse TLR4-MD-2 independent of CD14, trigger MyD88- and TRIF-dependent signaling, and stimulate tumor necrosis factor α (TNFα) and type I interferon (IFN) production in mouse macrophages. In contrast to the agonist activity toward the mouse receptor, the tested sulfatides antagonize TLR4-MD-2 activation by LPS in human macrophage-like cells. The agonistic and antagonistic activities of sulfatides require the presence of the sulfate group and are inversely related to the FA chain length. The crystal structure of mouse TLR4-MD-2 in complex with C16-sulfatide revealed that three C16-sulfatide molecules bound to the MD-2 hydrophobic pocket and induced an active dimer conformation of the receptor complex similar to that induced by LPS or lipid A. The three C16-sulfatide molecules partially mimicked the detailed interactions of lipid A to achieve receptor activation. Our results suggest that sulfatides may mediate sterile inflammation or suppress LPS-stimulated inflammation, and that additional endogenous negatively charged lipids with up to six lipid chains of limited length might also bind to TLR4-MD-2 and activate or inhibit this complex.


Sujet(s)
Protéines adaptatrices du transport vésiculaire/métabolisme , Antigène lymphocytaire-96/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Sulfoglycosphingolipides/pharmacologie , Récepteur de type Toll-4/métabolisme , Protéines adaptatrices du transport vésiculaire/génétique , Animaux , Lignée cellulaire , Femelle , Humains , Antigène lymphocytaire-96/génétique , Souris , Souris de lignée C57BL , Souris knockout , Simulation de dynamique moléculaire , Facteur de différenciation myéloïde-88/génétique , Sulfoglycosphingolipides/composition chimique , Récepteur de type Toll-4/génétique
7.
Stem Cell Res Ther ; 11(1): 541, 2020 12 14.
Article de Anglais | MEDLINE | ID: mdl-33317626

RÉSUMÉ

BACKGROUND: Mesenchymal stem cells (MSCs) have exerted their brilliant potential to promote heart repair following myocardial infarction. However, low survival rate of MSCs after transplantation due to harsh conditions with hypoxic and ischemic stress limits their therapeutic efficiency in treating cardiac dysfunction. ELABELA (ELA) serves as a peptide hormone which has been proved to facilitate cell growth, survival, and pluripotency in human embryonic stem cells. Although ELA works as an endogenous ligand of a G protein-coupled receptor APJ (Apelin receptor, APLNR), whether APJ is an essential signal for the function of ELA remains elusive. The effect of ELA on apoptosis of MSCs is still vague. OBJECTIVE: We studied the role of ELABELA (ELA) treatment on the anti-apoptosis of MSCs in hypoxic/ischemic (H/I) conditions which mimic the impaired myocardial microenvironment and explored the possible mechanisms in vitro. METHODS: MSCs were obtained from donated rats weighing between 80~120 g. MSCs were exposed to serum-free and hypoxic (1% O2) environments for 24 h, which mimics hypoxic/ischemic damage in vivo, using serum-containing normoxic conditions (20% O2) as a negative control. MSCs that were exposed to H/I injury with ELA processing were treated by 5 µM of ELA. Cell viability and apoptosis of MSCs were evaluated by CCK8 and flow cytometry, respectively. Mitochondrial function of MSCs was also assessed according to mitochondrial membrane potential (MMP) and ATP content. The protein expression of key kinases of the PI3K/AKT and ERK1/2 signaling pathways involving t-AKT, p-AKT, t-ERK1/2, and p-ERK1/2, as well as apoptosis-related protein expression of Bcl-2, Bax, and cleaved Caspase 3, were monitored by Western blot. RESULTS: We found that ELA treatment of H/I-induced MSCs improved overall cell viability, enhanced Bcl/Bax expression, and decreased Caspase 3 activity. ELA inhibited H/I-induced mitochondrial dysfunction by increasing ATP concentration and suppressing the loss of mitochondrial transmembrane potential. However, this anti-apoptotic property of ELA was restrained in APJ-silenced MSCs. Additionally, ELA treatment induced the phosphorylation of AKT and ERK, while the blockade of PI3K/AKT and ERK1/2 pathways with respective inhibitors, LY294002 and U0126, suppressed the action of ELA. CONCLUSION: ELA positively affected on the survival of MSCs and exhibited anti-apoptotic characteristics when exposed to hypoxic/ischemic condition in vitro. Also, the function of ELA was correlated with the APJ receptor, reduced mitochondrial damage, and activation of the PI3K/AKT and ERK1/2 signal axes.


Sujet(s)
Système de signalisation des MAP kinases , Cellules souches mésenchymateuses , Animaux , Apoptose , Cellules souches mésenchymateuses/métabolisme , Mitochondries/métabolisme , Hormones peptidiques , Phosphatidylinositol 3-kinases/génétique , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Rats
8.
Neurochem Int ; 140: 104814, 2020 11.
Article de Anglais | MEDLINE | ID: mdl-32758586

RÉSUMÉ

Peroxisome proliferator-activated receptor gamma (PPAR-γ) is one of the ligand-activated transcription factors which regulates a number of central events and considered as a promising target for various neurodegenerative disease conditions. Numerous reports implicate that PPAR-γ agonists have shown neuroprotective effects by regulating genes transcription associated with the pathogenesis of neurodegeneration. In regards, this review critically appraises the recent knowledge of PPAR-γ receptors in neuroprotection in order to hypothesize potential neuroprotective mechanism of PPAR-γ agonism in chronic neurological conditions. Of note, the PPAR-γ's interaction dynamics with PPAR-γ coactivator-1α (PGC-1α) has gained significant attention for neuroprotection. Likewise, a plethora of studies suggest that the PPAR-γ pathway can be actuated by the endogenous ligands present in the CNS and thus identification and development of novel agonist for the PPAR-γ receptor holds a vow to prevent neurodegeneration. Together, the critical insights of this review enlighten the translational possibilities of developing novel neuroprotective therapeutics targeting PPAR-γ for various neurodegenerative disease conditions.


Sujet(s)
Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/prévention et contrôle , Neuroprotection/physiologie , Récepteur PPAR gamma/agonistes , Récepteur PPAR gamma/métabolisme , Animaux , Humains , Mitochondries/génétique , Mitochondries/métabolisme , Maladies neurodégénératives/génétique , Stress oxydatif/physiologie , Récepteur PPAR gamma/génétique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme
9.
Cell ; 179(4): 895-908.e21, 2019 10 31.
Article de Anglais | MEDLINE | ID: mdl-31675498

RÉSUMÉ

The peptidergic system is the most abundant network of ligand-receptor-mediated signaling in humans. However, the physiological roles remain elusive for numerous peptides and more than 100 G protein-coupled receptors (GPCRs). Here we report the pairing of cognate peptides and receptors. Integrating comparative genomics across 313 species and bioinformatics on all protein sequences and structures of human class A GPCRs, we identify universal characteristics that uncover additional potential peptidergic signaling systems. Using three orthogonal biochemical assays, we pair 17 proposed endogenous ligands with five orphan GPCRs that are associated with diseases, including genetic, neoplastic, nervous and reproductive system disorders. We also identify additional peptides for nine receptors with recognized ligands and pathophysiological roles. This integrated computational and multifaceted experimental approach expands the peptide-GPCR network and opens the way for studies to elucidate the roles of these signaling systems in human physiology and disease. VIDEO ABSTRACT.


Sujet(s)
Génomique , Peptides/génétique , Conformation des protéines , Récepteurs couplés aux protéines G/génétique , Séquence d'acides aminés/génétique , Biologie informatique , Réseaux de régulation génique/génétique , Système génital/métabolisme , Système génital/anatomopathologie , Humains , Ligands , Tumeurs/génétique , Tumeurs/anatomopathologie , Maladies du système nerveux/génétique , Maladies du système nerveux/anatomopathologie , Transduction du signal/génétique
10.
Crit Rev Toxicol ; 48(7): 555-574, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-30226107

RÉSUMÉ

The aryl hydrocarbon receptor (AHR) is not essential to survival, but does act as a key regulator of many normal physiological events. The role of this receptor in toxicological processes has been studied extensively, primarily employing the high-affinity ligand 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). However, regulation of physiological responses by endogenous AHR ligands remains to be elucidated. Here, we review developments in this field, with a focus on 6-formylindolo[3,2-b]carbazole (FICZ), the endogenous ligand with the highest affinity to the receptor reported to date. The binding of FICZ to different isoforms of the AHR seems to be evolutionarily well conserved and there is a feedback loop that controls AHR activity through metabolic degradation of FICZ via the highly inducible cytochrome P450 1A1. Several investigations provide strong evidence that FICZ plays a critical role in normal physiological processes and can ameliorate immune diseases with remarkable efficiency. Low levels of FICZ are pro-inflammatory, providing resistance to pathogenic bacteria, stimulating the anti-tumor functions, and promoting the differentiation of cancer cells by repressing genes in cancer stem cells. In contrast, at high concentrations FICZ behaves in a manner similar to TCDD, exhibiting toxicity toward fish and bird embryos, immune suppression, and activation of cancer progression. The findings are indicative of a dual role for endogenously activated AHR in barrier tissues, aiding clearance of infections and suppressing immunity to terminate a vicious cycle that might otherwise lead to disease. There is not much support for the AHR ligand-specific immune responses proposed, the differences between FICZ and TCDD in this context appear to be explained by the rapid metabolism of FICZ.


Sujet(s)
Carbazoles/métabolisme , Différenciation cellulaire , Prolifération cellulaire , Récepteurs à hydrocarbure aromatique/métabolisme , Transduction du signal , Animaux , Humains
11.
J Reprod Immunol ; 121: 26-33, 2017 06.
Article de Anglais | MEDLINE | ID: mdl-28558299

RÉSUMÉ

Pre-eclampsia (PE) is characterized by an exaggerated systemic inflammatory response and generalized endothelial dysfunction. We have recently demonstrated that fibrinogen, an endogenous ligand of Toll-like receptor (TLR) 4, activates monocytes from women with pre-eclampsia (Al-ofi et al., 2014). Using an experimental co-culture model of primary human monocytes (derived from 9 women with PE (GA=33.18±5.8) and 9 normotensive pregnant women, NP (GA=33.15±4.0)) and human umbilical venous endothelial cells (HUVECs), we compared the effects of fibrinogen and lipopolysaccharide (LPS, bacterial ligand to TLR4) on the expression levels of inflammatory cytokines (IL-6 and IL-1ß), chemokines (IL-8 and MCP-1), and anti-angiogenic factor (soluble fms-like tyrosine kinase-1,sFLT-1), as well as the soluble vascular cell adhesion molecule-1 (sVCAM-1). Cytokines, VEGF and sVCAM-1 were measured in the supernatant media by cytometric array. The levels of sFLT-1 were measured by ELISA. Fibrinogen induced greater expression levels of IL-1ß and VCAM-1 from PE HUVEC-monocyte co-culture than from NP HUVEC-monocyte co-culture (P<0.05), similar to the effects of LPS. In contrast, unlike LPS, fibrinogen suppressed IL-6, IL-8, MCP-1 and sFLT-1 production by co-cultures that included PE monocytes compared to those with NP monocytes (P<0.05). In conclusion, fibrinogen promotes monocyte-endothelial cell adhesion and angiogenesis and suppresses the expression of some inflammatory markers in pre-eclampsia. Although the physiological implications of these intriguing observations are unclear our findings suggest that fibrinogen contributes to the regulation of cell adhesion, angiogenesis and inflammation by mechanisms not wholly dependent on TLR4 stimulation.


Sujet(s)
Cellules endothéliales/physiologie , Inflammation/immunologie , Monocytes/physiologie , Pré-éclampsie/immunologie , Adulte , Marqueurs biologiques/métabolisme , Adhérence cellulaire , Techniques de coculture , Cytokines/métabolisme , Femelle , Fibrinogène/immunologie , Cellules endothéliales de la veine ombilicale humaine , Humains , Médiateurs de l'inflammation/métabolisme , Ligands , Lipopolysaccharides/immunologie , Néovascularisation physiologique , Grossesse , Récepteur de type Toll-2/métabolisme , Récepteur de type Toll-4/métabolisme
12.
Exp Mol Pathol ; 102(2): 203-209, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28161441

RÉSUMÉ

BACKGROUND: Mesenchymal stem cells (MSCs) transplantation has been regarded as an optimal therapeutic approach for cardiovascular disease. However, the inferior survival and low vascularization potential of these cells in the local infarct site reduce the therapeutic efficacy. In this study, we investigated the influence of apelin on MSCs survival and vascularization under hypoxic-ischemic condition in vitro and explored the relevant mechanism. METHODS: MSCs were obtained from C57BL/6 mice and cultured in vitro. Cells of the third passage were divided into MSCs and MSCs+apelin groups. In the MSCs+apelin group, MSCs were stimulated with apelin-13 (5µM). The two groups experienced exposure to hypoxia (1% O2) and serum deprivation for 24h, using normoxia (20% O2) as a negative control during the process. Human umbilical vein endothelial cells (HUVECs) were used and incubated with conditioned media from both groups to promote vascularization for another 6h. Vascular densities were assessed and relevant biomarkers were detected thereafter. RESULTS: Compared with MSCs group, MSCs+apelin group presented more rapid growth. The proliferation rate was much higher. Cells apoptosis percentage was significantly declined both under normoxic and hypoxic conditions. Media produced from MSCs+apelin group triggered HUVECs to form a larger number of vascular branches on matrigel. The expression and secretion of vascular endothelial growth factor (VEGF) were significantly increased. CONCLUSION: Apelin could effectively promote MSCs survival and vascularization under hypoxic-ischemic condition in vitro, and this procedure was associated with the upregulation of VEGF. This study provides a new perspective for exploring novel approaches to enhance MSCs survival and vascularization potential.


Sujet(s)
Survie cellulaire/effets des médicaments et des substances chimiques , Protéines et peptides de signalisation intercellulaire/pharmacologie , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Néovascularisation pathologique/traitement médicamenteux , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Animaux , Apoptose , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire , Cellules cultivées , Milieux de culture conditionnés/composition chimique , Cellules endothéliales de la veine ombilicale humaine , Humains , Cellules souches mésenchymateuses/cytologie , Souris , Souris de lignée C57BL , Régulation positive , Facteur de croissance endothéliale vasculaire de type A/génétique
13.
J Lipid Res ; 58(2): 393-402, 2017 02.
Article de Anglais | MEDLINE | ID: mdl-28011707

RÉSUMÉ

Liver X receptor (LXR)α is a nuclear receptor that responds to oxysterols and cholesterol overload by stimulating cholesterol efflux, transport, conversion to bile acids, and excretion. LXRα binds to and is regulated by synthetic (T-0901317, GW3695) and endogenous (oxysterols) ligands. LXRα activity is also modulated by FAs, but the ligand binding specificity of FA and acyl-CoA derivatives for LXRα remains unknown. We investigated whether LXRα binds FA or FA acyl-CoA with affinities that mimic in vivo concentrations, examined the effect of FA chain length and the degree of unsaturation on binding, and investigated whether FAs regulate LXRα activation. Saturated medium-chain FA (MCFA) displayed binding affinities in the low nanomolar concentration range, while long-chain fatty acyl-CoA did not bind or bound weakly to LXRα. Circular dichroic spectra and computational docking experiments confirmed that MCFA bound to the LXRα ligand binding pocket similar to the known synthetic agonist of LXRα (T0901317), but with limited change to the conformation of the receptor. Transactivation assays showed that MCFA activated LXRα, whereas long-chain FA caused no effect. Our results suggest that LXRα functions as a receptor for saturated FA or acyl-CoA of C10 and C12 in length.


Sujet(s)
Acyl coenzyme A/métabolisme , Cholestérol/métabolisme , Acides gras/métabolisme , Récepteurs hépatiques X/métabolisme , Acyl coenzyme A/composition chimique , Animaux , Cellules COS , Chlorocebus aethiops , Cholestérol/composition chimique , Acides gras/composition chimique , Humains , Hydrocarbures fluorés/composition chimique , Hydrocarbures fluorés/métabolisme , Ligands , Oxystérols/composition chimique , Oxystérols/métabolisme , Liaison aux protéines , Sulfonamides/composition chimique , Sulfonamides/métabolisme
14.
Trends Neurosci ; 39(12): 830-839, 2016 12.
Article de Anglais | MEDLINE | ID: mdl-27793433

RÉSUMÉ

Currently, drug discovery focusses only on quantifying pharmacological parameters, sometimes including binding kinetics, of drug candidates. For a complete understanding of a drug's desired binding kinetics, the kinetics of both the target and its endogenous ligands should be considered. This is because the release and binding kinetics of endogenous ligands in addition to receptor internalization rates are significant contributors to drug-target interactions. Here, we discuss the kinetic profile of three neuropeptides and their receptors; gonadotropin-releasing hormone receptor (GnRHR), neuropeptide Y receptors, and corticotropin-releasing factor receptor 1 (CRF1R). These three examples provide new insights into the importance of kinetic profiles which could improve the understanding of desired drug-target binding kinetics and advance drug discovery for various neurological and psychiatric illnesses.


Sujet(s)
Neuropeptides/métabolisme , Récepteur aux neuropeptides/métabolisme , Animaux , Découverte de médicament , Humains , Cinétique
15.
Front Cell Dev Biol ; 4: 45, 2016.
Article de Anglais | MEDLINE | ID: mdl-27243009

RÉSUMÉ

Traditionally considered as a critical intermediate in the toxic and carcinogenic response to dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin, TCDD), the Aryl hydrocarbon/Dioxin receptor (AhR) has proven to be also an important regulator of cell physiology and organ homeostasis. AhR has become an interesting and actual area of research mainly boosted by a significant number of recent studies analyzing its contribution to the proper functioning of the immune, hepatic, cardiovascular, vascular and reproductive systems. At the cellular level, AhR establishes functional interactions with signaling pathways governing cell proliferation and cell cycle, cell morphology, cell adhesion and cell migration. Two exciting new aspects in AhR biology deal with its implication in the control of cell differentiation and its more than likely involvement in cell pluripotency and stemness. In fact, it is possible that AhR could help modulate the balance between differentiation and pluripotency in normal and transformed tumor cells. At the molecular level, AhR regulates an increasingly large array of physiologically relevant genes either by traditional transcription-dependent mechanisms or by unforeseen processes involving genomic insulators, chromatin dynamics and the transcription of mobile genetic elements. AhR is also closely related to epigenetics, not only from the point of view of target gene expression but also with respect to its own regulation by promoter methylation. It is reasonable to consider that deregulation of these many functions could have a causative role, or at least contribute to, human disease. Consequently, several laboratories have proposed that AhR could be a valuable tool as diagnostic marker and/or therapeutic target in human pathologies. An additional point of interest is the possibility of regulating AhR activity by endogenous non-toxic low weight molecules agonist or antagonist molecules that could be present or included in the diet. In this review, we will address these molecular and functional features of AhR biology within physiological and pathological contexts.

16.
Mol Immunol ; 68(2 Pt A): 94-7, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26141240

RÉSUMÉ

The nature of the endogenous ligands for natural killer T (NKT) cells has been debated for more than a decade. Because the mammalian glycosylceramide synthases are invertases, it is believed that in mammals all glycosylceramides are ß anomers. However, the possibility that an alternative enzymatic pathway, an unfaithful enzyme, or unique physico-chemical environments could allow the production of small quantities of α anomers should be entertained. Classic biochemical and chemical analysis approaches are not well suited for this challenge as they lack sensitivity. Using a combination of biological assays and new technological approaches, we have unequivocally demonstrated that α glycosylceramides were constitutively produced by mammalian immune cells, loaded onto CD1d and presented to NKT cells both in the thymus and in the periphery. Their amount is controlled tightly by catabolic enzymes, and can be altered in vitro and in vivo to modify NKT cell behavior.


Sujet(s)
Cellules présentatrices d'antigène/immunologie , Céramides/immunologie , Cellules tueuses naturelles/immunologie , Thymocytes/immunologie , Animaux , Présentation d'antigène/génétique , Cellules présentatrices d'antigène/cytologie , Antigène CD1d/immunologie , Antigène CD1d/métabolisme , Céramides/composition chimique , Céramides/classification , Céramides/métabolisme , Glucosyltransferases/génétique , Glucosyltransferases/immunologie , Humains , Cellules tueuses naturelles/cytologie , N-Acylsphingosine galactosyltransferase/génétique , N-Acylsphingosine galactosyltransferase/immunologie , Thymocytes/cytologie , Thymus (glande)
17.
Arch Biochem Biophys ; 579: 73-84, 2015 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-26032337

RÉSUMÉ

The gypsy moth utilizes a pheromone, (7R,8S)-2-methyl-7,8-epoxyoctadecane, for mate location. The pheromone is detected by sensory hairs (sensilla) on the antennae of adult males. Sensilla contain the dendrites of olfactory neurons bathed in lymph, which contains pheromone binding proteins (PBPs). We have extracted and identified free fatty acids from lymph of sensory hairs, and we demonstrate that these function as endogenous ligands for gypsy moth PBP1 and PBP2. Homology modeling of both PBPs, and docking of fatty acids reveal multiple binding sites: one internal, the others external. Pheromone binding assays suggest that these fatty acids increase PBP-pheromone binding affinity. We show that fatty acid binding causes an increase in α-helix content in the N-terminal domain, but not in the C-terminal peptide of both proteins. The C-terminal peptide was shown to form a α-helix in a hydrophobic, homogeneous environment, but not in the presence of fatty acid micelles. Through partition assays we show that the fatty acids prevent adsorption of the pheromone on hydrophobic surfaces and facilitate pheromone partition into an aqueous phase. We propose that lymph is an emulsion of fatty acids and PBP that influence each other and thereby control the partition equilibria of hydrophobic odorants.


Sujet(s)
Protéines de transport/métabolisme , Acides gras/métabolisme , Protéines d'insecte/métabolisme , Papillons de nuit/métabolisme , Phéromones/métabolisme , Sensilles/métabolisme , Adsorption , Animaux , Sites de fixation , Protéines de transport/composition chimique , Acides gras/composition chimique , Protéines d'insecte/composition chimique , Protéines et peptides de signalisation intercellulaire , Mâle , Papillons de nuit/composition chimique , Phéromones/composition chimique , Liaison aux protéines , Sensilles/composition chimique , Odorat/physiologie , Relation structure-activité
18.
Biochem Pharmacol ; 98(2): 318-21, 2015 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-26119821

RÉSUMÉ

This article represents a timely opportunity to express my affection, admiration and gratitude to Professor David Triggle. David was my Ph.D. advisor as well as a key consultant in the 1980s and early 1990s for research programs at Miles Institute for Preclinical Pharmacology in West Haven, CT, the U.S. research operation of Bayer AG, in the areas of Ca(2+) and K(+) channel ligands. The binding methodology developed in his laboratory was used to search for an endogenous ligand for L-type Ca(2+) channels. We did not find the substance that we were searching for, a genetically-determined, competitive inhibitor for the 1,4-dihydropyridine binding site, but instead isolated the endogenous ligand for the brain's own marijuana, anandamide. Devane, Mechoulam and coworkers first discovered that this compound was the endogenous ligand for delta-9-tetrahydrocannabinol, the active substance in cannabis. The endogenous endocannabinoid system is now the target of many exciting new approaches to drug discovery.


Sujet(s)
Académies et instituts , Comportement coopératif , Recherche , Académies et instituts/histoire , Animaux , Acides arachidoniques/histoire , Acides arachidoniques/isolement et purification , Acides arachidoniques/métabolisme , Encéphale/métabolisme , Canaux calciques/histoire , Canaux calciques/physiologie , Dihydropyridines/histoire , Dihydropyridines/métabolisme , Découverte de médicament/histoire , Endocannabinoïdes/histoire , Endocannabinoïdes/isolement et purification , Endocannabinoïdes/métabolisme , Histoire du 20ème siècle , Histoire du 21ème siècle , Humains , Ligands , Amides gras polyinsaturés N-alkylés/histoire , Amides gras polyinsaturés N-alkylés/isolement et purification , Amides gras polyinsaturés N-alkylés/métabolisme , Canaux potassiques/histoire , Canaux potassiques/physiologie , Recherche/histoire , États-Unis
19.
Adv Pharmacol ; 72: 147-64, 2015.
Article de Anglais | MEDLINE | ID: mdl-25600369

RÉSUMÉ

Since their introduction in the 1960s, benzodiazepines (BZs) remain one of the most commonly prescribed medications, acting as potent sedatives, hypnotics, anxiolytics, anticonvulsants, and muscle relaxants. The primary neural action of BZs and related compounds is augmentation of inhibitory transmission, which occurs through allosteric modulation of the gamma-aminobutyric acid (GABA)-induced current at the gamma-aminobutyric acid receptor (GABAAR). The discovery of the BZ-binding site on GABAARs encouraged many to speculate that the brain produces its own endogenous ligands to this site (Costa & Guidotti, 1985). The romanticized quest for endozepines, endogenous ligands to the BZ-binding site, has uncovered a variety of ligands that might fulfill this role, including oleamides (Cravatt et al., 1995), nonpeptidic endozepines (Rothstein et al., 1992), and the protein diazepam-binding inhibitor (DBI) (Costa & Guidotti, 1985). Of these ligands, DBI, and affiliated peptide fragments, is the most extensively studied endozepine. The quest for the "brain's Valium" over the decades has been elusive as mainly negative allosteric modulatory effects have been observed (Alfonso, Le Magueresse, Zuccotti, Khodosevich, & Monyer, 2012; Costa & Guidotti, 1985), but recent evidence is accumulating that DBI displays regionally discrete endogenous positive modulation of GABA transmission through activation of the BZ receptor (Christian et al., 2013). Herein, we review the literature on this topic, focusing on identification of the endogenous molecule and its region-specific expression and function.


Sujet(s)
Inhibiteur de la liaison au diazépam/métabolisme , Récepteurs GABA-A/métabolisme , Acide gamma-amino-butyrique/métabolisme , Régulation allostérique , Animaux , Benzodiazépines/pharmacologie , Sites de fixation , Encéphale/métabolisme , Humains , Ligands , Récepteurs GABA-A/effets des médicaments et des substances chimiques
20.
Pharm Biol ; 53(3): 345-9, 2015 Mar.
Article de Anglais | MEDLINE | ID: mdl-25331826

RÉSUMÉ

CONTEXT: P-selectin is a promising target for inflammatory-related diseases. Polysaccharides are the active ingredients of Sanguisorba officinalis L. (Rosaceae) responsible for its anti-inflammatory activities; however, the molecular mechanism is not clear yet. OBJECTIVE: This study evaluates the effects of polysaccharides (SOPs) from Sanguisorba officinalis on their antagonistic function against P-selectin-mediated leukocyte adhesion. MATERIALS AND METHODS: The antagonistic function of SOPs was investigated by flow cytometry and static adhesion assay at the concentrations of 25 and 100 µg/ml. The dynamic interaction between HL-60 cells and CHO-P cell monolayer treated with SOPs (25 and 100 µg/ml) was analyzed in a parallel plate flow chamber, and quantitatively calculated by ImageJ software (NIH, Bethesda, MD). In vitro protein binding assay was carried out to evaluate the blocking effects of SOPs (25 and 100 µg/ml) on the interaction between P-selectin and PSGL-1. RESULTS: SOPs-treatment (100 µg/ml) significantly reduced the percentage of HL-60 cells binding to P-selectin (p < 0.01) determined by flow cytometry. In addition, SOPs (25 and 100 µg/ml) markedly blocked the adhesion between HL-60 cells and CHO-P cells under static condition, and the inhibitory rates reached 39.9% and 71.2%, respectively. Compared with the positive control group, SOPs-treatment (25 and 100 µg/ml) significantly reduced the percentage of HL-60 cells rolling on CHO-P cell monolayers by 43.5% and 75.2%, respectively. Protein binding assay showed the interaction between P-selectin and PSGL-1 was significantly blocked by SOPs. DISCUSSION AND CONCLUSION: SOPs possess a significant antagonistic function against P-selectin-mediated leukocyte adhesion, and SOPs could be considered as a promising candidate for amelioration of inflammation-related diseases.


Sujet(s)
Leucocytes/effets des médicaments et des substances chimiques , Sélectine P/antagonistes et inhibiteurs , Polyosides bactériens/isolement et purification , Polyosides bactériens/pharmacologie , Sanguisorba , Animaux , Cellules CHO , Adhérence cellulaire/effets des médicaments et des substances chimiques , Adhérence cellulaire/physiologie , Cricetinae , Cricetulus , Cellules HL-60 , Humains , Leucocytes/métabolisme , Sélectine P/métabolisme , Polyosides bactériens/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE