Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.771
Filter
1.
Biosci Rep ; 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39361899

ABSTRACT

Bimolecular fluorescence complementation (BiFC) methodology uses split fluorescent proteins to detect interactions between proteins in living cells. To date, BiFC has been used to investigate receptor dimerization by splitting the fluorescent protein between the intracellular portions of different receptor components. We reasoned that attaching these split proteins to the extracellular N-terminus instead may improve the flexibility of this methodology and reduce the likelihood of impaired intracellular signal transduction. As a proof-of-concept we used receptors for calcitonin gene-related peptide, which comprise heterodimers of either the calcitonin or calcitonin receptor-like receptor in complex with an accessory protein (receptor activity-modifying protein 1). We created fusion constructs in which split mVenus fragments were attached to either the C-termini or N-termini of receptor subunits. The resulting constructs were transfected into Cos7 and HEK293S cells, where we measured cAMP production in response to ligand stimulation, cell surface expression of receptor complexes, and BiFC fluorescence. Additionally, we investigated ligand-dependent internalization in HEK293S cells. We found N-terminal fusions were better tolerated with regards to cAMP signaling and receptor internalization. N-terminal fusions also allowed reconstitution of functional fluorescent mVenus proteins, however fluorescence yields were lower than with C-terminal fusion. Our results suggest that BiFC methodologies can be applied to the receptor N-terminus, thereby increasing the flexibility of this approach, and enabling further insights into receptor dimerization.

2.
Biomed Pharmacother ; 180: 117467, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39362066

ABSTRACT

Commensal microbiota is crucial for nutrient digestion and production of biologically active molecules, many of which mimic endogenous ligands of human GPCRs. Bacteroides spp. are among the most abundant bacteria residing in the human gut and their absence has been positively correlated with metabolic disorders. In the present study, we focused on N-acylated glycines (NAGlys) as products of Bacteroides spp. and potential GPCR ligands modulating GLP-1 secretion. Representative strains of the most abundant commensal Bacteroides were cultured in either yeast- or animal-based nutrient broths. The broths post-culture were investigated in terms of the contents of NAGlys and stimulatory effects towards GLP-1 production in GLUTag and NCI-H716 cell lines. Pure preparations of the detected NAGlys were further studied to evaluate stimulation of GLP-1 production and related cellular signalling evoked. The most potent NAGlys were also tested as ligands of key lipid GPCRs involved in the regulation of carbohydrate metabolism: GPR40/FFAR1, GPR55, GPR119, and GPR120/FFAR4. We found that Bacteroides potentiate GLP-1 production, depending on the strain and provided nutrient mix. Long-chain unsaturated oleoyl and arachidonoyl glycines, produced by B. thetaiotaomicron and B. intestinalis in the animal-based broth, were particularly effective in stimulation of GLP-1 secretion. They served as agonists of all the receptors under study expressed in GLP-1-producing cells. The obtained results broaden the knowledge of microbial signalling molecules and their role in regulation of carbohydrate homeostasis. They also emphasise the importance of balanced diet as a source of building blocks for commensal bacteria to produce efficient agonists of lipid GPCRs.

3.
Cell Biochem Biophys ; 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39365517

ABSTRACT

Olfactory receptors (ORs) are a class of G protein-coupled receptors (GPCR) widely distributed in olfactory sensory neurons and various non-olfactory tissues, serving significant physiological and pathological functions in the human body. Increasing evidence reveals the heightened expression of olfactory receptors in tumorous tissues and cells alongside normal tissues. Olfactory receptors have demonstrated influence over tumor cell proliferation and metastasis, establishing a close relationship with tumor initiation and progression. This review highlights the specific molecular actions and signaling pathways of olfactory receptors in the development of human tumors. The potential for precise tumor diagnosis and targeted therapy through therapeutic targeting of olfactory receptors as an adjunct anticancer treatment strategy is being considered.

4.
Front Endocrinol (Lausanne) ; 15: 1392418, 2024.
Article in English | MEDLINE | ID: mdl-39363899

ABSTRACT

Bone is a dynamic tissue that is constantly remodeled throughout adult life. Recently, it has been shown that bone turnover decreases shortly after food consumption. This process has been linked to the fermentation of non-digestible food ingredients such as inulin by gut microbes, which results in the production of the short-chain fatty acids (SCFAs) acetate, propionate and butyrate. SCFAs exert various metabolic functions, which in part can be explained by activation of G protein-coupled receptors (Gpr) 41 and 43. However, the potential relevance of a SCFA-Gpr41/43 signaling axis for bone metabolism has not been established. The aim of our study is to investigate the role of Gpr41/43 in bone metabolism and osteogenic differentiation of mesenchymal stem cells. For this purpose, we analyzed the skeletal phenotype of wild type controls (WT) and Gpr41/43 double knockout (Gpr41/43 dKO) mice fed either a chow or an inulin-enriched diet. In addition, we isolated bone marrow derived mesenchymal stem cells from WT and Gpr41/43 dKO mice and differentiated them into osteoblasts in the absence or presence of acetate. MicroCT scanning of femoral bones of Gpr41/43 dKO mice revealed a significant increase of trabecular bone volume and trabecular compared to WT controls. Treatment of WT bone marrow-derived osteoblasts with acetate resulted in decreased mineralization and substantial downregulation of bone formation markers such as Phex, Ptgs2 and Col1a1. Notably, this effect was strongly attenuated in differentiated osteoblasts lacking Gpr41/43. Inversely, acetate supplementation resulted in higher levels of adipocyte marker genes including Pparg, Lpl and Adipoq in bone marrow-derived cells from WT mice, an effect blunted in differentiated cells isolated from Gpr41/43 dKO mice. Overall, these data indicate that acetate regulates bone architecture via SCFA-Gpr41/43 signaling by modulating the osteogenic versus adipogenic differentiation of mesenchymal stem cells.


Subject(s)
Adipogenesis , Cell Differentiation , Mesenchymal Stem Cells , Mice, Knockout , Osteogenesis , Receptors, G-Protein-Coupled , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Adipogenesis/physiology , Osteogenesis/physiology , Fatty Acids, Volatile/metabolism , Mice, Inbred C57BL , Bone Density , Male , Osteoblasts/metabolism , Osteoblasts/cytology , Cells, Cultured
5.
Sci Rep ; 14(1): 23595, 2024 10 09.
Article in English | MEDLINE | ID: mdl-39384825

ABSTRACT

Formyl peptide receptors (FPR), part of the G-protein coupled receptor superfamily, are pivotal in directing phagocyte migration towards chemotactic signals from bacteria and host tissues. Although their roles in acute bacterial infections are well-documented, their involvement in immunity against tuberculosis (TB) remains unexplored. Here, we investigate the functions of Fpr1 and Fpr2 in defense against Mycobacterium tuberculosis (Mtb), the causative agent of TB. Elevated levels of Fpr1 and Fpr2 were found in the lungs of mice, rabbits and peripheral blood of humans infected with Mtb, suggesting a crucial role in the immune response. The effects of Fpr1 and Fpr2 deletion on bacterial load, lung damage, and cellular inflammation were assessed in a murine TB model utilizing hypervirulent strain of Mtb from the W-Beijing lineage. While Fpr2 deletion had no impact on disease outcome, Fpr1-deficient mice demonstrated improved bacterial control, especially by macrophages. Bone marrow-derived macrophages from these Fpr1-/- mice exhibited an enhanced ability to contain bacterial growth over time. Contrarily, treating genetically susceptible mice with Fpr1-specific inhibitors caused impaired early bacterial control, corresponding with increased Mtb persistence in necrotic neutrophils. Furthermore, ex vivo assays revealed that Fpr1-/- neutrophils were unable to restrain Mtb growth, indicating a differential function of Fpr1 among myeloid cells. These findings highlight the distinct and complex roles of Fpr1 in myeloid cell-mediated immunity against Mtb infection, underscoring the need for further research into these mechanisms for a better understanding of TB immunity.


Subject(s)
Macrophages , Mycobacterium tuberculosis , Neutrophils , Receptors, Formyl Peptide , Tuberculosis , Receptors, Formyl Peptide/metabolism , Receptors, Formyl Peptide/genetics , Animals , Neutrophils/immunology , Neutrophils/metabolism , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Mice , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Humans , Tuberculosis/immunology , Tuberculosis/microbiology , Mice, Knockout , Rabbits , Mice, Inbred C57BL , Lung/microbiology , Lung/immunology , Lung/pathology , Lung/metabolism , Disease Models, Animal , Female
6.
Pharmacol Res ; 208: 107395, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39241934

ABSTRACT

G protein-coupled receptors (GPCRs), widely expressed in the human central nervous system (CNS), perform numerous physiological functions and play a significant role in the pathogenesis of diseases. Consequently, identifying key therapeutic GPCRs targets for CNS-related diseases is garnering immense interest in research labs and pharmaceutical companies. However, using GPCRs drugs for treating neurodegenerative diseases has limitations, including side effects and uncertain effective time frame. Recognizing the rich history of herbal treatments for neurological disorders like stroke, Alzheimer's disease (AD), and Parkinson's disease (PD), modern pharmacological research is now focusing on the understanding of the efficacy of traditional Chinese medicinal herbs and compounds in modulating GPCRs and treatment of neurodegenerative conditions. This paper will offer a comprehensive, critical review of how certain natural products and compounds target GPCRs to treat neurological diseases. Conducting an in-depth study of herbal remedies and their efficacies against CNS-related disorders through GPCRs targeting will augment our strategies for treating neurological disorders. This will not only broaden our understanding of effective therapeutic methodologies but also identify the root causes of altered GPCRs signaling in the context of pathophysiological mechanisms in neurological diseases. Moreover, it would be informative for the creation of safer and more effective GPCR-mediated drugs, thereby establishing a foundation for future treatment of various neurological diseases.


Subject(s)
Biological Products , Nervous System Diseases , Receptors, G-Protein-Coupled , Humans , Receptors, G-Protein-Coupled/metabolism , Animals , Biological Products/therapeutic use , Biological Products/pharmacology , Nervous System Diseases/drug therapy , Nervous System Diseases/metabolism , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/pharmacology , Signal Transduction/drug effects
7.
Br J Pharmacol ; 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261899

ABSTRACT

Class A G protein-coupled receptors (GPCRs) continue to garner interest for their essential roles in cell signalling and their importance as drug targets. Although numerous drugs in the clinic target these receptors, over 60% GPCRs remain unexploited. Moreover, the adverse effects triggered by the available unbiased GPCR modulators, limit their use and therapeutic value. In this context, the elucidation of biased signalling has opened up new pharmacological avenues holding promise for safer therapeutics. Functionally selective ligands favour receptor conformations facilitating the recruitment of specific effectors and the modulation of the associated pathways. This review surveys the current drug discovery landscape of GPCR-biased modulators with a focus on recent advances. Understanding the biological effects of this preferential coupling is at different stages depending on the Class A GPCR family. Therefore, with a focus on individual GPCR families, we present a compilation of the functionally selective modulators reported over the past few years. In doing so, we dissect their therapeutic relevance, molecular determinants and potential clinical applications.

8.
Cell Commun Signal ; 22(1): 423, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223553

ABSTRACT

Acute kidney injury (AKI) is a clinical condition characterized by a rapid decline in kidney function, which is associated with local inflammation and programmed cell death in the kidney. The G protein-coupled receptors (GPCRs) represent the largest family of signaling transduction proteins in the body, and approximately 40% of drugs on the market target GPCRs. The expressions of various GPCRs, prostaglandin receptors and purinergic receptors, to name a few, are significantly altered in AKI models. And the role of GPCRs in AKI is catching the eyes of researchers due to their distinctive biological functions, such as regulation of hemodynamics, metabolic reprogramming, and inflammation. Therefore, in this review, we aim to discuss the role of GPCRs in the pathogenesis of AKI and summarize the relevant clinical trials involving GPCRs to assess the potential of GPCRs and their ligands as therapeutic targets in AKI and the transition to AKI-CKD.


Subject(s)
Acute Kidney Injury , Receptors, G-Protein-Coupled , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Humans , Receptors, G-Protein-Coupled/metabolism , Animals , Signal Transduction
9.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273318

ABSTRACT

The paradigm "one drug fits all" or "one dose fits all" will soon be challenged by pharmacogenetics research and application. Drug response-efficacy or safety-depends on interindividual variability. The current clinical practice does not include genetic screening as a routine procedure and does not account for genetic variation. Patients with the same illness receive the same treatment, yielding different responses. Integrating pharmacogenomics in therapy would provide critical information about how a patient will respond to a certain drug. Worldwide, great efforts are being made to achieve a personalized therapy-based approach. Nevertheless, a global harmonized guideline is still needed. Plasma membrane proteins, like receptor tyrosine kinase (RTK) and G protein-coupled receptors (GPCRs), are ubiquitously expressed, being involved in a diverse array of physiopathological processes. Over 30% of drugs approved by the FDA target GPCRs, reflecting the importance of assessing the genetic variability among individuals who are treated with these drugs. Pharmacogenomics of transmembrane protein receptors is a dynamic field with profound implications for precision medicine. Understanding genetic variations in these receptors provides a framework for optimizing drug therapies, minimizing adverse reactions, and advancing the paradigm of personalized healthcare.


Subject(s)
Pharmacogenetics , Precision Medicine , Receptors, G-Protein-Coupled , Humans , Pharmacogenetics/methods , Precision Medicine/methods , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Genetic Variation
10.
Mol Pharmacol ; 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39284672

ABSTRACT

Aberrant type 2 inflammatory responses are the underlying cause of the pathophysiology of allergic asthma, allergic rhinitis and other atopic diseases with an alarming prevalence in relevant parts of the western world. A bulk of evidence points out the important role of the DP2 receptor in this inflammation processes. A screening of different polyunsaturated fatty acids (PUFAs) at a fluorescence resonance energy transfer (FRET)-based DP2 receptor conformation sensor expressed in HEK cells revealed an agonistic effect of the prostaglandin (PG) D2 precursor arachidonic acid (AA) on DP2 receptor activity of about 80% of the effect induced by PGD2 In a combination of experiments at the conformation sensor and using a BRET-based G protein activation sensor expressed together with DP2 receptor-wt in HEK cells, we found that arachidonic acid act as a direct activator of the DP2 receptor but not DP1 receptor, in a concentration range considered physiologically relevant. Pharmacological inhibition of cyclooxygenases and lipoxygenases as well as cytochrome P450 did not lead to a diminished arachidonic acid response on the DP2 receptor, confirming a direct action of arachidonic acid on the receptor. Significance Statement We identified the prostaglandin precursor arachidonic acid to directly activate the DP2 receptor, a G protein-coupled receptor that is known to play an important role in type 2 inflammation.

11.
Int J Mol Sci ; 25(18)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39337509

ABSTRACT

Inflammatory bowel diseases (IBDs) involve chronic inflammation of the gastrointestinal tract, where effector CD4+ T-cells play a central role. Thereby, the recruitment of T-cells into the colonic mucosa represents a key process in IBD. We recently found that CCR9 and DRD5 might form a heteromeric complex on the T-cell surface. The increase in CCL25 production and the reduction in dopamine levels associated with colonic inflammation represent a dual signal stimulating the CCR9:DRD5 heteromer, which promotes the recruitment of CD4+ T-cells into the colonic lamina propria. Here, we aimed to analyse the molecular requirements involved in the heteromer assembly as well as to determine the underlying cellular mechanisms involved in the colonic tropism given by the stimulation of the CCR9:DRD5 complex. The results show that dual stimulation of the CCR9:DRD5 heteromer potentiates the phosphorylation of the myosin light chain 2 (MLC2) and the migration speed in confined microchannels. Accordingly, disrupting the CCR9:DRD5 assembly induced a sharp reduction in the pMLC2 in vitro, decreased the migratory speed in confined microchannels, and dampened the recruitment of CD4+ T-cells into the inflamed colonic mucosa. Furthermore, in silico analysis confirmed that the interface of interaction of CCR9:DRD5 is formed by the transmembrane segments 5 and 6 from each protomer. Our findings demonstrated that the CCR9:DRD5 heteromeric complex plays a fundamental role in the migration of CD4+ T-cells into the colonic mucosa upon inflammation. Thereby, the present study encourages the design of strategies for disassembling the formation of the CCR9:DRD5 as a therapeutic opportunity to treat IBD.


Subject(s)
CD4-Positive T-Lymphocytes , Intestinal Mucosa , Receptors, CCR , Receptors, Dopamine D5 , Signal Transduction , Receptors, CCR/metabolism , Receptors, CCR/genetics , Humans , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Receptors, Dopamine D5/metabolism , Receptors, Dopamine D5/genetics , Intestinal Mucosa/metabolism , Colon/metabolism , Cell Movement , Dopamine/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/immunology
12.
bioRxiv ; 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39314495

ABSTRACT

Adhesion G protein-coupled receptors (ADGRs) belong to Class B2 of GPCRs and are involved in a wide array of important physiological processes. ADGRs contain a GPCR autoproteolysis-inducing (GAIN) domain that is proximal to the receptor N-terminus and undergoes autoproteolysis during biosynthesis to generate two fragments: the N-terminal fragment (NTF) and C-terminal fragment (CTF). Dissociation of NTF reveals a tethered agonist to activate CTF of ADGRs for G protein signaling. Synthetic peptides that mimic the tethered agonist can also activate the ADGRs. However, mechanisms of peptide agonist dissociation and deactivation of ADGRs remain poorly understood. In this study, we have performed all-atom enhanced sampling simulations using a novel Protein-Protein Interaction-Gaussian accelerated Molecular Dynamics (PPI-GaMD) method on the ADGRG2-IP15 and ADGRG1-P7 complexes. The PPI-GaMD simulations captured dissociation of the IP15 and P7 peptide agonists from their target receptors. We were able to identify important low-energy conformations of ADGRG2 and ADGRG1 in the active, intermediate, and inactive states, as well as exploring different states of the peptide agonists IP15 and P7 during dissociation. Therefore, our PPI-GaMD simulations have revealed dynamic mechanisms of peptide agonist dissociation and deactivation of ADGRG1 and ADGRG2, which will facilitate rational design of peptide regulators of the two receptors and other ADGRs.

14.
Function (Oxf) ; 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39322240

ABSTRACT

Essential hypertension (HT) is a highly prevalent cardiovascular disease of unclear physiopathology. Pharmacological studies suggest that purinergic P2Y6 receptors (P2ry6) play important roles in cardiovascular function and may contribute to angiotensin II (AgtII) pathophysiological effects. Here, we tested the hypothesis that functional coupling between P2ry6 and AgtII receptors mediates altered vascular reactivity in HT. For this, a multipronged approach was implemented using mesenteric vascular smooth muscle cells (VSMCs) and arteries from BPN (Blood Pressure Normal) and BPH (Blood Pressure High) mice. Differential transcriptome profiling of mesenteric artery VSMCs identified P2ry6 purinergic receptor mRNA as one of the top upregulated transcripts in BPH. P2Y receptor activation elicited distinct vascular responses in mesenteric arteries from BPN and BPH mice. Accordingly, 10 µM UTP produced a contraction close to half-maximal activation in BPH arteries but no response in BPN vessels. AgtII-induced contraction was also higher in BPH mice despite having lower AgtII receptor type-1 (Agtr1) expression and was sensitive to P2ry6 modulators. Proximity Ligation Assay (PLA) and super-resolution microscopy (SRM) showed closer localization of Agtr1 and P2ry6 at/near the membrane of BPH mice. This proximal association was reduced in BPN mice, suggesting a functional role for Agtr1-P2ry6 complexes in the hypertensive phenotype. Intriguingly, BPN mice were resistant to AgtII-induced HT and showed reduced P2ry6 expression in VSMCs. Altogether, results suggest that increased functional coupling between P2ry6 and Agtr1 may contribute to enhanced vascular reactivity during HT. In this regard, blocking P2ry6 could be a potential pharmacological strategy to treat HT.

15.
Microorganisms ; 12(8)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39203464

ABSTRACT

This study explores whether Escherichia coli Nissle 1917 (EcN) can preserve the integrity of the intestinal barrier by modulating the metabolism pathway of short-chain fatty acids (SCFAs) in a C57BL/6J mouse model of lipopolysaccharide (LPS)-induced acute enteritis and a model of a Caco-2 monolayer. The study involved establishing a septic shock model in mice through lipopolysaccharide (LPS) injection. Clinical scores and intestinal permeability were meticulously documented. Immunofluorescence was utilized to localize the tight junction proteins. A quantitative real-time polymerase chain reaction (qRT-PCR) was employed to assess the expression of G protein-coupled receptors (GPRs) signaling. Additionally, the supplement of acetate and butyrate with Caco-2 monolayers to elucidate the potential of EcN in augmenting the intestinal barrier primarily via the modulation of SCFAs and qRT-PCR was performed to detect the expression of tight junction proteins and the activation of the GPRs protein signaling pathway. EcN mitigated the clinical symptoms and reduced intestinal permeability in the colon of LPS-induced mice. It also enhanced the production of SCFAs in the gut and upregulated the expression of SCFA receptor proteins GPR41 and GPR43 in the colon tissue. Our findings reveal that EcN activates the SCFA/GPRs pathway, thereby preserving intestinal barrier function and alleviating inflammation in a mouse sepsis model.

16.
Mol Pharmacol ; 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39187389

ABSTRACT

The orexigenic gut peptide ghrelin is an endogenous ligand for the growth hormone secretagogue receptor type 1a (GHSR1a). Systemic ghrelin administration has previously been shown to increase gastric motility and emptying. While these effects are known to be mediated by the vagus nerve, the cellular mechanism underlying these effects remains unclear. Therefore, the purpose of the present study was to investigate the signaling mechanism by which GHSR1a inhibits voltage-gated Ca2+ channels in isolated rat gastric vagal afferent neurons using whole-cell patch-clamp electrophysiology. The ghrelin pharmacological profile indicated that Ca2+ currents were inhibited with a log (Ic50)=-2.10 {plus minus} 0.44 and a maximal inhibition of 42.8 {plus minus} 5.0%. Exposure to the GHSR1a receptor antagonist (D-Lys3)-GHRP-6 reduced ghrelin-mediated Ca2+ channel inhibition (29.4 {plus minus} 16.7% vs 1.9 {plus minus} 2.5%, n=6, p=0.0064). Interestingly, we observed that activation of GHSR1a inhibited Ca2+ currents through both voltage-dependent and voltage-independent pathways. We also treated the gastric neurons with either pertussis toxin (PTX) or YM-254890 to examine whether the Ca2+ current inhibition was mediated by Gαi/o or Gαq/11 family of subunits. Treatment with both PTX (Ca2+ current inhibition=15.7 {plus minus} 10.6%, n=8, p=0.0327) and YM-254890 (15.2 {plus minus} 11.9%, n=8, p=0.0269) blocked ghrelin's effects on Ca2+ currents, as compared to control neurons (34.3 {plus minus} 18.9%, n=8). These results indicate GHSR1a can couple to both Gαi/o and Gαq/11 in gastric vagal afferent neurons. Overall, our findings suggest GHSR1a-mediated inhibition of Ca2+ currents occurs through two distinct pathways, offering necessary insights into the cellular mechanisms underlying ghrelin's regulation of gastric vagal afferents. Significance Statement This study demonstrated that in gastric vagal afferent neurons, activation of GHSR1a by ghrelin inhibits voltage-gated Ca2+ channels through both voltage-dependent and voltage-independent signaling pathways. These results provide necessary insight into the cellular mechanism underlying ghrelin regulation of gastric vagal afferent activity, which may benefit future studies investigating ghrelin mimetics to treat gastric motility disorders.

17.
Pharmacol Rev ; 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214702

ABSTRACT

Bitter perception plays a critical role for the detection of potentially harmful substances in food items for most vertebrates. The detection of bitter compounds is facilitated by specialized receptors located in taste buds of the oral cavity. This work focuses on the receptors, including their sensitivities, structure-function relationships, agonists and antagonists. Moreover, the existence of numerous bitter taste receptor variants in the human population and the fact that several of them affect individual bitter tasting profoundly, is discussed as well. The identification of bitter taste receptors in numerous tissues outside the oral cavity and their multiple proposed roles in these tissues is also described briefly. Although this work is mainly focused on human bitter taste receptors, it is imperative to compare human bitter taste with that of other animals to understand which evolutionary forces might have shaped bitter taste receptors and their functions and to distinguish apparent typical human from rather general features. For the readers who are not too familiar with the gustatory system short descriptions of taste anatomy, signal transduction and oral bitter taste receptor expression are included in the beginning of this article. Significance Statement Apart from their role as sensors for potentially harmful substances in the oral cavity, the numerous additional roles of bitter taste receptors in tissues outside the gustatory system have received much attention recently. For the careful assessment of functions inside and outside the taste system a solid knowledge about the specific and general pharmacological features of these receptors and the growing toolbox available for studying them is imperative and provided in this work.

18.
Cell ; 187(17): 4751-4769.e25, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39089252

ABSTRACT

The Duffy antigen receptor is a seven-transmembrane (7TM) protein expressed primarily at the surface of red blood cells and displays strikingly promiscuous binding to multiple inflammatory and homeostatic chemokines. It serves as the basis of the Duffy blood group system in humans and also acts as the primary attachment site for malarial parasite Plasmodium vivax and pore-forming toxins secreted by Staphylococcus aureus. Here, we comprehensively profile transducer coupling of this receptor, discover potential non-canonical signaling pathways, and determine the cryoelectron microscopy (cryo-EM) structure in complex with the chemokine CCL7. The structure reveals a distinct binding mode of chemokines, as reflected by relatively superficial binding and a partially formed orthosteric binding pocket. We also observe a dramatic shortening of TM5 and 6 on the intracellular side, which precludes the formation of the docking site for canonical signal transducers, thereby providing a possible explanation for the distinct pharmacological and functional phenotype of this receptor.


Subject(s)
Cryoelectron Microscopy , Duffy Blood-Group System , Receptors, Cell Surface , Humans , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/chemistry , Duffy Blood-Group System/metabolism , Duffy Blood-Group System/chemistry , Signal Transduction , Binding Sites , Chemokines/metabolism , Chemokines/chemistry , Protein Binding
19.
Asian Pac J Cancer Prev ; 25(8): 2605-2613, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39205557

ABSTRACT

BACKGROUND: Sphingosine kinase 1 (SphK1) is a lipid enzyme whose role in the etiology of cancer has been well explored. Here, a systematic review and meta-analysis were conducted to evaluate the association of SphK1 expression with hematological malignancy. MATERIALS AND METHODS: Relevant studies were identified through electronic databases (PubMed, Scopus, Embase, and OVID) and evaluated based on predefined inclusion and exclusion criteria. Quality assessment using the Newcastle-Ottawa Scale (NOS) was conducted, and pooled odds ratio (OR) was calculated to assess the association between SphK1 expression and hematological malignancy. RESULTS: Nine studies meeting the inclusion criteria were included in the systematic review. These studies utilized various techniques to assess SphK1 expression in hematological malignancies. The quality assessment reported that the included studies were of moderate quality. Meta-analysis of eligible studies revealed a positive association between SphK1 expression and hematological malignancies at the protein level (OR = 52.37, 95% CI = 10.10 to 271.47, and P = 0.00001). The funnel plot indicated no publication bias among the included studies. However, the certainty of the evidence was low according to the GRADE assessment. CONCLUSION: Our study's findings support the link between SphK1 expression and hematological malignancies. SphK1 gene dysregulation may contribute to various malignancies, suggesting it could be a therapeutic target to improve patient outcomes. Further research is needed to understand SphK1's mechanistic role in hematological malignancies and its therapeutic potential.


Subject(s)
Hematologic Neoplasms , Phosphotransferases (Alcohol Group Acceptor) , Humans , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Hematologic Neoplasms/epidemiology , Hematologic Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/analysis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Prognosis
20.
Neuron ; 112(19): 3295-3310.e8, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39094559

ABSTRACT

Understanding the kinetics of LSD in receptors and subsequent induced signaling is crucial for comprehending both the psychoactive and therapeutic effects of LSD. Despite extensive research on LSD's interactions with serotonin 2A and 2B receptors, its behavior on other targets, including dopamine receptors, has remained elusive. Here, we present cryo-EM structures of LSD/PF6142-bound dopamine D1 receptor (DRD1)-legobody complexes, accompanied by a ß-arrestin-mimicking nanobody, NBA3, shedding light on the determinants of G protein coupling versus ß-arrestin coupling. Structural analysis unveils a distinctive binding mode of LSD in DRD1, particularly with the ergoline moiety oriented toward TM4. Kinetic investigations uncover an exceptionally rapid dissociation rate of LSD in DRD1, attributed to the flexibility of extracellular loop 2 (ECL2). Moreover, G protein can stabilize ECL2 conformation, leading to a significant slowdown in ligand's dissociation rate. These findings establish a solid foundation for further exploration of G protein-coupled receptor (GPCR) dynamics and their relevance to signal transduction.


Subject(s)
Cryoelectron Microscopy , Lysergic Acid Diethylamide , Receptors, Dopamine D1 , Receptors, Dopamine D1/metabolism , Lysergic Acid Diethylamide/pharmacology , Humans , Hallucinogens/pharmacology , Hallucinogens/chemistry , HEK293 Cells , beta-Arrestins/metabolism , Animals , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL