Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (811)

Search Parameters:
Keywords = G protein-coupled receptor (GPCR)

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
23 pages, 5203 KB  
Article
On–DNA Platform Molecules Based on a Diazide Scaffold II: A Compact Diazide Platform Designed for Small–Molecule Drug Discovery
by Hiroyuki Miyachi, Masaki Koshimizu and Masashi Suzuki
Int. J. Mol. Sci. 2026, 27(2), 828; https://doi.org/10.3390/ijms27020828 - 14 Jan 2026
Viewed by 117
Abstract
Expanding the chemical diversity of DNA–encoded libraries (DELs) is crucial for identifying binders to emerging drug targets using DEL technology. In the present study, as part of our ongoing efforts to develop on–DNA diazide platforms (D–DAPs)—platform molecules possessing both aromatic and aliphatic azide [...] Read more.
Expanding the chemical diversity of DNA–encoded libraries (DELs) is crucial for identifying binders to emerging drug targets using DEL technology. In the present study, as part of our ongoing efforts to develop on–DNA diazide platforms (D–DAPs)—platform molecules possessing both aromatic and aliphatic azide groups on a single core reactive scaffold—we designed and synthesized a new compact diazide platform, designated as a compact D–DAP (C–D–DAP). This molecule is based on a low–molecular–weight reactive scaffold, 3–azido–5–(azidomethyl)benzoic acid, to facilitate small–molecule drug discovery targeting enzymes and G protein–coupled receptors (GPCRs). Furthermore, we established two distinct stepwise warhead construction strategies that exploit the chemoselective transformations of the azide groups in the C–D–DAP, which exhibit different reactivities. In addition, four virtual DELs were generated based on stepwise warhead elaboration from the C–D–DAP scaffold. Comparative chemical diversity analysis against bioactive compounds from ChEMBL revealed that these virtual libraries populate structural regions that are sparsely represented among known molecules. Each virtual library also occupies a distinct region of structural space relative to the others and displays intermediate quantitative estimate of drug–likeness (QED) values. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

13 pages, 733 KB  
Review
G Protein-Coupled Receptors in Irritable Bowel Syndrome: Mechanisms and Therapeutic Opportunities
by Zhenya Zhu, Ziyu Liu, Yate He, Xiaorui He, Wei Zheng and Mizu Jiang
Int. J. Mol. Sci. 2026, 27(2), 752; https://doi.org/10.3390/ijms27020752 - 12 Jan 2026
Viewed by 260
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder characterized by abdominal pain, altered motility, and visceral hypersensitivity. Emerging evidence implicates G protein-coupled receptors (GPCRs) as key integrators of microbial, immune, endocrine, and neural signals in IBS pathophysiology. This review summarizes recent advances [...] Read more.
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder characterized by abdominal pain, altered motility, and visceral hypersensitivity. Emerging evidence implicates G protein-coupled receptors (GPCRs) as key integrators of microbial, immune, endocrine, and neural signals in IBS pathophysiology. This review summarizes recent advances in understanding how GPCRs mediate gut immune regulation, microbiota–host crosstalk, metabolic signaling, and pain processing in IBS. Recent studies show that microbial metabolites (e.g., short-chain fatty acids, biogenic amines, and lipid mediators) signal through GPCRs on immune cells, epithelia, and neurons to influence intestinal homeostasis. On immune cells and neurons, GPCRs also mediate signals from external substances (such as fats, sugars, histamine, etc.) to regulate immune and neural functions. And there are challenges and future directions in targeting GPCRs for IBS, including patient heterogeneity and the complexity of host–microbiome interactions. This review provides a mechanistic framework for GPCR-based therapies in IBS. Full article
(This article belongs to the Special Issue Emerging Roles of the Gut-Brain Axis (GBA) in Health and Disease)
Show Figures

Figure 1

20 pages, 1108 KB  
Review
G Protein-Coupled Receptors in Cerebrovascular Diseases: Signaling Mechanisms and Therapeutic Opportunities
by Qiuxiang Gu, Jia Yao, Jiajing Sheng and Dong Liu
Int. J. Mol. Sci. 2026, 27(2), 736; https://doi.org/10.3390/ijms27020736 - 11 Jan 2026
Viewed by 319
Abstract
G protein-coupled receptors (GPCRs) are key regulators of cerebrovascular function, integrating vascular, inflammatory, and neuronal signaling within the neurovascular unit (NVU). Increasing evidence suggests that GPCR actions are highly dependent on cell type, signaling pathway, and disease stage, leading to distinct, and sometimes [...] Read more.
G protein-coupled receptors (GPCRs) are key regulators of cerebrovascular function, integrating vascular, inflammatory, and neuronal signaling within the neurovascular unit (NVU). Increasing evidence suggests that GPCR actions are highly dependent on cell type, signaling pathway, and disease stage, leading to distinct, and sometimes opposing, effects during acute ischemic injury and post-stroke recovery. In this review, we reorganize GPCR signaling mechanisms using a disease-stage-oriented and NVU-centered framework. We synthesize how GPCR-mediated intercellular communication among neurons, glial cells, and vascular elements dynamically regulates cerebral blood flow, neuroinflammation, blood–brain barrier (BBB) integrity, and neuronal circuit remodeling. Particular emphasis is placed on phase-dependent GPCR signaling, highlighting receptors whose functions shift across acute injury, secondary damage, and recovery phases. We further critically evaluated the translational implications of GPCR-targeted therapies, discussing why promising preclinical neuroprotection has frequently failed to translate into clinical benefit. By integrating molecular mechanisms with temporal dynamics and translational constraints, this review provides a framework for the rational development of cell-type and stage-specific GPCR-based therapeutic strategies in cerebrovascular disease. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

21 pages, 1549 KB  
Article
Identification and Classification of Fungal GPCR Gene Families
by Zhiyin Liu, Asaf Salamov and Igor V. Grigoriev
J. Fungi 2026, 12(1), 30; https://doi.org/10.3390/jof12010030 - 30 Dec 2025
Viewed by 437
Abstract
G protein-coupled receptors (GPCRs) are transmembrane proteins crucial for signal transduction in eukaryotes, responding to diverse extracellular signals. Researchers have found and systematically summarized 14 distinct types of GPCRs in fungi but their distribution among numerous fungal species remained largely unexamined. Additionally, three [...] Read more.
G protein-coupled receptors (GPCRs) are transmembrane proteins crucial for signal transduction in eukaryotes, responding to diverse extracellular signals. Researchers have found and systematically summarized 14 distinct types of GPCRs in fungi but their distribution among numerous fungal species remained largely unexamined. Additionally, three families of mammalian homologs (Rhodopsin, Glutamate, and Frizzled) have been found in previous studies, but they are not included in the systematic classification of fungal GPCRs. Our study establishes a unified classification of 17 GPCR classes in fungi, combining 14 fungal and 3 mammalian previously recognized groups, and classifies 28,294 GPCRs across 1357 fungal species, significantly expanding the scale of GPCRs in fungi and demonstrating their broader distribution. We found that mammalian homologs are notably more prevalent in Early Diverging Fungi (EDF), whereas the previous 14 classes are predominantly found in Ascomycota and Basidiomycota. The most abundant class detected in fungi was Pth11-like GPCRs, exclusively found in Pezizomycotina and involved in fungal pathogenicity. Our analysis suggested that Pezizomycotina ancestor possessed an extensive array of Pth11-like GPCRs, but over time, some species underwent considerable reductions in these GPCRs in conjunction with genome contractions. Utilizing a custom-built convolutional neural network (CNN) for the identification of fungal GPCRs, we identified several putative novel fungal GPCRs. Predicted interactions between these prospective new GPCRs and G-alpha proteins, as simulated by AlphaFold Multimer, provided additional support for their functional relevance. In conclusion, our work defines the first large-scale, unified classification of fungal GPCRs, reveals lineage-specific expansions and contractions, and uncovers previously unrecognized GPCR candidates with potential functional roles in fungal signaling. Full article
(This article belongs to the Section Fungal Genomics, Genetics and Molecular Biology)
Show Figures

Figure 1

54 pages, 6191 KB  
Review
Integration of cAMP and TRPV4 Signaling to Optimize Collagen Remodeling for Management of Fibrosis
by Connie Di Raimo and Christopher McCulloch
Cells 2026, 15(1), 56; https://doi.org/10.3390/cells15010056 - 28 Dec 2025
Viewed by 471
Abstract
Fibrosis manifests as an excessive accumulation of fibrillar collagen in tissues where secreted collagen exceeds degradation. Myofibroblasts are important contributors to the excessive collagen seen in fibrotic lesions. Accordingly, targeting signaling pathways that enhance collagen degradation and subdue myofibroblast differentiation has the potential [...] Read more.
Fibrosis manifests as an excessive accumulation of fibrillar collagen in tissues where secreted collagen exceeds degradation. Myofibroblasts are important contributors to the excessive collagen seen in fibrotic lesions. Accordingly, targeting signaling pathways that enhance collagen degradation and subdue myofibroblast differentiation has the potential to optimize collagen remodeling and improve organ fibrosis. One of the most promising molecular targets for therapeutic development is the G protein-coupled receptor (GPCR) family, which is diverse, cell-type-specific, multi-pass transmembrane receptors that participate in the regulation of extracellular matrix remodeling. GPCRs are categorized into multiple subclasses, some of which activate signaling cascades that can augment or reduce pro-fibrotic processes, depending on which Gα class is activated. Specifically, activation of Gαs GPCR stimulates production of the second messenger, cyclic adenosine monophosphate (cAMP), which generally inhibits pro-fibrotic mediators. A related, second approach for control of fibrosis is the blockade of a specific mechanosensitive, Ca2+-permeable channel that is implicated in fibrosis and contributes to myofibroblast differentiation, the transient receptor potential vanilloid type 4 (TRPV4). In health, TRPV4 activation regulates collagen remodeling, but when dysregulated, it promotes pro-fibrotic gene expression through mechanosensitive transcription factors. In this review, we focus on the functions of the Gαs GPCR pathway and TRPV4 activation through the interplay of the second messengers cAMP and Ca2+ ions. Ca2+ influx modulates cAMP levels by regulating phosphodiesterases and adenylyl cyclases. We consider evidence that Gαs GPCR and TRPV4 signaling pathways interact antagonistically to either promote collagen degradation or to increase the formation of myofibroblasts through signaling that involves cAMP and Ca2+ conductance. Coordinated activation of the Gαs GPCR pathway and inhibition of TRPV4 could provide a novel, bimodal approach to control tissue fibrosis. Full article
(This article belongs to the Special Issue Transient Receptor Potential (TRP) Channels and Health and Disease)
Show Figures

Figure 1

23 pages, 1065 KB  
Review
The Emerging Roles of Metabolite-Activated GPCRs in Teleost Physiology and Aquaculture Development
by Guan-Yuan Wei, Ming-Yuan Wu, Lan Ding, Zhen-Fa Qin, Zheng-Xiang Zhang, Liang-Jia Wei and Zhi-Shuai Hou
Metabolites 2026, 16(1), 29; https://doi.org/10.3390/metabo16010029 - 26 Dec 2025
Viewed by 340
Abstract
Metabolites, once viewed mainly as energy substrates or structural precursors, are now increasingly recognized as key extracellular signaling mediators that regulate diverse physiological processes. This review synthesizes and systematizes current knowledge on metabolite-mediated signaling through G-protein-coupled receptors (GPCRs) in teleosts and, importantly, highlights [...] Read more.
Metabolites, once viewed mainly as energy substrates or structural precursors, are now increasingly recognized as key extracellular signaling mediators that regulate diverse physiological processes. This review synthesizes and systematizes current knowledge on metabolite-mediated signaling through G-protein-coupled receptors (GPCRs) in teleosts and, importantly, highlights new conceptual links between specific metabolite–GPCR axes and key physiological functions relevant to aquaculture. By integrating evidence across metabolite–GPCRs axes, including succinate–SUCNR1, aromatic amino acids (tryptophan and phenylalanine)–GPR142, basic amino acids (L-arginine)–GPRC6A, and lactate–GPR81. We clarify how metabolite–receptor interactions have the potential to modulate glucose homeostasis, immune responses, energy metabolism, and stress coping. A major contribution of this review is illustrating how metabolites act not only as nutrients but also as extracellular signaling molecules governing core physiological processes via GPCRs. Particularly from an evolutionary perspective, compared with peptide-activated GPCRs, metabolite-sensing GPCRs are relatively conserved across different species, suggesting that relevant findings from biomedical research could be translated to aquaculture applications. Therefore, understanding GPCR-mediated metabolite sensing provides a molecular foundation for improving nutrient formulation, developing functional feeds, and designing selective breeding strategies in precision aquaculture. Full article
(This article belongs to the Special Issue Nutrition, Metabolism and Physiology in Aquatic Animals)
Show Figures

Figure 1

13 pages, 2934 KB  
Article
TAS1R3 Regulates GTPase Signaling in Human Skeletal Muscle Cells for Glucose Uptake
by Joseph M. Hoolachan, Rekha Balakrishnan, Karla E. Merz, Debbie C. Thurmond and Rajakrishnan Veluthakal
Int. J. Mol. Sci. 2026, 27(1), 103; https://doi.org/10.3390/ijms27010103 - 22 Dec 2025
Viewed by 575
Abstract
Taste receptor type 1 member 3 (TAS1R3) is a class C G protein-coupled receptor (GPCR) traditionally associated with taste perception. While its role in insulin secretion is established, its contribution to skeletal muscle glucose uptake, a process responsible for 70–80% of postprandial glucose [...] Read more.
Taste receptor type 1 member 3 (TAS1R3) is a class C G protein-coupled receptor (GPCR) traditionally associated with taste perception. While its role in insulin secretion is established, its contribution to skeletal muscle glucose uptake, a process responsible for 70–80% of postprandial glucose disposal, remains unclear. TAS1R3 expression was assessed in skeletal muscle biopsies from non-diabetic and type 2 diabetes (T2D) donors using qPCR and immunoblotting. Functional studies in human LHCN-M2 myotubes involved TAS1R3 inhibition with lactisole or siRNA-mediated knockdown, followed by the measurement of insulin-stimulated glucose uptake using radiolabeled glucose assays. Rac1 activation and phospho-cofilin were analyzed by G-LISA and Western blotting, and Gαq/11 involvement was tested using YM-254890. TAS1R3 mRNA and protein levels were significantly reduced in T2D skeletal muscle. Pharmacological inhibition or the knockdown of TAS1R3 impaired insulin-stimulated glucose uptake in myotubes. TAS1R3 regulates skeletal muscle glucose uptake through a non-canonical insulin signaling pathway involving Rac1 and phospho-cofilin, independent of IRS1-AKT and Gαq/11 signaling. These findings identify TAS1R3 as a key determinant of Rac1-mediated glucose uptake and a potential therapeutic target for improving insulin sensitivity in T2D. Full article
Show Figures

Figure 1

22 pages, 4291 KB  
Article
Photodynamic Activation of Mammalian and Avian Cholecystokinin Type 1 Receptor Outside of the Pancreatic Acinar Cell Microenvironment
by Jie Wang and Zong Jie Cui
Int. J. Mol. Sci. 2025, 26(24), 12011; https://doi.org/10.3390/ijms262412011 - 13 Dec 2025
Viewed by 380
Abstract
Cholecystokinin 1 receptor (CCK1R) is activated by singlet oxygen (1O2) in type II photodynamic action in isolated rat, mouse, and Peking duck pancreatic acini. To examine whether this is maintained outside the microenvironment of pancreatic acinar cell, photodynamic activation [...] Read more.
Cholecystokinin 1 receptor (CCK1R) is activated by singlet oxygen (1O2) in type II photodynamic action in isolated rat, mouse, and Peking duck pancreatic acini. To examine whether this is maintained outside the microenvironment of pancreatic acinar cell, photodynamic activation of CCK1R from human, rat, mouse, and Peking duck expressed in CHO-K1 cells was examined, as monitored with Fura-2 fluorescence calcium imaging. Photodynamic action with sulphonated aluminum phthalocyanine was found to trigger persistent calcium oscillations in CCK1R-CHO-K1 cells transfected with human, rat, mouse or Peking duck CCK1R gene, which were blocked by 1O2 quencher Trolox C. After tagging protein photosensitizer miniSOG to C-terminus of these CCK1R, photodynamic action was found to similarly trigger persistent calcium oscillations in CCK1R-miniSOG-CHO-K1 cells expressing human, rat, mouse, and Peking duck receptor constructs. Incubation with Trolox C 300 μM during LED light irradiation also prevented photodynamic CCK1R activation in CCK1R-miniSOG-CHO-K1 cells. In contrast, human M3R was not photodynamically activated with SALPC or tagged miniSOG as the photosensitizer. These data, together, suggest that photodynamic CCK1R activation is maintained outside of the pancreatic acinar cell, making possible photodynamic CCK1R activation in CCK1R-expressing organs and tissues other than the pancreas, with high spatiotemporal precision. Full article
(This article belongs to the Special Issue Novel Mechanisms of Receptor Activation)
Show Figures

Figure 1

23 pages, 1525 KB  
Review
The CB2 Receptor in Immune Regulation and Disease: Genetic Architecture, Epigenetic Control, and Emerging Therapeutic Strategies
by Hilal Kalkan and Nicolas Flamand
DNA 2025, 5(4), 59; https://doi.org/10.3390/dna5040059 - 11 Dec 2025
Viewed by 824
Abstract
The cannabinoid receptor type 2 (CB2) is increasingly recognized as a crucial regulator of neuroimmune balance in the brain. In addition to its well-established role in immunity, the CB2 receptor has been identified in specific populations of neurons and glial [...] Read more.
The cannabinoid receptor type 2 (CB2) is increasingly recognized as a crucial regulator of neuroimmune balance in the brain. In addition to its well-established role in immunity, the CB2 receptor has been identified in specific populations of neurons and glial cells throughout various brain regions, and its expression is dynamically increased during inflammatory and neuropathological conditions, positioning it as a potential non-psychoactive target for modifying neurological diseases. The expression of the CB2 gene (CNR2) is finely tuned by epigenetic processes, including promoter CpG methylation, histone modifications, and non-coding RNAs, which regulate receptor availability and signaling preferences in response to stress, inflammation, and environmental factors. CB2 signaling interacts with TRP channels (such as TRPV1), nuclear receptors (PPARγ), and orphan G Protein-Coupled Receptors (GPCRs, including GPR55 and GPR18) within the endocannabinoidome (eCBome), influencing microglial characteristics, cytokine production, and synaptic activity. We review how these interconnected mechanisms affect neurodegenerative and neuropsychiatric disorders, underscore the species- and cell-type-specificities that pose challenges for translation, and explore emerging strategies, including selective agonists, positive allosteric modulators, and biased ligands, that leverage the signaling adaptability of the CB2 receptor while reducing central effects mediated by the CB1 receptor. This focus on the neuro-centric perspective repositions the CB2 receptor as an epigenetically informed, context-dependent hub within the eCBome, making it a promising candidate for precision therapies in conditions featuring neuroinflammation. Full article
Show Figures

Figure 1

17 pages, 3156 KB  
Article
Type-1 Cannabinoid Receptor Promiscuous Coupling: Computational Insights into Receptor-G Protein Interaction Dynamics
by Alessandro Berghella, Tomasz Maciej Stepniewski, Annalaura Sabatucci, Marta Lopez-Balastegui, Krzysztof Nowicki, Beatrice Dufrusine, Jana Selent and Enrico Dainese
Int. J. Mol. Sci. 2025, 26(24), 11905; https://doi.org/10.3390/ijms262411905 - 10 Dec 2025
Viewed by 439
Abstract
Cannabinoid receptor (CB1), a G protein coupled receptor (GPCR), is a known pharmacological target in several diseases and modulates key physiological processes through Gi protein-mediated signaling. However, recent evidence suggests that CB1 can also activate other G proteins, including the stimulatory Gs protein, [...] Read more.
Cannabinoid receptor (CB1), a G protein coupled receptor (GPCR), is a known pharmacological target in several diseases and modulates key physiological processes through Gi protein-mediated signaling. However, recent evidence suggests that CB1 can also activate other G proteins, including the stimulatory Gs protein, a phenomenon with unclear structural determinants. Here, we use a computational approach to elucidate the structural basis of the CB1-Gs interaction. Protein–protein docking and extensive molecular dynamics simulations yield a model for the CB1-Gs complex that agrees well with both existing experimental data and available GPCR-Gs structures, supporting its validity. This work provides new insights into the structural basis of CB1’s ability to couple with different G-proteins. The model provides a basis for future studies dissecting the functional consequences of CB1-Gs signaling and the development of improved therapeutics targeting the CB1 receptor and the wider endocannabinoid system. Full article
Show Figures

Figure 1

30 pages, 1934 KB  
Review
Low pH, High Stakes: A Narrative Review Exploring the Acid-Sensing GPR65 Pathway as a Novel Approach in Renal Cell Carcinoma
by Michael Grant, Barbara Cipriani, Alastair Corbin, David Miller, Alan Naylor, Stuart Hughes, Tom McCarthy, Sumeet Ambarkhane, Danish Memon, Michael Millward, Sumanta Pal and Ignacio Melero
Cancers 2025, 17(23), 3883; https://doi.org/10.3390/cancers17233883 - 4 Dec 2025
Viewed by 1027
Abstract
Renal cell carcinoma (RCC) is a biologically heterogeneous malignancy accounting for 3% of adult cancers globally. Despite advances in immune checkpoint inhibitors (ICIs) and vascular endothelial growth factor (VEGF)-targeted therapies, durable disease control remains elusive for many patients. Increasing evidence implicates the acidic [...] Read more.
Renal cell carcinoma (RCC) is a biologically heterogeneous malignancy accounting for 3% of adult cancers globally. Despite advances in immune checkpoint inhibitors (ICIs) and vascular endothelial growth factor (VEGF)-targeted therapies, durable disease control remains elusive for many patients. Increasing evidence implicates the acidic tumour microenvironment (TME) as a critical mediator of RCC progression, immune evasion, and therapeutic resistance. Solid tumours, including RCC, exhibit reversed pH gradients, characterised by acidic extracellular (pH 6.2–6.9) and alkaline intracellular conditions. This dysregulation arises from enhanced glycolysis, hypoxia-driven lactate accumulation, and the overexpression of pH-regulating enzymes such as carbonic anhydrase (CA9). Acidic TMEs impair cytotoxic T-cell and NK-cell activity, promote tumour-associated macrophage (TAM) polarisation towards an immunosuppressive phenotype, and upregulate alternative immune checkpoints. These mechanisms collectively undermine ICI efficacy and contribute to primary and secondary treatment resistance. Proton-sensing G-protein-coupled receptors (GPCRs), notably GPR65, have emerged as pivotal mediators linking extracellular acidosis to immune dysfunction. Preclinical studies demonstrate that GPR65 antagonists restore anti-tumour immune activity by reversing acidosis-driven immunosuppression and enhancing antigen processing. In RCC models, selective GPR65 inhibitors have shown the ability to reduce immunosuppressive cytokine IL-10 production, induce immunoproteasome activation, and synergise with anti-PD-1 therapy. The first-in-class GPR65 inhibitor, PTT-4256, is now under evaluation in the Phase I/II RAISIC-1 trial (NCT06634849) in solid tumours, including RCC. Targeting acid-sensing pathways represents a novel and promising therapeutic strategy in RCC, aiming to remodel the TME and overcome ICI resistance. Integrating GPR65 inhibition with existing immunotherapies may define the next era of RCC management, warranting continued translational and clinical investigation. Full article
Show Figures

Figure 1

26 pages, 703 KB  
Review
To Be Biased or Not to Be: A Play for G-Protein Coupled Receptors
by Nikitas G. Liolitsas, Evangelia Pantazaka and Evangelia Papadimitriou
Int. J. Transl. Med. 2025, 5(4), 56; https://doi.org/10.3390/ijtm5040056 - 4 Dec 2025
Viewed by 1258
Abstract
G protein-coupled receptors (GPCRs) are the largest family of diverse receptors in eukaryotic organisms, playing a critical role in modulating human physiology. It therefore comes as no surprise that about 36% of all currently available drugs target this superfamily. When an agonist binds [...] Read more.
G protein-coupled receptors (GPCRs) are the largest family of diverse receptors in eukaryotic organisms, playing a critical role in modulating human physiology. It therefore comes as no surprise that about 36% of all currently available drugs target this superfamily. When an agonist binds to a GPCR, it induces conformational changes in the receptor that allow it to interact with intracellular proteins. This interaction triggers downstream signaling cascades that alter the cell’s activity. GPCR signaling is complex, as GPCRs transmit signals through coupling with G proteins, arrestins, and numerous other intracellular effectors. Different ligands, receptor subtypes, and cellular environments can result in the activation of distinct signaling pathways. Biased signaling through GPCRs has emerged as a frontier area in pharmacological research efforts towards designing targeted therapeutic interventions and enhancing drug efficacy and safety. This review presents the types of bias associated with GPCRs and the mechanisms underlying biased signaling. Examples of biased ligands and their therapeutic implications will be discussed. In addition, the inherent challenges in measuring signaling bias, and especially the translational gap between in vitro and in vivo assays and clinical outcomes, will be outlined. Full article
Show Figures

Figure 1

17 pages, 1560 KB  
Review
Biological Breakthroughs and Drug Discovery Revolution via Cryo-Electron Microscopy of Membrane Proteins
by Vitor Hugo Balasco Serrão
Membranes 2025, 15(12), 368; https://doi.org/10.3390/membranes15120368 - 1 Dec 2025
Viewed by 1769
Abstract
The application of cryo-electron microscopy (cryo-EM) in membrane protein structural biology has catalyzed unprecedented advances in our understanding of fundamental biological processes and transformed drug discovery paradigms. This review briefly describes the biological achievements enabled using cryo-EM techniques, including single particle analysis (SPA), [...] Read more.
The application of cryo-electron microscopy (cryo-EM) in membrane protein structural biology has catalyzed unprecedented advances in our understanding of fundamental biological processes and transformed drug discovery paradigms. This review briefly describes the biological achievements enabled using cryo-EM techniques, including single particle analysis (SPA), micro-electron diffraction (microED), and subtomogram averaging (STA), in elucidating the structures and functions of membrane proteins, ion channels, transporters, and viral glycoproteins. We highlight how these structural insights have revealed druggable sites, enabled structure-based drug design, and provided mechanistic understanding of disease processes. Key biological targets include G protein-coupled receptors (GPCRs), ion channels implicated in neurological disorders, respiratory chain complexes, viral entry machinery, and membrane transporters. The integration of cryo-EM with computational drug design has already yielded clinical candidates and approved therapeutics, marking a new era in membrane protein pharmacology. Full article
Show Figures

Figure 1

18 pages, 6298 KB  
Article
Molecular Dynamics Insights into TAS1R2 Transmembrane Domain Activation
by Yongcheng Lu, Xinyi Ma, Ziyue Meng and Meng Cui
Int. J. Mol. Sci. 2025, 26(23), 11464; https://doi.org/10.3390/ijms262311464 - 26 Nov 2025
Viewed by 481
Abstract
Sweet taste receptors (STRs) are class C G protein-coupled receptors (GPCRs) that function as heterodimers of TAS1R2 and TAS1R3. These receptors possess multiple binding sites and can be activated by a wide range of sweet-tasting compounds. Interestingly, TAS1R2 alone or even its extracellular [...] Read more.
Sweet taste receptors (STRs) are class C G protein-coupled receptors (GPCRs) that function as heterodimers of TAS1R2 and TAS1R3. These receptors possess multiple binding sites and can be activated by a wide range of sweet-tasting compounds. Interestingly, TAS1R2 alone or even its extracellular domain-truncated form (TAS1R2-TMD), can act as a functional receptor. Previous studies demonstrated that the sweetener S819 and the sweet inhibitor amiloride act through the transmembrane domain (TMD) of TAS1R2; however, the molecular mechanisms underlying these ligand-specific effects remain unclear, largely due to the historical lack of experimentally determined full-length STR structures. Recent breakthroughs in cryo-EM structural determination of the full-length TAS1R2/TAS1R3 complex now offer an unprecedented opportunity to elucidate receptor activation mechanisms at atomic resolution. In this study, we investigated ligand-induced conformational dynamics of hTAS1R2-TMD using microsecond-scale molecular dynamics (MD) simulations on three systems: hTAS1R2-TMD/S819 (agonist-bound), hTAS1R2-TMD/amiloride (antagonist-bound), and hTAS1R2-TMD (apo). Comparative analyses revealed that agonist and antagonist binding distinctly modulate key structural switches, including the conserved ionic lock (E6.35-R3.50), which stabilizes the inactive state and disrupts upon activation. Notably, we identified a novel salt bridge (D7.32-R3.32) that forms preferentially in the active state, potentially serving as a unique molecular switch for TAS1R2. Additional analyses uncovered ligand-specific rearrangements in hydrogen-bonding and hydrophobic interaction networks. These results provide atomistic insights into how agonists and antagonists differentially modulate TAS1R2 activation and lay a structural foundation for designing novel sweeteners and taste modulators. Full article
Show Figures

Figure 1

6 pages, 178 KB  
Editorial
Special Issue “Molecular Research on Adenosine Receptors: From Cell Biology to Human Diseases”
by Federica Mannino
Int. J. Mol. Sci. 2025, 26(23), 11298; https://doi.org/10.3390/ijms262311298 - 22 Nov 2025
Viewed by 618
Abstract
Adenosine receptors (ARs), members of the G protein-coupled receptor (GPCR) superfamily, have emerged as central regulators of numerous physiological and pathological processes [...] Full article
Back to TopTop