Next Article in Journal
Co-Immunoprecipitation-Coupled Mass Spectrometry Analysis of Zyxin’s Interactome and Phosphosites in Early Xenopus laevis Development
Previous Article in Journal
Evaluation of the Unintended Effects of Herbicide-Resistant Soybean Seeds via TMT Quantitative Proteomics and Flavonoid-Targeted Metabolomics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

G Protein-Coupled Receptors in Cerebrovascular Diseases: Signaling Mechanisms and Therapeutic Opportunities

Nantong Laboratory of Development and Diseases, School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2026, 27(2), 736; https://doi.org/10.3390/ijms27020736 (registering DOI)
Submission received: 11 December 2025 / Revised: 9 January 2026 / Accepted: 10 January 2026 / Published: 11 January 2026
(This article belongs to the Section Molecular Neurobiology)

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 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.

1. Introduction

Cerebrovascular diseases remain a leading cause of mortality and long-term disability worldwide, with ischemic stroke representing the most prevalent and devastating manifestation [1,2,3]. Despite substantial advances in acute reperfusion therapies, effective strategies to limit secondary injury and promote long-term functional recovery remain limited [4,5,6,7]. A deeper understanding of the molecular signaling pathways governing cerebrovascular dysfunction is therefore essential.
G protein-coupled receptors (GPCRs) constitute the largest family of signal transduction receptors and regulate diverse physiological processes relevant to cerebrovascular pathology, including vascular tone, immune activation, and neuronal survival [8,9,10]. However, GPCR signaling in the brain is inherently complex. The same receptor may engage distinct downstream pathways depending on cell type, ligand availability, and microenvironmental conditions, leading to context-dependent and sometimes contradictory biological outcomes [11,12,13,14]. This complexity has contributed to inconsistent interpretations across studies and has hindered successful clinical translation.
Most existing reviews describe GPCRs primarily in a receptor-centered or single-cell-type manner, often emphasizing vascular smooth muscle cells (VSMCs), endothelial cells, or the blood–brain barrier (BBB) in isolation [15]. Such approaches insufficiently capture the coordinated, multicellular signaling processes that underlie cerebrovascular injury and recovery. Increasing evidence indicates that neurons, glial cells, vascular cells, and infiltrating immune cells function as an integrated neurovascular unit (NVU), in which intercellular communication critically shapes both acute injury and repair responses [16]. Moreover, the temporal dynamics of GPCR signaling across acute ischemia, secondary injury, and post-stroke repair phases are rarely addressed in an integrated manner.
In this review, we adopt a disease-process-oriented framework to systematically examine GPCR signaling in cerebrovascular disorders. By incorporating an NVU-centered perspective and emphasizing temporal phase-specific effects, we aim to clarify how GPCR signaling contributes to injury progression and recovery, and to identify conceptual principles relevant for therapeutic development.

2. Canonical GPCR Signaling Pathways

2.1. GPCRs and the G Protein Family

GPCRs, a large superfamily conserved from yeast to humans, mediate most signaling pathways and sensory functions, with ~800 mammalian genes and ~30% of FDA-approved drugs [17]. Despite the structural diversity of GPCR ligands (from photons to peptides), all ligands induce GPCR conformational changes to activate G proteins. Structurally, all GPCRs share an extracellular N-terminus, seven transmembrane α-helices, three extracellular loops, three intracellular loops, and a cytoplasmic C-terminus [18]. Phylogenetic analysis classifies human GPCRs into five major families with distinct functions:
  • Rhodopsin family (Class A): The largest subgroup (~70% of GPCRs), including adrenergic receptors, endothelin receptors, and chemokine receptors (e.g., CCR5, CXCR4).
  • Secretin family (Class B): Ligands are peptide hormones (e.g., glucagon), with receptors involved in metabolic regulation.
  • Glutamate family (Class C): Includes metabotropic glutamate receptors (mGluRs) and γ-aminobutyric acid (GABA) receptors, critical for neural signaling.
  • Adhesion family: Regulates cell adhesion and tissue development, with roles in vascular integrity.
  • Frizzled/Taste2 family: Mediates Wnt signaling (Frizzled) and taste perception (Taste2).
G proteins are subdivided into Gαs, Gαi/o, Gαq/11, and Gα12/13 families. Ligand binding to GPCRs induces GTP loading on Gα and dissociation from Gβγ, enabling both subunits to engage downstream effectors.

2.2. Core GPCR Signal Transduction Mechanisms

The main effectors of G proteins include the adenylate cyclase (AC) system, phospholipase C (PLC), and others. Different G protein families activate their respective effectors, thereby initiating downstream signaling pathways (Figure 1).
Gαs activates AC to convert ATP into cAMP, which stimulates PKA and cyclic nucleotide-gated channels, whereas Gαi inhibits AC, reducing cAMP levels [19]. Gαq activates phospholipase Cβ (PLCβ) to hydrolyze PIP2 into IP3 and DAG; IP3 triggers Ca2+ release from the endoplasmic reticulum, and DAG activates PKC, inducing downstream cellular responses [20]. Members of the RGS (Regulator of G-protein Signaling) family can activate the GTPase activity of specific Gα subunits. p115RhoGEF, a guanine nucleotide exchange factor for Rho proteins, can specifically stimulate the GTPase activity of Gα12/13 in G proteins [21]. Additionally, Gα12 can exert its functions by interacting with receptor tyrosine kinase family members, cadherins, and other proteins [22]. In addition to Gα proteins, Gβγ proteins can also regulate AC [23,24], PLCβ [25,26], inward rectifier K+ channels [27,28], and voltage-gated Ca2+ channels [29,30], thereby facilitating signal transduction.
These signaling pathways regulate intracellular cAMP production, Ca2+ release, PKC activation, and RhoA-dependent cytoskeletal remodeling—mechanisms highly relevant to vascular tone, endothelial permeability, and inflammatory activation.

3. GPCR Signaling Within the NVU

Within the NVU, GPCR signaling is organized across distinct cell types to enable coordinated responses to neuronal activity, metabolic stress, and inflammation [31]. Rather than functioning as isolated components, GPCRs form intercellular signaling axes that couple vascular, immune, and neuronal functions [31]. The functional outcome of GPCR activation depends on both cellular context and disease stage, so that disruption of these signaling networks contributes to neurovascular dysfunction and impaired recovery [32,33].

3.1. Endothelial–Pericyte GPCR Crosstalk in Vascular Stability

Endothelial cells and pericytes form the structural and functional backbone of the cerebral microvasculature. GPCR signaling between these two cell types regulates capillary tone, vascular permeability, and microvascular stability [34,35]. Endothelial GPCRs respond to circulating ligands such as adenosine [36], sphingosine-1-phosphate (S1P) [37,38], and prostaglandins [39], thereby modulating nitric oxide production, cytoskeletal dynamics, and junctional integrity. These endothelial responses, in turn, influence pericyte contractility and capillary perfusion.
Conversely, pericyte-expressed GPCRs sense local metabolic and neurotransmitter cues and relay signals back to endothelial cells to fine-tune microvascular resistance [40]. Disruption of this bidirectional GPCR-mediated communication destabilizes capillary networks and predisposes the vasculature to hypoperfusion and leakage during cerebrovascular injury.

3.2. Astrocyte–Endothelial GPCR Signaling at the BBB

Astrocytes play a pivotal role in regulating BBB function through close physical and biochemical interactions with endothelial cells. Astrocyte-derived ligands, including S1P [37,38], prostaglandins [39], and neurotransmitter metabolites [40], activate endothelial GPCRs to modulate tight junction organization and transcellular transport pathways. Through these mechanisms, astrocytic GPCR signaling indirectly governs BBB permeability and endothelial responsiveness to injury [32].
Endothelial GPCR activation also feeds back to astrocytes by altering extracellular ion balance and metabolic substrate availability, thereby influencing astrocytic reactivity [31]. This reciprocal GPCR-mediated signaling axis establishes a regulatory loop that maintains BBB homeostasis under physiological conditions and determines barrier vulnerability under pathological stress.

3.3. Microglia–Neuron GPCR Communication in Immune Surveillance

Microglia serve as the resident immune cells of the central nervous system and continuously monitor neuronal activity and tissue integrity. GPCRs expressed on microglia detect extracellular nucleotides, chemokines, and neurotransmitters released by stressed or active neurons [41]. Activation of these receptors shapes microglial surveillance behavior, cytokine secretion, and phagocytic activity [42].
Neuronal GPCR signaling reciprocally responds to microglia-derived mediators, thereby modulating neuronal excitability and stress responses [43]. This bidirectional GPCR-dependent communication ensures adaptive immune surveillance under homeostatic conditions but may become maladaptive when dysregulated, contributing to inflammatory amplification in cerebrovascular disease [44].

3.4. Integrated GPCR Networks Across the NVU

Although individual GPCR pathways can be assigned to specific NVU cell types, their functional consequences emerge from integrated network behavior. Endothelial activation alters immune cell recruitment [45], microglial responses influence vascular permeability [32], and astrocytic signaling shapes both vascular and neuronal function [46,47]. GPCRs act as nodal regulators within this network, enabling rapid and coordinated adaptation to metabolic and inflammatory cues.
The repeated involvement of GPCRs such as S1P, PGE2, and A2AR across endothelial, glial, and neuronal compartments does not reflect redundancy, but rather context-specific signal partitioning within the NVU (Figure 2) [36,37,38,48,49,50]. By coupling to distinct intracellular effectors in different cell types, these receptors enable parallel regulation of vascular stability, immune activation, and neuronal excitability. This multi-level integration provides a mechanistic explanation for the pronounced context- and stage-dependence of GPCR signaling observed in cerebrovascular disease.
This NVU-centered view provides a conceptual framework for understanding how GPCR signaling synchronizes vascular, immune, and neuronal processes. It also highlights why therapeutic modulation of GPCRs often yields context-dependent outcomes, as perturbation of one signaling node can propagate across multiple NVU components.

4. GPCRs in Acute Cerebrovascular Injury

Acute cerebrovascular injury, particularly ischemic stroke, triggers a rapid cascade of pathological events, including cerebral hypoperfusion, sterile inflammation, and BBB disruption. GPCR signaling plays a central role in coordinating these processes through tightly regulated yet highly context-dependent actions across NVU cell types. Importantly, during the acute phase, GPCR-mediated responses frequently exert dual effects, with protective or deleterious outcomes determined by receptor subtype, cellular localization, and temporal window of activation [32,33].

4.1. Regulation of Cerebral Blood Flow During Acute Ischemia

Cerebral blood flow dysregulation is a defining feature of acute ischemic stroke [72]. GPCRs expressed on VSMCs and pericytes critically regulate vessel tone and microvascular perfusion during this phase. Activation of Gs-coupled GPCRs, including A2AR and prostaglandin E2 receptors EP2 and EP4, promotes cAMP-dependent vasodilation, thereby facilitating collateral circulation and partial restoration of perfusion in ischemic regions [50,66]. Experimental evidence further indicates that EP2/EP4 signaling supports functional hyperemia and capillary recruitment, consistent with a protective role in acute ischemia [73].
In contrast, activation of Gq- and G12/13-coupled GPCRs, such as AT1R, endothelin receptors, and prostaglandin F2α receptor (FP), induces intracellular Ca2+ elevation and RhoA-mediated contractile signaling [54,74,75]. These pathways exacerbate vasoconstriction, impair microvascular reperfusion, and contribute to the no-reflow phenomenon observed after recanalization. The balance between these opposing GPCR signaling axes critically shapes perfusion heterogeneity during the hyperacute stage of stroke and influences infarct expansion.

4.2. GPCR-Mediated Neuroinflammation and Secondary Injury

Neuroinflammation rapidly emerges following ischemic insult and represents a major contributor to secondary brain injury [76]. GPCRs expressed on endothelial cells, microglia, and infiltrating immune cells coordinate leukocyte recruitment, cytokine production, and inflammatory amplification within the ischemic microenvironment.
Endothelial GPCR activation regulates the initial inflammatory interface between circulating immune cells and the cerebral vasculature. For example, the proton-sensing GPCR GPR4 responds to ischemia-induced acidosis by enhancing endothelial activation, leukocyte adhesion, and vascular permeability, thereby facilitating immune cell infiltration [45]. Concurrently, microglial GPCRs, including chemokine and purinergic receptors, modulate inflammatory polarization and cytokine release, indirectly influencing neuronal vulnerability [77].
These GPCR-driven inflammatory responses do not occur in isolation but form feed-forward signaling loops across NVU components. Endothelial activation promotes immune cell entry, which in turn amplifies microglial reactivity and further disrupts vascular homeostasis. During the acute phase, excessive or prolonged activation of pro-inflammatory GPCR signaling exacerbates tissue damage, underscoring the importance of temporally controlled modulation rather than indiscriminate suppression of inflammation [77,78].

4.3. GPCR Regulation of BBB Disruption in the Acute Phase

BBB breakdown is a hallmark of acute cerebrovascular injury and a major contributor to cerebral edema and hemorrhagic transformation [79]. GPCR signaling influences BBB integrity primarily through rapid modulation of endothelial cytoskeletal dynamics and tight junction organization during the early injury phase [80,81,82].
Activation of GPCRs such as S1P3, protease-activated receptors (PARs), and bradykinin B2 receptors (B2Rs) promotes RhoA-ROCK signaling, actin stress fiber formation, and junctional disassembly, thereby increasing paracellular permeability [58,83]. These pathways exacerbate vascular leakage under ischemic and inflammatory conditions. In contrast, endothelial S1P1 signaling counteracts barrier disruption by stabilizing tight junctions and limiting cytoskeletal contraction, underscoring receptor subtype-specific effects within the same ligand system [37].
Notably, acute BBB disruption is mechanistically distinct from later-stage barrier remodeling and repair. During the acute phase, GPCR-mediated signaling primarily determines the severity and duration of barrier breakdown, thereby shaping early edema formation and secondary injury progression.

5. GPCRs in Neuronal Survival, Plasticity, and Functional Recovery

Beyond their established roles in acute vascular regulation, GPCRs critically shape long-term outcomes by orchestrating neurovascular remodeling. During the subacute and recovery phases, GPCR signaling transitions from injury mitigation to the promotion of neuronal resilience, synaptic plasticity, and circuit reorganization [84]. Importantly, these processes are not driven solely by neurons but rely on coordinated signaling among neurons, astrocytes, and remodeling vessels.

5.1. GPCR Regulation of Delayed Neuronal Survival

Following the acute insult, neurons in the ischemic penumbra remain vulnerable to delayed apoptosis and metabolic stress. During this window, GPCRs function as molecular “survival switches” that modulate cell fate decisions.
GPR37, a Gi-coupled receptor enriched in neurons and oligodendrocytes, acts as an endogenous neuroprotective brake [85]. Genetic deletion of GPR37 exacerbates ischemic injury by promoting caspase-3 activation and impairing mTOR signaling [85].
Furthermore, proton-sensing GPCRs link the acidic ischemic microenvironment to adaptive survival responses. TDAG8 (GPR65), although predominantly expressed in immune and glial cells, exerts paracrine neuroprotective effects, whereas neuronal GPR68 senses acidosis to trigger PKC-dependent survival pathways [86,87]. These observations highlight how GPCRs decode metabolic cues, such as extracellular pH, to preserve neuronal integrity during the subacute phase.

5.2. GPCR Modulation of Synaptic Plasticity and NVU Remodeling

Functional recovery after stroke depends on synaptic reorganization, a process tightly regulated by interactions between neurons and astrocytes within the tripartite synapse. GPCRs modulate this form of plasticity by controlling dendritic excitability, neurotransmission, and neurotrophic support.
The chemokine receptor CCR5 acts as a critical molecular “brake” on post-stroke plasticity [88,89]. Inhibition of CCR5 enhances motor recovery by disinhibiting CREB-dependent transcriptional programs that drive axonal remodeling and synaptic strengthening [88,89]. This finding represents a paradigm shift, suggesting that suppressing maladaptive GPCR signaling can be as crucial as activating pro-repair pathways during recovery.
Additionally, neuropeptide signaling via PACAP/PAC1 exemplifies coordinated neuro-glial coupling within the NVU [71]. PAC1 activation not only enhances neuronal excitability via Kv4.2 channels but also modulates astrocytic support for synaptic strengthening [90]. Such coordinated signaling ensures that circuit rewiring is accompanied by appropriate metabolic and trophic support from surrounding NVU components.

5.3. Stage-Dependent GPCR Signaling Switches and Functional Outcomes

A recurring challenge in translational research is the pronounced context dependence of GPCR signaling. The same receptor may exert opposing effects depending on disease stage, cell type, and duration of activation. β2ARs exemplify this duality. While β2AR activation may alter acute inflammation by inhibiting NF-κB, prolonged activation during the recovery phase may alter neuronal excitability and compromise network stability [91,92].
Similarly, GPCRs that drive pro-inflammatory responses during the acute phase may become necessary at later stages to facilitate debris clearance, synaptic remodeling, and remyelination [93]. These observations emphasize that GPCRs function as dynamic modulators rather than static therapeutic targets. Effective intervention, therefore, requires precise targeting of defined temporal windows, rather than blanket activation or inhibition.
Collectively, failure to account for these temporal switches can adversely affect functional outcomes. GPCR interventions that are beneficial for acute neuroprotection may impair cognitive recovery or long-term network stability if applied during later phases, highlighting the need for temporally optimized strategies to maximize sustained motor and cognitive rehabilitation.

6. Translational and Therapeutic Challenges of Targeting GPCRs in Cerebrovascular Diseases

GPCRs regulate multiple pathological processes in cerebrovascular diseases, including dysregulation of cerebral blood flow, neuroinflammation, BBB breakdown, and neuronal injury (Figure 3). Accordingly, GPCRs have emerged as attractive therapeutic targets in experimental stroke models. However, despite robust neuroprotective effects reported in rodents, the clinical translation of GPCR-targeted strategies has largely been unsuccessful.

6.1. Clinically Validated and Repurposed GPCR Targets

Several GPCR families have been investigated as therapeutic targets in stroke (Table 1).
Adenosine receptors (ARs)
ARs regulate cerebral perfusion, inflammation, neuronal excitability, and BBB stability. Adenosine A1 receptor (A1AR) activation reduces excitotoxicity, while A2AR modulation promotes vasodilation and can limit ischemic injury in preclinical models [118,123]. Selective A2AR antagonists, such as istradefylline, approved for PD, are being explored for post-stroke neuroprotection due to their anti-inflammatory and neuroprotective potential [124,125].
Endothelin receptors (ETRs)
ETARs mediate ET-1-induced cerebral vasoconstriction and contribute to ischemic damage [126]. Bosentan, a dual endothelin receptor antagonist, improves cerebral blood flow in preclinical ischemia models but may exacerbate systemic or infectious complications, highlighting the need for context-specific targeting strategies [52,127].
Sphingosine-1-phosphate receptors (S1P receptors)
S1P signaling influences BBB integrity, neuroinflammation, and neurogenesis. FTY720 (fingolimod) activates S1P receptors on neurons, glia, and endothelial cells, promoting anti-apoptotic pathways and facilitating the proliferation of neural precursor cells [128,129]. In this regard, S1P-targeted therapies exemplify the multicellular actions of GPCRs within the NVU.
Chemokine receptors (CCR5 and CXCR4)
These GPCRs regulate immune cell recruitment and post-stroke neuroplasticity [130]. CCR5 antagonism with maraviroc enhances motor and cognitive recovery in rodent models, and emerging human data support its potential for post-stroke neural repair [88,89,131]. In parallel, its therapeutic applications in long COVID/post-acute sequelae of COVID-19 are being evaluated in ongoing or planned clinical trials [132].
Serotonin receptors (5-HTRs)
Several serotonin receptor subtypes modulate cerebrovascular tone. Activation of 5-HT1 and 5-HT2 receptors reduces cerebrovascular constriction, while 5-HT3 functions as a ligand-gated ion channel [119,120]. Agents such as sumatriptan, widely used in migraine therapy, influence cerebrovascular dynamics and may hold relevance for cerebrovascular modulation [121,122].

6.2. Discrepancy Between Preclinical Efficacy and Clinical Outcomes

A recurring challenge in GPCR-targeted stroke therapy is the poor concordance between preclinical neuroprotection and clinical efficacy [133]. Broad-spectrum modulation of adenosine receptors exemplifies this translational gap. In experimental ischemia, pharmacological antagonism or genetic deletion of A2ARs reduces infarct volume, attenuates neuroinflammation, and preserves synaptic integrity [65,134,135]. However, clinical attempts to modulate adenosine signaling have been constrained by narrow therapeutic windows, cardiovascular side effects, and context-dependent disruption of BBB integrity [136,137].
Similar discrepancies have been observed for other GPCR systems, including endothelin and chemokine receptors, which demonstrate protective effects in tightly controlled experimental paradigms but yield inconsistent or adverse outcomes in heterogeneous patient populations [138]. These findings indicate that GPCR-mediated neuroprotection is highly dependent on injury stage, cell type, and signaling context, and cannot be reliably extrapolated from simplified models lacking temporal and spatial resolution.

6.3. Pharmacological Barriers: BBB Penetration, Off-Target Effects, and Therapeutic Windows

Pharmacokinetic and pharmacodynamic limitations further impede the clinical translation of GPCR-targeted therapies [139]. Many GPCR ligands fail to achieve effective concentrations within ischemic brain tissue due to restricted BBB permeability or active efflux by transporters such as P-glycoprotein [140]. Even when BBB penetration is achieved, systemic administration frequently leads to off-target effects, particularly within the peripheral vasculature, resulting in hypotension or impaired cerebral autoregulation [141].
Moreover, the temporal dynamics of GPCR signaling present a fundamental challenge. GPCRs such as A2ARs and β2ARs exhibit phase-dependent effects, conferring vascular or anti-inflammatory benefits during early ischemia while potentially exacerbating BBB leakage or immune activation at later stages [65,91,92]. Therapeutic interventions that fail to account for these shifting windows may inadvertently convert protective signaling into deleterious outcomes [142,143].

6.4. Species Differences and Limitations of Rodent Models

Species-specific differences in GPCR expression patterns, coupling preferences, and signaling bias further limit translational predictability. GPCRs implicated in cerebrovascular pathology, including A2ARs, CXCR4, and β2ARs, exhibit divergent cell-type distributions and downstream signaling cascades in rodents compared with humans, particularly within endothelial cells, microglia, and pericytes [65,91,92,106]. These discrepancies likely contribute to the frequent failure of GPCR-targeted neuroprotective strategies during clinical translation.
In addition, commonly used rodent stroke models inadequately capture the vascular comorbidities, aging, and immune heterogeneity characteristic of human cerebrovascular diseases [144]. Consequently, GPCR-dependent mechanisms identified in young, otherwise healthy animals may not accurately reflect receptor function in the diseased human brain.

6.5. Emerging Human-Relevant Models for GPCR Research

To bridge this translational gap, human-relevant experimental platforms are increasingly being adopted. Induced pluripotent stem cell (iPSC)-derived NVU models enable controlled investigation of GPCR signaling across human neurons, astrocytes, endothelial cells, and pericytes within an integrated multicellular context [145,146]. Brain organoids and microfluidic NVU-on-chip systems further permit spatiotemporal modeling of ischemia–reperfusion injury and intercellular GPCR-mediated communication under human-specific conditions [147].
In parallel, single-cell and spatial transcriptomic analyses of human stroke tissue provide unprecedented resolution of GPCR expression dynamics across disease stages and cell types [148]. Together, these approaches offer critical guidance for identifying phase-specific and cell-selective GPCR targets with improved translational potential (Figure 4).

6.6. Perspectives: Toward Precision GPCR Targeting in Cerebrovascular Diseases

Collectively, these challenges highlight the need for a paradigm shift in GPCR-based therapeutic strategies for cerebrovascular diseases, from receptor-centric modulation toward phase-specific and cell-type-resolved targeting within the NVU. Advances in GPCR pharmacology and drug design are beginning to enable this transition.
Structural biology approaches, including cryo-electron microscopy and X-ray crystallography, have provided high-resolution insights into GPCR–ligand interactions, facilitating the rational development of subtype-selective and signaling-biased ligands that preferentially engage protective pathways while minimizing deleterious effects [149,150]. In parallel, emerging therapeutic modalities, such as nanobody-based biologics and RNA-based interventions, offer enhanced specificity and improved delivery to defined GPCR targets in the brain [151,152].
Precision targeting may be further supported by biomarker-guided strategies. Circulating GPCR ligands or cell-specific receptor expression patterns, such as A2AR levels on peripheral blood mononuclear cells, have shown promise as indicators of disease state and therapeutic responsiveness [153,154]. In addition, AI-assisted ligand screening and computational modeling are accelerating the discovery and optimization of selective GPCR modulators with improved pharmacokinetic and safety profiles [155].
Integrating these advances with human-relevant disease models and temporal knowledge of GPCR signaling may enable selective engagement of beneficial GPCR pathways while avoiding adverse vascular and inflammatory consequences. Such precision-oriented strategies are likely essential for translating GPCR biology into effective therapies for cerebrovascular diseases.

Author Contributions

Conceptualization, Q.G.; validation, Q.G. and J.S.; formal analysis, Q.G. and J.S.; investigation, Q.G. and J.S.; writing—original draft preparation, Q.G.; writing—review and editing, Q.G., J.Y. and D.L.; supervision, D.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Wu, J.W.; Wang, B.X.; Shen, L.P.; Chen, Y.L.; Du, Z.Y.; Du, S.Q.; Lu, X.J.; Zhao, X.D. Investigating the Potential Therapeutic Targeting of the JAK-STAT Pathway in Cerebrovascular Diseases: Opportunities and Challenges. Mol. Neurobiol. 2025, 62, 9338–9364. [Google Scholar] [CrossRef]
  2. Chen, Q.; Yu, Y.; Zhu, L.; Duan, X.; Zhong, Y.; Li, C.; Wu, W. Recent progress of carbon dots in the theranostics of cardiovascular and cerebrovascular diseases. Nanoscale 2025, 17, 14521–14557. [Google Scholar] [CrossRef]
  3. Yin, X.; Li, S.; Wang, J.; Wang, M.; Yang, J. Research progress of active compounds from traditional Chinese medicine in the treatment of stroke. Eur. J. Med. Chem. 2025, 291, 117599. [Google Scholar] [CrossRef]
  4. Hu, S.; Wang, D.; Liu, W.; Wang, Y.; Chen, J.; Cai, X. Apelin receptor dimer: Classification, future prospects, and pathophysiological perspectives. Biochim. Biophys. Acta Mol. Basis Dis. 2024, 1870, 167257. [Google Scholar] [CrossRef]
  5. Wang, Y.; Zhu, C.L.; Li, P.; Liu, Q.; Li, H.R.; Yu, C.M.; Deng, X.M.; Wang, J.F. The role of G protein-coupled receptor in neutrophil dysfunction during sepsis-induced acute respiratory distress syndrome. Front. Immunol. 2023, 14, 1112196. [Google Scholar] [CrossRef] [PubMed]
  6. Annamneedi, A.; Gora, C.; Dudas, A.; Leray, X.; Bozon, V.; Crepieux, P.; Pellissier, L.P. Towards the convergent therapeutic potential of G protein-coupled receptors in autism spectrum disorders. Br. J. Pharmacol. 2025, 182, 3044–3067. [Google Scholar] [CrossRef]
  7. Shao, Y.; Mei, Y.; Tan, Y.; Yang, M.; Wu, H. The regulatory functions of G protein-coupled receptors signaling pathways in B cell differentiation and development contributing to autoimmune diseases. Cell Biosci. 2025, 15, 57. [Google Scholar] [CrossRef]
  8. Chen, S.; Niu, Z.; Shen, Y.; Lu, W.; Zhao, J.; Yang, H.; Guo, M.; Zhang, L.; Zheng, R.; Du, G.; et al. Naodesheng decoction regulating vascular function via G-protein-coupled receptors: Network analysis and experimental investigations. Front. Pharmacol. 2024, 15, 1355169. [Google Scholar] [CrossRef] [PubMed]
  9. Wang, B.; Wang, M.; Ao, D.; Wei, X. CXCL13-CXCR5 axis: Regulation in inflammatory diseases and cancer. Biochim. Biophys. Acta Rev. Cancer 2022, 1877, 188799. [Google Scholar] [CrossRef]
  10. Oz-Arslan, D.; Yavuz, M.; Kan, B. Exploring orphan GPCRs in neurodegenerative diseases. Front. Pharmacol. 2024, 15, 1394516. [Google Scholar] [CrossRef] [PubMed]
  11. Vessey, D.A.; Li, L.; Imhof, I.; Honbo, N.; Karliner, J.S. FTY720 postconditions isolated perfused heart by a mechanism independent of sphingosine kinase 2 and different from S1P or ischemic postconditioning. Med. Sci. Monit. Basic. Res. 2013, 19, 126–132. [Google Scholar] [CrossRef]
  12. Ricart-Ortega, M.; Font, J.; Llebaria, A. GPCR photopharmacology. Mol. Cell Endocrinol. 2019, 488, 36–51. [Google Scholar] [CrossRef] [PubMed]
  13. Hussain, M.; Liu, J.; Wang, G.Z.; Zhou, G.B. CXCL13 Signaling in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2021, 1302, 71–90. [Google Scholar] [CrossRef]
  14. Janicot, R.; Maziarz, M.; Park, J.C.; Zhao, J.; Luebbers, A.; Green, E.; Philibert, C.E.; Zhang, H.; Layne, M.D.; Wu, J.C.; et al. Direct interrogation of context-dependent GPCR activity with a universal biosensor platform. Cell 2024, 187, 1527–1546 e1525. [Google Scholar] [CrossRef]
  15. Mazarura, G.R.; Dallagnol, J.C.C.; Chatenet, D.; Allen, B.G.; Hebert, T.E. The complicated lives of GPCRs in cardiac fibroblasts. Am. J. Physiol. Cell Physiol. 2022, 323, C813–C822. [Google Scholar] [CrossRef]
  16. Schaeffer, S.; Iadecola, C. Revisiting the neurovascular unit. Nat. Neurosci. 2021, 24, 1198–1209. [Google Scholar] [CrossRef]
  17. Liu, S.; Anderson, P.J.; Rajagopal, S.; Lefkowitz, R.J.; Rockman, H.A. G Protein-Coupled Receptors: A Century of Research and Discovery. Circ. Res. 2024, 135, 174–197. [Google Scholar] [CrossRef]
  18. Hilger, D.; Masureel, M.; Kobilka, B.K. Structure and dynamics of GPCR signaling complexes. Nat. Struct. Mol. Biol. 2018, 25, 4–12. [Google Scholar] [CrossRef] [PubMed]
  19. Sprang, S.R.; Chen, Z.; Du, X. Structural basis of effector regulation and signal termination in heterotrimeric Galpha proteins. Adv. Protein Chem. 2007, 74, 1–65. [Google Scholar] [CrossRef] [PubMed]
  20. Kamato, D.; Thach, L.; Bernard, R.; Chan, V.; Zheng, W.; Kaur, H.; Brimble, M.; Osman, N.; Little, P.J. Structure, Function, Pharmacology, and Therapeutic Potential of the G Protein, Galpha/q,11. Front. Cardiovasc. Med. 2015, 2, 14. [Google Scholar] [CrossRef]
  21. Suzuki, N.; Hajicek, N.; Kozasa, T. Regulation and physiological functions of G12/13-mediated signaling pathways. Neurosignals 2009, 17, 55–70. [Google Scholar] [CrossRef]
  22. Kelly, P.; Casey, P.J.; Meigs, T.E. Biologic functions of the G12 subfamily of heterotrimeric g proteins: Growth, migration, and metastasis. Biochemistry 2007, 46, 6677–6687. [Google Scholar] [CrossRef]
  23. Halls, M.L.; Cooper, D.M. Regulation by Ca2+-signaling pathways of adenylyl cyclases. Cold Spring Harb. Perspect. Biol. 2011, 3, a004143. [Google Scholar] [CrossRef] [PubMed]
  24. Yen, Y.C.; Li, Y.; Chen, C.L.; Klose, T.; Watts, V.J.; Dessauer, C.W.; Tesmer, J.J.G. Structure of adenylyl cyclase 5 in complex with Gbetagamma offers insights into ADCY5-related dyskinesia. Nat. Struct. Mol. Biol. 2024, 31, 1189–1197. [Google Scholar] [CrossRef]
  25. Giri, L.; Patel, A.K.; Karunarathne, W.K.; Kalyanaraman, V.; Venkatesh, K.V.; Gautam, N. A G-protein subunit translocation embedded network motif underlies GPCR regulation of calcium oscillations. Biophys. J. 2014, 107, 242–254. [Google Scholar] [CrossRef] [PubMed]
  26. Ubeysinghe, S.; Wijayaratna, D.; Kankanamge, D.; Karunarathne, A. Molecular regulation of PLCbeta signaling. Methods Enzymol. 2023, 682, 17–52. [Google Scholar] [CrossRef] [PubMed]
  27. Whorton, M.R.; MacKinnon, R. X-ray structure of the mammalian GIRK2-betagamma G-protein complex. Nature 2013, 498, 190–197. [Google Scholar] [CrossRef]
  28. Wang, W.; Touhara, K.K.; Weir, K.; Bean, B.P.; MacKinnon, R. Cooperative regulation by G proteins and Na(+) of neuronal GIRK2 K(+) channels. elife 2016, 5, e15751. [Google Scholar] [CrossRef]
  29. Behrendt, M.; Gruss, F.; Enzeroth, R.; Dembla, S.; Zhao, S.; Crassous, P.A.; Mohr, F.; Nys, M.; Louros, N.; Gallardo, R.; et al. The structural basis for an on-off switch controlling Gbetagamma-mediated inhibition of TRPM3 channels. Proc. Natl. Acad. Sci. USA 2020, 117, 29090–29100. [Google Scholar] [CrossRef]
  30. Zhao, C.; MacKinnon, R. Structural and functional analyses of a GPCR-inhibited ion channel TRPM3. Neuron 2023, 111, 81–91. e7. [Google Scholar] [CrossRef]
  31. Toussay, X.; Tiberi, M.; Lacoste, B. Laser Doppler Flowmetry to Study the Regulation of Cerebral Blood Flow by G Protein-Coupled Receptors in Rodents. Methods Mol. Biol. 2019, 1947, 377–387. [Google Scholar] [CrossRef]
  32. Akhter, N.; Lambay, A.; Almotairi, R.; Hamadi, A.; Bhatia, K.; Ahmad, S.; Ducruet, A.F. The Complex Role of the Complement C3a Receptor (C3aR) in Cerebral Injury and Recovery Following Ischemic Stroke. Cells 2025, 14, 1440. [Google Scholar] [CrossRef]
  33. Rada, C.C.; Mejia-Pena, H.; Grimsey, N.J.; Canto Cordova, I.; Olson, J.; Wozniak, J.M.; Gonzalez, D.J.; Nizet, V.; Trejo, J. Heat shock protein 27 activity is linked to endothelial barrier recovery after proinflammatory GPCR-induced disruption. Sci. Signal 2021, 14, eabc1044. [Google Scholar] [CrossRef]
  34. Ma, Y.; Zheng, K.; Zhao, C.; Chen, J.; Chen, L.; Zhang, Y.; Chen, T.; Yao, X.; Cai, Y.; Wu, J. Microglia LILRB4 upregulation reduces brain damage after acute ischemic stroke by limiting CD8(+) T cell recruitment. J. Neuroinflammation 2024, 21, 214. [Google Scholar] [CrossRef]
  35. Jayaraj, R.L.; Azimullah, S.; Beiram, R.; Jalal, F.Y.; Rosenberg, G.A. Neuroinflammation: Friend and foe for ischemic stroke. J. Neuroinflammation 2019, 16, 142. [Google Scholar] [CrossRef]
  36. Khan, H.; Kaur, P.; Singh, T.G.; Grewal, A.K.; Sood, S. Adenosine as a Key Mediator of Neuronal Survival in Cerebral Ischemic Injury. Neurochem. Res. 2022, 47, 3543–3555. [Google Scholar] [CrossRef]
  37. Garcia, J.G.; Liu, F.; Verin, A.D.; Birukova, A.; Dechert, M.A.; Gerthoffer, W.T.; Bamberg, J.R.; English, D. Sphingosine 1-phosphate promotes endothelial cell barrier integrity by Edg-dependent cytoskeletal rearrangement. J. Clin. Investig. 2001, 108, 689–701. [Google Scholar] [CrossRef] [PubMed]
  38. Thangada, S.; Shapiro, L.H.; Silva, C.; Yamase, H.; Hla, T.; Ferrer, F.A. Treatment with the immunomodulator FTY720 (fingolimod) significantly reduces renal inflammation in murine unilateral ureteral obstruction. J. Urol. 2014, 191, 1508–1516. [Google Scholar] [CrossRef] [PubMed]
  39. Li, W.; Wu, S.; Hickey, R.W.; Rose, M.E.; Chen, J.; Graham, S.H. Neuronal cyclooxygenase-2 activity and prostaglandins PGE2, PGD2, and PGF2 alpha exacerbate hypoxic neuronal injury in neuron-enriched primary culture. Neurochem. Res. 2008, 33, 490–499. [Google Scholar] [CrossRef] [PubMed]
  40. Kostandy, B.B. The role of glutamate in neuronal ischemic injury: The role of spark in fire. Neurol. Sci. 2012, 33, 223–237. [Google Scholar] [CrossRef]
  41. Fung, S.; Cherry, A.E.; Xu, C.; Stella, N. Alkylindole-sensitive receptors modulate microglial cell migration and proliferation. Glia 2015, 63, 1797–1808. [Google Scholar] [CrossRef] [PubMed]
  42. McHugh, D.; Hu, S.S.; Rimmerman, N.; Juknat, A.; Vogel, Z.; Walker, J.M.; Bradshaw, H.B. N-arachidonoyl glycine, an abundant endogenous lipid, potently drives directed cellular migration through GPR18, the putative abnormal cannabidiol receptor. BMC Neurosci. 2010, 11, 44. [Google Scholar] [CrossRef] [PubMed]
  43. Gu, C.; Chen, Y.; Chen, Y.; Liu, C.F.; Zhu, Z.; Wang, M. Role of G Protein-Coupled Receptors in Microglial Activation: Implication in Parkinson’s Disease. Front. Aging Neurosci. 2021, 13, 768156. [Google Scholar] [CrossRef]
  44. Reyes-Gibby, C.C.; Yuan, C.; Wang, J.; Yeung, S.C.; Shete, S. Gene network analysis shows immune-signaling and ERK1/2 as novel genetic markers for multiple addiction phenotypes: Alcohol, smoking and opioid addiction. BMC Syst. Biol. 2015, 9, 25. [Google Scholar] [CrossRef] [PubMed]
  45. Yang, L.V.; Oppelt, K.A.; Thomassen, M.J.; Marie, M.A.; Nik Akhtar, S.; McCallen, J.D. Can GPR4 Be a Potential Therapeutic Target for COVID-19? Front. Med. 2020, 7, 626796. [Google Scholar] [CrossRef]
  46. Negri, S.; Faris, P.; Maniezzi, C.; Pellavio, G.; Spaiardi, P.; Botta, L.; Laforenza, U.; Biella, G.; Moccia, D.F. NMDA receptors elicit flux-independent intracellular Ca(2+) signals via metabotropic glutamate receptors and flux-dependent nitric oxide release in human brain microvascular endothelial cells. Cell Calcium 2021, 99, 102454. [Google Scholar] [CrossRef]
  47. Kuhr, F.; Lowry, J.; Zhang, Y.; Brovkovych, V.; Skidgel, R.A. Differential regulation of inducible and endothelial nitric oxide synthase by kinin B1 and B2 receptors. Neuropeptides 2010, 44, 145–154. [Google Scholar] [CrossRef]
  48. Jiang, W.; Jin, Y.; Zhang, S.; Ding, Y.; Huo, K.; Yang, J.; Zhao, L.; Nian, B.; Zhong, T.P.; Lu, W.; et al. PGE2 activates EP4 in subchondral bone osteoclasts to regulate osteoarthritis. Bone Res. 2022, 10, 27. [Google Scholar] [CrossRef]
  49. McCullough, L.; Wu, L.; Haughey, N.; Liang, X.; Hand, T.; Wang, Q.; Breyer, R.M.; Andreasson, K. Neuroprotective function of the PGE2 EP2 receptor in cerebral ischemia. J. Neurosci. 2004, 24, 257–268. [Google Scholar] [CrossRef]
  50. Lacroix, A.; Toussay, X.; Anenberg, E.; Lecrux, C.; Ferreiros, N.; Karagiannis, A.; Plaisier, F.; Chausson, P.; Jarlier, F.; Burgess, S.A.; et al. COX-2-Derived Prostaglandin E2 Produced by Pyramidal Neurons Contributes to Neurovascular Coupling in the Rodent Cerebral Cortex. J. Neurosci. 2015, 35, 11791–11810. [Google Scholar] [CrossRef]
  51. Liu, X.; Deng, F.; Yu, Z.; Xie, Y.; Hu, C.; Chen, L. Inhibition of endothelin A receptor protects brain microvascular endothelial cells against hypoxia-induced injury. Int. J. Mol. Med. 2014, 34, 313–320. [Google Scholar] [CrossRef]
  52. Bearzi, P.; Navarini, L.; Currado, D.; Marino, A.; Minerba, M.; Salvolini, C.; Perrone, A.; Frasca, L.; Liakouli, V.; Vomero, M.; et al. Bosentan effect on echocardiographic systolic pulmonary arterial pressure in systemic sclerosis-related pulmonary hypertension: A systematic review and metanalysis. Clin. Exp. Rheumatol. 2024, 42, 1615–1622. [Google Scholar] [CrossRef]
  53. Davenport, A.P.; Hyndman, K.A.; Dhaun, N.; Southan, C.; Kohan, D.E.; Pollock, J.S.; Pollock, D.M.; Webb, D.J.; Maguire, J.J. Endothelin. Pharmacol. Rev. 2016, 68, 357–418. [Google Scholar] [CrossRef]
  54. Hennenberg, M.; Trebicka, J.; Biecker, E.; Schepke, M.; Sauerbruch, T.; Heller, J. Vascular dysfunction in human and rat cirrhosis: Role of receptor-desensitizing and calcium-sensitizing proteins. Hepatology 2007, 45, 495–506. [Google Scholar] [CrossRef]
  55. Sauge, E.; Pechkovsky, D.; Atmuri, N.D.P.; Tehrani, A.Y.; White, Z.; Dong, Y.; Cait, J.; Hughes, M.; Tam, A.; Donen, G.; et al. Losartan metabolite EXP3179 is a unique blood pressure-lowering AT1R antagonist with direct, rapid endothelium-dependent vasoactive properties. Vascul Pharmacol. 2022, 147, 107112. [Google Scholar] [CrossRef] [PubMed]
  56. Tobo, M.; Tomura, H.; Mogi, C.; Wang, J.Q.; Liu, J.P.; Komachi, M.; Damirin, A.; Kimura, T.; Murata, N.; Kurose, H.; et al. Previously postulated “ligand-independent” signaling of GPR4 is mediated through proton-sensing mechanisms. Cell Signal. 2007, 19, 1745–1753. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, A.; Dong, L.; Leffler, N.R.; Asch, A.S.; Witte, O.N.; Yang, L.V. Activation of GPR4 by acidosis increases endothelial cell adhesion through the cAMP/Epac pathway. PLoS ONE 2011, 6, e27586. [Google Scholar] [CrossRef]
  58. Xu, D.; Gao, Q.; Wang, F.; Peng, Q.; Wang, G.; Wei, Q.; Lei, S.; Zhao, S.; Zhang, L.; Guo, F. Sphingosine-1-phosphate receptor 3 is implicated in BBB injury via the CCL2-CCR2 axis following acute intracerebral hemorrhage. CNS Neurosci. Ther. 2021, 27, 674–686. [Google Scholar] [CrossRef]
  59. Blaho, V.A.; Hla, T. Regulation of mammalian physiology, development, and disease by the sphingosine 1-phosphate and lysophosphatidic acid receptors. Chem. Rev. 2011, 111, 6299–6320. [Google Scholar] [CrossRef]
  60. Vahidinia, Z.; Joghataei, M.T.; Beyer, C.; Karimian, M.; Tameh, A.A. G-Protein-Coupled Receptors and Ischemic Stroke: A Focus on Molecular Function and Therapeutic Potential. Mol. Neurobiol. 2021, 58, 4588–4614. [Google Scholar] [CrossRef] [PubMed]
  61. Rabbani, P.; Ramkhelawon, B.; Cronstein, B.N. Adenosine metabolism and receptors in aging of the skin, musculoskeletal, immune and cardiovascular systems. Ageing Res. Rev. 2025, 106, 102695. [Google Scholar] [CrossRef] [PubMed]
  62. Goncalves, F.Q.; Valada, P.; Matos, M.; Cunha, R.A.; Tome, A.R. Feedback facilitation by adenosine A(2A) receptors of ATP release from mouse hippocampal nerve terminals. Purinergic Signal. 2024, 20, 247–255. [Google Scholar] [CrossRef]
  63. Lin, S.; Landon, B.; Zhang, H.; Jin, K. Pericyte Dysfunction Contributes to Vascular Cognitive Impairment Induced by Chronic Cerebral Hypoperfusion in Rats. Aging Dis. 2024, 15, 1357–1372. [Google Scholar] [CrossRef]
  64. Yamamoto, M.; Guo, D.H.; Hernandez, C.M.; Stranahan, A.M. Endothelial Adora2a Activation Promotes Blood-Brain Barrier Breakdown and Cognitive Impairment in Mice with Diet-Induced Insulin Resistance. J. Neurosci. 2019, 39, 4179–4192. [Google Scholar] [CrossRef]
  65. Cheng, C.; Cheng, W.; Wang, Y.; Chen, X.; Zhang, L.; Li, Y.; Shen, F.; Yuan, D.; Hong, P.; Huang, W. The Dual Role of A2aR in Neuroinflammation: Modulating Microglial Polarization in White Matter Lesions. eNeuro 2025, 12, 1–21. [Google Scholar] [CrossRef]
  66. Carr, R., 3rd; Schilling, J.; Song, J.; Carter, R.L.; Du, Y.; Yoo, S.M.; Traynham, C.J.; Koch, W.J.; Cheung, J.Y.; Tilley, D.G.; et al. beta-arrestin-biased signaling through the beta2-adrenergic receptor promotes cardiomyocyte contraction. Proc. Natl. Acad. Sci. USA 2016, 113, E4107–E4116. [Google Scholar] [CrossRef] [PubMed]
  67. Bhattacharya, S.; Hall, S.E.; Li, H.; Vaidehi, N. Ligand-stabilized conformational states of human beta(2) adrenergic receptor: Insight into G-protein-coupled receptor activation. Biophys. J. 2008, 94, 2027–2042. [Google Scholar] [CrossRef] [PubMed]
  68. Li, Y.J.; Shi, S.X.; Liu, Q.; Shi, F.D.; Gonzales, R.J. Targeted role for sphingosine-1-phosphate receptor 1 in cerebrovascular integrity and inflammation during acute ischemic stroke. Neurosci. Lett. 2020, 735, 135160. [Google Scholar] [CrossRef]
  69. Alves, N.G.; Yuan, S.Y.; Breslin, J.W. Sphingosine-1-phosphate protects against brain microvascular endothelial junctional protein disorganization and barrier dysfunction caused by alcohol. Microcirculation 2019, 26, e12506. [Google Scholar] [CrossRef]
  70. Zhu, H.; Cao, Y.; Chen, X.; Xu, N.; Zhou, Y.; Yang, P.; Xu, F.; Ji, J. CX3CL1 attenuates neurological deficit and neuroinflammation through CX3CR1/p38 MAPK/ERK1/2 signaling pathway in traumatic brain injury. Int. Immunopharmacol. 2026, 168, 115765. [Google Scholar] [CrossRef]
  71. Reglodi, D.; Tamas, A.; Jungling, A.; Vaczy, A.; Rivnyak, A.; Fulop, B.D.; Szabo, E.; Lubics, A.; Atlasz, T. Protective effects of pituitary adenylate cyclase activating polypeptide against neurotoxic agents. Neurotoxicology 2018, 66, 185–194. [Google Scholar] [CrossRef]
  72. Wu, Y.; Li, Z.; Ding, T.; Yang, Y.; Wei, C.; Zhang, S.; Fan, X. Bidirectional regulation of neuronal autophagy in ischemic stroke: Mechanisms and therapeutic potential. Ageing Res. Rev. 2025, 111, 102842. [Google Scholar] [CrossRef] [PubMed]
  73. Zhu, W.M.; Neuhaus, A.; Beard, D.J.; Sutherland, B.A.; DeLuca, G.C. Neurovascular coupling mechanisms in health and neurovascular uncoupling in Alzheimer’s disease. Brain 2022, 145, 2276–2292. [Google Scholar] [CrossRef]
  74. Stenman, E.; Malmsjo, M.; Uddman, E.; Gido, G.; Wieloch, T.; Edvinsson, L. Cerebral ischemia upregulates vascular endothelin ET(B) receptors in rat. Stroke 2002, 33, 2311–2316. [Google Scholar] [CrossRef]
  75. Edvinsson, L. Cerebrovascular endothelin receptor upregulation in cerebral ischemia. Curr. Vasc. Pharmacol. 2009, 7, 26–33. [Google Scholar] [CrossRef]
  76. Cai, L.; Gong, Q.; Qi, L.; Xu, T.; Suo, Q.; Li, X.; Wang, W.; Jing, Y.; Yang, D.; Xu, Z.; et al. ACT001 attenuates microglia-mediated neuroinflammation after traumatic brain injury via inhibiting AKT/NFkappaB/NLRP3 pathway. Cell Commun. Signal. 2022, 20, 56. [Google Scholar] [CrossRef]
  77. Kumar, A.; Stoica, B.A.; Loane, D.J.; Yang, M.; Abulwerdi, G.; Khan, N.; Kumar, A.; Thom, S.R.; Faden, A.I. Microglial-derived microparticles mediate neuroinflammation after traumatic brain injury. J. Neuroinflammation 2017, 14, 47. [Google Scholar] [CrossRef]
  78. Lattanzi, R.; Miele, R. Versatile Role of Prokineticins and Prokineticin Receptors in Neuroinflammation. Biomedicines 2021, 9, 1648. [Google Scholar] [CrossRef]
  79. Yang, Y.; Rosenberg, G.A. Blood-brain barrier breakdown in acute and chronic cerebrovascular disease. Stroke 2011, 42, 3323–3328. [Google Scholar] [CrossRef] [PubMed]
  80. De Bock, M.; Wang, N.; Decrock, E.; Bol, M.; Gadicherla, A.K.; Culot, M.; Cecchelli, R.; Bultynck, G.; Leybaert, L. Endothelial calcium dynamics, connexin channels and blood-brain barrier function. Prog. Neurobiol. 2013, 108, 1–20. [Google Scholar] [CrossRef] [PubMed]
  81. Williams, J.; Bogwu, J.; Oyekan, A. The role of the RhoA/Rho-kinase signaling pathway in renal vascular reactivity in endothelial nitric oxide synthase null mice. J. Hypertens. 2006, 24, 1429–1436. [Google Scholar] [CrossRef]
  82. Masago, K.; Kihara, Y.; Yanagida, K.; Hamano, F.; Nakagawa, S.; Niwa, M.; Shimizu, T. Lysophosphatidic acid receptor, LPA(6), regulates endothelial blood-brain barrier function: Implication for hepatic encephalopathy. Biochem. Biophys. Res. Commun. 2018, 501, 1048–1054. [Google Scholar] [CrossRef]
  83. Ji, B.; Cheng, B.; Pan, Y.; Wang, C.; Chen, J.; Bai, B. Neuroprotection of bradykinin/bradykinin B2 receptor system in cerebral ischemia. Biomed. Pharmacother. 2017, 94, 1057–1063. [Google Scholar] [CrossRef]
  84. Panossian, A.; Lemerond, T.; Efferth, T. Adaptogens in Long-Lasting Brain Fatigue: An Insight from Systems Biology and Network Pharmacology. Pharmaceuticals 2025, 18, 261. [Google Scholar] [CrossRef] [PubMed]
  85. McCrary, M.R.; Jiang, M.Q.; Giddens, M.M.; Zhang, J.Y.; Owino, S.; Wei, Z.Z.; Zhong, W.; Gu, X.; Xin, H.; Hall, R.A.; et al. Protective effects of GPR37 via regulation of inflammation and multiple cell death pathways after ischemic stroke in mice. FASEB J. 2019, 33, 10680–10691. [Google Scholar] [CrossRef] [PubMed]
  86. Ma, X.D.; Hang, L.H.; Shao, D.H.; Shu, W.W.; Hu, X.L.; Luo, H. TDAG8 activation attenuates cerebral ischaemia-reperfusion injury via Akt signalling in rats. Exp. Neurol. 2017, 293, 115–123. [Google Scholar] [CrossRef] [PubMed]
  87. Wang, T.; Zhou, G.; He, M.; Xu, Y.; Rusyniak, W.G.; Xu, Y.; Ji, Y.; Simon, R.P.; Xiong, Z.G.; Zha, X.M. GPR68 Is a Neuroprotective Proton Receptor in Brain Ischemia. Stroke 2020, 51, 3690–3700. [Google Scholar] [CrossRef]
  88. Villanueva, M.T. Repurposing CCR5 inhibitors for stroke recovery. Nat. Rev. Drug Discov. 2019, 18, 253. [Google Scholar] [CrossRef]
  89. Wu, Q.L.; Cui, L.Y.; Ma, W.Y.; Wang, S.S.; Zhang, Z.; Feng, Z.P.; Sun, H.S.; Chu, S.F.; He, W.B.; Chen, N.H. A novel small-molecular CCR5 antagonist promotes neural repair after stroke. Acta Pharmacol. Sin. 2023, 44, 1935–1947. [Google Scholar] [CrossRef]
  90. Gupte, R.P.; Kadunganattil, S.; Shepherd, A.J.; Merrill, R.; Planer, W.; Bruchas, M.R.; Strack, S.; Mohapatra, D.P. Convergent phosphomodulation of the major neuronal dendritic potassium channel Kv4.2 by pituitary adenylate cyclase-activating polypeptide. Neuropharmacology 2016, 101, 291–308. [Google Scholar] [CrossRef]
  91. White, R.E.; Palm, C.; Xu, L.; Ling, E.; Ginsburg, M.; Daigle, B.J.; Han, R.; Patterson, A.; Altman, R.B.; Giffard, R.G. Mice lacking the beta2 adrenergic receptor have a unique genetic profile before and after focal brain ischaemia. ASN Neuro 2012, 4, AN20110020. [Google Scholar] [CrossRef]
  92. Chen, H.; Yuan, M.; Huang, C.; Xu, Z.; Li, M.; Zhang, C.; Gao, Z.; Zhang, M.; Xu, J.; Qian, H.; et al. Endothelial Cell Inflammation and Barriers Are Regulated by the Rab26-Mediated Balance between beta2-AR and TLR4 in Pulmonary Microvessel Endothelial Cells. Mediators Inflamm. 2019, 2019, 7538071. [Google Scholar] [CrossRef]
  93. Lin, B.; Zhou, Y.; Huang, Z.; Ma, M.; Qi, M.; Jiang, Z.; Li, G.; Xu, Y.; Yan, J.; Wang, D.; et al. GPR34 senses demyelination to promote neuroinflammation and pathologies. Cell Mol. Immunol. 2024, 21, 1131–1144. [Google Scholar] [CrossRef]
  94. Nasu-Tada, K.; Koizumi, S.; Inoue, K. Involvement of beta1 integrin in microglial chemotaxis and proliferation on fibronectin: Different regulations by ADP through PKA. Glia 2005, 52, 98–107. [Google Scholar] [CrossRef]
  95. Tsigkou, V.; Siasos, G.; Rovos, K.; Tripyla, N.; Tousoulis, D. Peripheral artery disease and antiplatelet treatment. Curr. Opin. Pharmacol. 2018, 39, 43–52. [Google Scholar] [CrossRef] [PubMed]
  96. Cattaneo, M. P2Y12 receptors: Structure and function. J. Thromb. Haemost. 2015, 13, S10–S16. [Google Scholar] [CrossRef] [PubMed]
  97. Hansen, L.; Lohfink, N.; Vutukuri, R.; Kestner, R.I.; Trautmann, S.; Hecht, M.; Wagner, P.V.; Spitzer, D.; Khel, M.I.; Macas, J.; et al. Endothelial Sphingosine-1-Phosphate Receptor 4 Regulates Blood-Brain Barrier Permeability and Promotes a Homeostatic Endothelial Phenotype. J. Neurosci. 2022, 42, 1908–1929. [Google Scholar] [CrossRef] [PubMed]
  98. Bierwirth, J.; Tan, J.B.; Mendez Padilla, B.D.; Zhang, L.; Schroeder, H.; Liu, T.; Power, G.G.; Blood, A.B. Role of alpha1 adrenergic receptors in the cerebral cortical blood flow response to acute hypoxia in low- and high-altitude near-term fetal lambs. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2025, 328, R364–R373. [Google Scholar] [CrossRef]
  99. Hein, L. Adrenoceptors and signal transduction in neurons. Cell Tissue Res. 2006, 326, 541–551. [Google Scholar] [CrossRef]
  100. Garcia, V.; Gilani, A.; Shkolnik, B.; Pandey, V.; Zhang, F.F.; Dakarapu, R.; Gandham, S.K.; Reddy, N.R.; Graves, J.P.; Gruzdev, A.; et al. 20-HETE Signals Through G-Protein-Coupled Receptor GPR75 (G(q)) to Affect Vascular Function and Trigger Hypertension. Circ. Res. 2017, 120, 1776–1788. [Google Scholar] [CrossRef]
  101. Feldman, R.D.; Gros, R. Vascular effects of aldosterone: Sorting out the receptors and the ligands. Clin. Exp. Pharmacol. Physiol. 2013, 40, 916–921. [Google Scholar] [CrossRef] [PubMed]
  102. Thomas, P.; Alyea, R.; Pang, Y.; Peyton, C.; Dong, J.; Berg, A.H. Conserved estrogen binding and signaling functions of the G protein-coupled estrogen receptor 1 (GPER) in mammals and fish. Steroids 2010, 75, 595–602. [Google Scholar] [CrossRef] [PubMed]
  103. Wang, T.; Shao, J.; Kumar, S.; Alnouri, M.W.; Carvalho, J.; Gunther, S.; Krasel, C.; Murphy, K.T.; Bunemann, M.; Offermanns, S.; et al. Orphan GPCR GPRC5C Facilitates Angiotensin II-Induced Smooth Muscle Contraction. Circ. Res. 2024, 134, 1259–1275. [Google Scholar] [CrossRef]
  104. Freundt, G.V.; von Samson-Himmelstjerna, F.A.; Nitz, J.T.; Luedde, M.; Waltenberger, J.; Wieland, T.; Frey, N.; Preusch, M.; Hippe, H.J. The orphan receptor GPRC5B activates pro-inflammatory signaling in the vascular wall via Fyn and NFkappaB. Biochem. Biophys. Res. Commun. 2022, 592, 60–66. [Google Scholar] [CrossRef]
  105. Albee, L.J.; Gao, X.; Majetschak, M. Plasticity of seven-transmembrane-helix receptor heteromers in human vascular smooth muscle cells. PLoS ONE 2021, 16, e0253821. [Google Scholar] [CrossRef] [PubMed]
  106. Tan, X.; Sanders, P.; Bolado, J., Jr.; Whitney, M. Integration of G-protein coupled receptor signaling pathways for activation of a transcription factor (EGR-3). Genom. Proteom. Bioinform. 2003, 1, 173–179. [Google Scholar] [CrossRef]
  107. Chen, G.; Jin, Z.; Wang, X.; Yu, Q.H.; Hu, G.B. Danshen injection mitigated the cerebral ischemia/reperfusion injury by suppressing neuroinflammation via the HIF-1alpha/CXCR4/NF-kappaB signaling pathway. Neuroreport 2024, 35, 601–611. [Google Scholar] [CrossRef]
  108. Ofek, O.; Attar-Namdar, M.; Kram, V.; Dvir-Ginzberg, M.; Mechoulam, R.; Zimmer, A.; Frenkel, B.; Shohami, E.; Bab, I. CB2 cannabinoid receptor targets mitogenic Gi protein-cyclin D1 axis in osteoblasts. J. Bone Miner. Res. 2011, 26, 308–316. [Google Scholar] [CrossRef]
  109. Sanchez, T.; Skoura, A.; Wu, M.T.; Casserly, B.; Harrington, E.O.; Hla, T. Induction of vascular permeability by the sphingosine-1-phosphate receptor-2 (S1P2R) and its downstream effectors ROCK and PTEN. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 1312–1318. [Google Scholar] [CrossRef]
  110. Grimm, M.; Tischner, D.; Troidl, K.; Albarran Juarez, J.; Sivaraj, K.K.; Ferreiros Bouzas, N.; Geisslinger, G.; Binder, C.J.; Wettschureck, N. S1P2/G12/13 Signaling Negatively Regulates Macrophage Activation and Indirectly Shapes the Atheroprotective B1-Cell Population. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 37–48. [Google Scholar] [CrossRef]
  111. Saligrama, N.; Noubade, R.; Case, L.K.; del Rio, R.; Teuscher, C. Combinatorial roles for histamine H1-H2 and H3-H4 receptors in autoimmune inflammatory disease of the central nervous system. Eur. J. Immunol. 2012, 42, 1536–1546. [Google Scholar] [CrossRef]
  112. Wang, D.; Guo, Q.; Wu, Z.; Li, M.; He, B.; Du, Y.; Zhang, K.; Tao, Y. Molecular mechanism of antihistamines recognition and regulation of the histamine H(1) receptor. Nat. Commun. 2024, 15, 84. [Google Scholar] [CrossRef]
  113. Leurs, R.; Smit, M.J.; Timmerman, H. Molecular pharmacological aspects of histamine receptors. Pharmacol. Ther. 1995, 66, 413–463. [Google Scholar] [CrossRef]
  114. Karlstedt, K.; Jin, C.; Panula, P. Expression of histamine receptor genes Hrh3 and Hrh4 in rat brain endothelial cells. Br. J. Pharmacol. 2013, 170, 58–66. [Google Scholar] [CrossRef]
  115. Mogi, C.; Tobo, M.; Tomura, H.; Murata, N.; He, X.D.; Sato, K.; Kimura, T.; Ishizuka, T.; Sasaki, T.; Sato, T.; et al. Involvement of proton-sensing TDAG8 in extracellular acidification-induced inhibition of proinflammatory cytokine production in peritoneal macrophages. J. Immunol. 2009, 182, 3243–3251. [Google Scholar] [CrossRef]
  116. Meyer, R.C.; Giddens, M.M.; Schaefer, S.A.; Hall, R.A. GPR37 and GPR37L1 are receptors for the neuroprotective and glioprotective factors prosaptide and prosaposin. Proc. Natl. Acad. Sci. USA 2013, 110, 9529–9534. [Google Scholar] [CrossRef] [PubMed]
  117. Leeb-Lundberg, L.M.; Marceau, F.; Muller-Esterl, W.; Pettibone, D.J.; Zuraw, B.L. International union of pharmacology. XLV. Classification of the kinin receptor family: From molecular mechanisms to pathophysiological consequences. Pharmacol. Rev. 2005, 57, 27–77. [Google Scholar] [CrossRef]
  118. Reiss, A.B.; Grossfeld, D.; Kasselman, L.J.; Renna, H.A.; Vernice, N.A.; Drewes, W.; Konig, J.; Carsons, S.E.; DeLeon, J. Adenosine and the Cardiovascular System. Am. J. Cardiovasc. Drugs 2019, 19, 449–464. [Google Scholar] [CrossRef] [PubMed]
  119. Larsen, K.; Lindberg, U.; Ozenne, B.; McCulloch, D.E.; Armand, S.; Madsen, M.K.; Johansen, A.; Stenbaek, D.S.; Knudsen, G.M.; Fisher, P.M. Acute psilocybin and ketanserin effects on cerebral blood flow: 5-HT2AR neuromodulation in healthy humans. J. Cereb. Blood Flow. Metab. 2025, 45, 1385–1401. [Google Scholar] [CrossRef]
  120. Comer, M.B. Pharmacology of the selective 5-HT(1B/1D) agonist frovatriptan. Headache 2002, 42, S47–S53. [Google Scholar] [CrossRef] [PubMed]
  121. Razzaque, Z.; Heald, M.A.; Pickard, J.D.; Maskell, L.; Beer, M.S.; Hill, R.G.; Longmore, J. Vasoconstriction in human isolated middle meningeal arteries: Determining the contribution of 5-HT1B- and 5-HT1F-receptor activation. Br. J. Clin. Pharmacol. 1999, 47, 75–82. [Google Scholar] [CrossRef]
  122. Elhusseiny, A.; Hamel, E. Sumatriptan elicits both constriction and dilation in human and bovine brain intracortical arterioles. Br. J. Pharmacol. 2001, 132, 55–62. [Google Scholar] [CrossRef]
  123. Stockwell, J.; Jakova, E.; Cayabyab, F.S. Adenosine A1 and A2A Receptors in the Brain: Current Research and Their Role in Neurodegeneration. Molecules 2017, 22, 676. [Google Scholar] [CrossRef]
  124. Zhao, Y.; Zhou, Y.G.; Chen, J.F. Targeting the adenosine A(2A) receptor for neuroprotection and cognitive improvement in traumatic brain injury and Parkinson’s disease. Chin. J. Traumatol. 2024, 27, 125–133. [Google Scholar] [CrossRef]
  125. Merighi, S.; Travagli, A.; Nigro, M.; Pasquini, S.; Cappello, M.; Contri, C.; Varani, K.; Vincenzi, F.; Borea, P.A.; Gessi, S. Caffeine for Prevention of Alzheimer’s Disease: Is the A(2A) Adenosine Receptor Its Target? Biomolecules 2023, 13, 967. [Google Scholar] [CrossRef]
  126. Cosenzi, A. Enrasentan, an antagonist of endothelin receptors. Cardiovasc. Drug Rev. 2003, 21, 1–16. [Google Scholar] [CrossRef]
  127. Gonsalves, A.M.; Baker, S.E.; Jacob, D.W.; Harper, J.L.; Manrique-Acevedo, C.M.; Limberg, J.K. Effect of endothelin-1 on the blood pressure response to acute hypoxia and hyperoxia in healthy young men. Physiol. Rep. 2024, 12, e70004. [Google Scholar] [CrossRef] [PubMed]
  128. Binish, F.; Xiao, J. Deciphering the role of sphingosine 1-phosphate in central nervous system myelination and repair. J. Neurochem. 2025, 169, e16228. [Google Scholar] [CrossRef] [PubMed]
  129. Wang, Z.; Kawabori, M.; Houkin, K. FTY720 (Fingolimod) Ameliorates Brain Injury through Multiple Mechanisms and is a Strong Candidate for Stroke Treatment. Curr. Med. Chem. 2020, 27, 2979–2993. [Google Scholar] [CrossRef] [PubMed]
  130. Ping, S.; Qiu, X.; Kyle, M.; Zhao, L.R. Brain-derived CCR5 Contributes to Neuroprotection and Brain Repair after Experimental Stroke. Aging Dis. 2021, 12, 72–92. [Google Scholar] [CrossRef]
  131. Joy, M.T.; Ben Assayag, E.; Shabashov-Stone, D.; Liraz-Zaltsman, S.; Mazzitelli, J.; Arenas, M.; Abduljawad, N.; Kliper, E.; Korczyn, A.D.; Thareja, N.S.; et al. CCR5 Is a Therapeutic Target for Recovery after Stroke and Traumatic Brain Injury. Cell 2019, 176, 1143–1157 e1113. [Google Scholar] [CrossRef]
  132. Patterson, B.K.; Yogendra, R.; Guevara-Coto, J.; Mora-Rodriguez, R.A.; Osgood, E.; Bream, J.; Parikh, P.; Kreimer, M.; Jeffers, D.; Rutland, C.; et al. Case series: Maraviroc and pravastatin as a therapeutic option to treat long COVID/Post-acute sequelae of COVID (PASC). Front. Med. 2023, 10, 1122529. [Google Scholar] [CrossRef] [PubMed]
  133. Lorente, J.S.; Sokolov, A.V.; Ferguson, G.; Schioth, H.B.; Hauser, A.S.; Gloriam, D.E. GPCR drug discovery: New agents, targets and indications. Nat. Rev. Drug Discov. 2025, 24, 458–479. [Google Scholar] [CrossRef] [PubMed]
  134. Lemes Dos Santos Sanna, P.; Bernardes Carvalho, L.; Cristina Dos Santos Afonso, C.; de Carvalho, K.; Aires, R.; Souza, J.; Rodrigues Ferreira, M.; Birbrair, A.; Martha Bernardi, M.; Latini, A.; et al. Adora2A downregulation promotes caffeine neuroprotective effect against LPS-induced neuroinflammation in the hippocampus. Brain Res. 2024, 1833, 148866. [Google Scholar] [CrossRef]
  135. Company-Marin, I.; Gunner, J.; Poyner, D.; Simms, J.; Pitt, A.R.; Spickett, C.M. The effect of oxidative stress on the adenosine A(2A) receptor activity and signalling. Biochim. Biophys. Acta Biomembr. 2025, 1867, 184412. [Google Scholar] [CrossRef] [PubMed]
  136. Wala, K.; Szlasa, W.; Saczko, J.; Rudno-Rudzinska, J.; Kulbacka, J. Modulation of Blood-Brain Barrier Permeability by Activating Adenosine A2 Receptors in Oncological Treatment. Biomolecules 2021, 11, 633. [Google Scholar] [CrossRef]
  137. Folkesson, H.G.; Kuzenko, S.R.; Lipson, D.A.; Matthay, M.A.; Simmons, M.A. The adenosine 2A receptor agonist GW328267C improves lung function after acute lung injury in rats. Am. J. Physiol. Lung Cell Mol. Physiol. 2012, 303, L259–L271. [Google Scholar] [CrossRef]
  138. Yan, L.; Burbiel, J.C.; Maass, A.; Muller, C.E. Adenosine receptor agonists: From basic medicinal chemistry to clinical development. Expert. Opin. Emerg. Drugs 2003, 8, 537–576. [Google Scholar] [CrossRef] [PubMed]
  139. Davenport, A.P.; Scully, C.C.G.; de Graaf, C.; Brown, A.J.H.; Maguire, J.J. Advances in therapeutic peptides targeting G protein-coupled receptors. Nat. Rev. Drug Discov. 2020, 19, 389–413. [Google Scholar] [CrossRef]
  140. Bauer, B.; Hartz, A.M.; Fricker, G.; Miller, D.S. Modulation of p-glycoprotein transport function at the blood-brain barrier. Exp. Biol. Med. 2005, 230, 118–127. [Google Scholar] [CrossRef]
  141. Li, J.; Zheng, M.; Shimoni, O.; Banks, W.A.; Bush, A.I.; Gamble, J.R.; Shi, B. Development of Novel Therapeutics Targeting the Blood-Brain Barrier: From Barrier to Carrier. Adv. Sci. 2021, 8, e2101090. [Google Scholar] [CrossRef] [PubMed]
  142. McAuley, M.A.; Breu, V.; Graham, D.I.; McCulloch, J. The effects of bosentan on cerebral blood flow and histopathology following middle cerebral artery occlusion in the rat. Eur. J. Pharmacol. 1996, 307, 171–181. [Google Scholar] [CrossRef]
  143. Hanna, L.A.; Basalious, E.B.; ON, E.L. Respirable controlled release polymeric colloid (RCRPC) of bosentan for the management of pulmonary hypertension: In vitro aerosolization, histological examination and in vivo pulmonary absorption. Drug Deliv. 2016, 24, 188–198. [Google Scholar] [CrossRef] [PubMed]
  144. Macrae, I.M. Preclinical stroke research--advantages and disadvantages of the most common rodent models of focal ischaemia. Br. J. Pharmacol. 2011, 164, 1062–1078. [Google Scholar] [CrossRef]
  145. Stebbins, M.J.; Gastfriend, B.D.; Canfield, S.G.; Lee, M.S.; Richards, D.; Faubion, M.G.; Li, W.J.; Daneman, R.; Palecek, S.P.; Shusta, E.V. Human pluripotent stem cell-derived brain pericyte-like cells induce blood-brain barrier properties. Sci. Adv. 2019, 5, eaau7375. [Google Scholar] [CrossRef]
  146. Noorani, B.; Bhalerao, A.; Raut, S.; Nozohouri, E.; Bickel, U.; Cucullo, L. A Quasi-Physiological Microfluidic Blood-Brain Barrier Model for Brain Permeability Studies. Pharmaceutics 2021, 13, 1474. [Google Scholar] [CrossRef]
  147. Papamichail, L.; Koch, L.S.; Veerman, D.; Broersen, K.; van der Meer, A.D. Organoids-on-a-chip: Microfluidic technology enables culture of organoids with enhanced tissue function and potential for disease modeling. Front. Bioeng. Biotechnol. 2025, 13, 1515340. [Google Scholar] [CrossRef] [PubMed]
  148. Kaur, H.; Carvalho, J.; Looso, M.; Singh, P.; Chennupati, R.; Preussner, J.; Gunther, S.; Albarran-Juarez, J.; Tischner, D.; Classen, S.; et al. Author Correction: Single-cell profiling reveals heterogeneity and functional patterning of GPCR expression in the vascular system. Nat. Commun. 2019, 10, 1448. [Google Scholar] [CrossRef]
  149. Kogut-Gunthel, M.M.; Zara, Z.; Nicoli, A.; Steuer, A.; Lopez-Balastegui, M.; Selent, J.; Karanth, S.; Koehler, M.; Ciancetta, A.; Abiko, L.A.; et al. The path to the G protein-coupled receptor structural landscape: Major milestones and future directions. Br. J. Pharmacol. 2025, 182, 3225–3248. [Google Scholar] [CrossRef]
  150. Lopez-Balastegui, M.; Stepniewski, T.M.; Kogut-Gunthel, M.M.; Di Pizio, A.; Rosenkilde, M.M.; Mao, J.; Selent, J. Relevance of G protein-coupled receptor (GPCR) dynamics for receptor activation, signalling bias and allosteric modulation. Br. J. Pharmacol. 2025, 182, 3211–3224. [Google Scholar] [CrossRef]
  151. Yu, J.; Kumar, A.; Zhang, X.; Martin, C.; Van Holsbeeck, K.; Raia, P.; Koehl, A.; Laeremans, T.; Steyaert, J.; Manglik, A.; et al. Structural basis of mu-opioid receptor targeting by a nanobody antagonist. Nat. Commun. 2024, 15, 8687. [Google Scholar] [CrossRef]
  152. Stolwijk, J.A.; Sauer, L.; Ackermann, K.; Nassios, A.; Aung, T.; Haerteis, S.; Baumner, A.J.; Wegener, J.; Schreml, S. pH sensing in skin tumors: Methods to study the involvement of GPCRs, acid-sensing ion channels and transient receptor potential vanilloid channels. Exp. Dermatol 2020, 29, 1055–1061. [Google Scholar] [CrossRef] [PubMed]
  153. Dashti, M.R.; Gorbanzadeh, F.; Jafari-Gharabaghlou, D.; Farhoudi Sefidan Jadid, M.; Zarghami, N. G Protein-Coupled Receptor 75 (GPR75) As a Novel Molecule for Targeted Therapy of Cancer and Metabolic Syndrome. Asian Pac. J. Cancer Prev. 2023, 24, 1817–1825. [Google Scholar] [CrossRef] [PubMed]
  154. Godoy-Marin, H.; Duroux, R.; Jacobson, K.A.; Soler, C.; Colino-Lage, H.; Jimenez-Sabado, V.; Montiel, J.; Hove-Madsen, L.; Ciruela, F. Adenosine A(2A) Receptors Are Upregulated in Peripheral Blood Mononuclear Cells from Atrial Fibrillation Patients. Int. J. Mol. Sci. 2021, 22, 3467. [Google Scholar] [CrossRef] [PubMed]
  155. Hashem, S.; Dougha, A.; Tuffery, P. Ligand-Induced Biased Activation of GPCRs: Recent Advances and New Directions from In Silico Approaches. Molecules 2025, 30, 1047. [Google Scholar] [CrossRef]
Figure 1. GPCR-G protein complexes in signal transduction. GPCRs activate heterotrimeric G proteins via GDP-GTP exchange on Gα, releasing Gα and βγ to regulate effectors like AC, PLCβ, and Rho, modulating cAMP, Ca2+, and gene expression. This figure was created with BioRender (agreement number: OT293PX79F).
Figure 1. GPCR-G protein complexes in signal transduction. GPCRs activate heterotrimeric G proteins via GDP-GTP exchange on Gα, releasing Gα and βγ to regulate effectors like AC, PLCβ, and Rho, modulating cAMP, Ca2+, and gene expression. This figure was created with BioRender (agreement number: OT293PX79F).
Ijms 27 00736 g001
Figure 2. Spatiotemporal shifts in GPCR signaling within the NVU during stroke progression. Acute injury phase (hours to days): deleterious GPCR signaling predominates. Neuronal signals activate pericyte/VSMC GPCRs (e.g., ETAR, AT1R), leading to vasoconstriction and no-reflow [51,52,53,54,55]. Concurrently, endothelial GPCRs (e.g., GPR4, S1P3) drive BBB leakage, while microglial GPCRs (e.g., P2Y12, CXCR4) promote neuroinflammation and neuronal apoptosis (red arrows) [45,56,57,58,59]. Recovery phase (Repair): GPCR signaling shifts toward protective pathways. Neuronal activity engages pericyte/VSMC GPCRs (e.g., A2AR, β2AR) to promote vascular stabilization [60,61,62,63,64,65,66,67]. Astrocyte–endothelial crosstalk via GPCRs (e.g., S1P1) restores BBB integrity [37,38,68,69]. Microglial (e.g., CX3CR1) and neuronal (e.g., PAC1, A2AR) GPCR signaling facilitates inflammation resolution, synaptic plasticity, and axonal spouting (green arrows) [60,61,62,63,64,65,70,71]. Abbreviations: AT1R, angiotensin II type 1 receptor; S1P3, sphingosine-1-phosphate receptor 3; A2AR, adenosine A2A receptor; β2AR, β2-adrenergic receptor.
Figure 2. Spatiotemporal shifts in GPCR signaling within the NVU during stroke progression. Acute injury phase (hours to days): deleterious GPCR signaling predominates. Neuronal signals activate pericyte/VSMC GPCRs (e.g., ETAR, AT1R), leading to vasoconstriction and no-reflow [51,52,53,54,55]. Concurrently, endothelial GPCRs (e.g., GPR4, S1P3) drive BBB leakage, while microglial GPCRs (e.g., P2Y12, CXCR4) promote neuroinflammation and neuronal apoptosis (red arrows) [45,56,57,58,59]. Recovery phase (Repair): GPCR signaling shifts toward protective pathways. Neuronal activity engages pericyte/VSMC GPCRs (e.g., A2AR, β2AR) to promote vascular stabilization [60,61,62,63,64,65,66,67]. Astrocyte–endothelial crosstalk via GPCRs (e.g., S1P1) restores BBB integrity [37,38,68,69]. Microglial (e.g., CX3CR1) and neuronal (e.g., PAC1, A2AR) GPCR signaling facilitates inflammation resolution, synaptic plasticity, and axonal spouting (green arrows) [60,61,62,63,64,65,70,71]. Abbreviations: AT1R, angiotensin II type 1 receptor; S1P3, sphingosine-1-phosphate receptor 3; A2AR, adenosine A2A receptor; β2AR, β2-adrenergic receptor.
Ijms 27 00736 g002
Figure 3. GPCR-mediated regulatory mechanisms in cerebrovascular pathologies. In stroke pathogenesis, GPCRs play central roles in cerebrovascular perfusion, neuroimmune responses, BBB homeostasis, and neuronal survival mechanisms. This figure was created with BioRender (agreement number: BU293PX64P).
Figure 3. GPCR-mediated regulatory mechanisms in cerebrovascular pathologies. In stroke pathogenesis, GPCRs play central roles in cerebrovascular perfusion, neuroimmune responses, BBB homeostasis, and neuronal survival mechanisms. This figure was created with BioRender (agreement number: BU293PX64P).
Ijms 27 00736 g003
Figure 4. Discovery of GPCR-targeted agents against cerebrovascular pathologies. GPCR-targeted drug discovery advances through cryo-EM/X-ray structural studies, high-throughput AI-assisted ligand screening, RNA-based therapeutics delivered via nanobody platforms, and precision medicine approaches for stroke. This figure was created with BioRender (agreement number: JB293PX58P).
Figure 4. Discovery of GPCR-targeted agents against cerebrovascular pathologies. GPCR-targeted drug discovery advances through cryo-EM/X-ray structural studies, high-throughput AI-assisted ligand screening, RNA-based therapeutics delivered via nanobody platforms, and precision medicine approaches for stroke. This figure was created with BioRender (agreement number: JB293PX58P).
Ijms 27 00736 g004
Table 1. GPCR signaling mechanisms and translational relevance in cerebrovascular diseases.
Table 1. GPCR signaling mechanisms and translational relevance in cerebrovascular diseases.
GPCRMajor NVU Cell TypesEndogenous Ligand/
Representative
Modulator
Ligand TypeDominant Signaling PathwayPhase-
Dependent
Effects
BBB
Permeability
Clinical
Development Status
Known Signaling BiasReferences
EP2/EP4VSMCs, neuronsPGE2Agonist (endogenous)Gs-cAMP-PKAAcute: neurovascular coupling, ↑CBF; Dysfunction: neurovascular uncouplingIndirectPreclinicalGs-biased[48,49,50]
P2Y12MicrogliaATP/ADPAgonist (endogenous)Gi-cAMPAcute: microglial process extension, injury surveillanceN/AApproved (antiplatelet, peripheral)Gi-biased[94,95,96]
GPR4Endothelial cellsProtons (low pH)Endogenous activatorG12/13-RhoA/Gs-cAMPAcute: leukocyte adhesion, edemaN/APreclinicalG12/13-biased[45,56,57]
ETARVSMCs, pericytesET-1/BosentanAgonist/antagonistGq-Ca2+Acute: vasospasm, no-reflow; blockade improves CBF but may worsen viral injuryLimitedApproved (PAH)Gq-biased[51,52,53]
CX3CR1MicrogliaCX3CL1 (fractalkine)Agonist (endogenous)Gi-cAMPRecovery: inflammation resolution, synaptic stabilizationN/APreclinical (stroke)Context-dependent[70]
S1P1Endothelial cells, astrocytesS1P/FingolimodFunctional agonistGi-Rac1Recovery: BBB repair, anti-inflammationYesApproved (MS)Functional agonism[37,38,68,69]
S1P4Endothelial cellsS1PAgonistG12/13-RhoA/GiBarrier-protective signalingUnknownPreclinicalPoorly defined[59,97]
A2ARNeurons, endothelial cells, microglia, pericytesAdenosine/CGS21680/IstradefyllineAgonist/AntagonistGs-cAMP-PKAAcute: ↑CBF, neuroprotection; Chronic: ↑BBB permeability, inflammationYesApproved (PD); repurposing explored for strokeContext-dependent[60,61,62,63,64,65]
β2ARVSMCs, neurons, astrocytesNorepinephrine/SalbutamolAgonistGs-cAMP; β-arrestinAcute: vasodilation; Chronic: β-arrestin-mediated inflammationYesApproved (non-stroke)G protein/β-arrestin-biased[66,67]
α1ARVSMCsNorepinephrineAgonistGq-Ca2+Acute: vasoconstriction, hypoperfusionLimitedApproved (CV indications)Gq-biased[98,99]
AT1RVSMCs, endothelial cellsAngiotensin II/LosartanAgonist/AntagonistGq-Ca2+Acute: vasoconstriction, hypoperfusionLimitedApproved (hypertension)Gq-biased[54,55]
GPR75VSMCs20-HETEEndogenous agonistGq-Ca2+-PKCAcute: vasoconstriction, hypertension-related hypoperfusionUnknownPreclinicalGq-biased[100]
GPEREndothelial cellsEstrogen/AldosteroneAgonistGs-cAMPAcute/Recovery: NO-mediated vasodilationIndirectPreclinicalGs-biased[101,102]
GPR5CVSMCsOrphanModulatorAT1R-Ca2+ facilitationAcute: enhances Ang II-induced vasoconstrictionN/APreclinicalContext-dependent[103]
GPR5BEndothelial cells, VSMCsOrphanModulatorInflammatory GPCR networksChronic: vascular inflammation, metabolic stressN/APreclinicalPoorly defined[104]
CXCR4Microglia, neurons, endothelial cellsCXCL12AgonistGi-cAMPAcute: inflammation, leukocyte recruitment; Recovery: repair signalingLimitedApproved (oncology); preclinical strokeGi-biased[105,106,107]
PKR1/2NeuronsPK2AgonistGs-cAMP/Gi-cAMP/Gq-Ca2+Acute: pro-inflammatory; Context-dependent neuro-glial signalingUnknownPreclinicalContext-dependent[78]
CB2Microglia, immune cellsEndocannabinoidsAgonistGi-cAMPAnti-inflammatory, neuroprotectionYesPreclinicalGi-biased[108]
S1P2Endothelial cellsS1PAgonistG12/13-RhoAAcute: BBB disruptionUnknownPreclinicalG12/13-biased[109,110]
S1P3Endothelial cellsS1PAgonistG12/13-RhoA/Gq- Ca2+/Gi-cAMPBBB protection in ICH via CCL2 suppressionUnknownPreclinicalContext-dependent[58,59]
H1R/H2REndothelial cellsHistamineAgonistGq-Ca2+ (H1R)/Gs-cAMP (H2R)Acute: ↑BBB permeability, capillary leakageNAApproved (allergy)Receptor-specific[111,112,113]
H3R/H4REndothelial cells, immune cellsHistamineAgonistGi-cAMPAcute/Chronic: immune cell trafficking, BBB modulationLimitedApproved (non-CNS/immune)Gi-biased[111,113,114]
TDAG8Immune cellsProtons (low pH)Endogenous activatorGs-cAMP-PKAAcute: anti-apoptotic, neuroprotectionN/APreclinicalGs-biased[86,115]
GPR68NeuronsProtons (low pH)Endogenous activatorGq-Ca2+-PKCAcute: neuroprotection under acidosisYesPreclinicalGq-biased[87]
GPR37Neurons, astrocytesProsaposinAgonist (endogenous)Gi-cAMPRecovery: neuronal survival, anti-inflammationYesPreclinicalGi-biased[85,116]
PAC1NeuronsPACAPAgonistGs-cAMP-PKA/Gq-Ca2+-PKCRecovery: neuroprotection, plasticityYesPreclinicalContext-dependent[71]
B2RNeurons, endothelial cellsBradykininAgonistGq-Ca2+-NOAcute/Recovery: neuroprotection, angiogenesisLimitedApproved (non-CNS)Gq-biased[83,117]
A1ARNeurons, astrocytesAdenosineAgonistGi-cAMPAcute: anti-excitotoxic; Excessive activation may impair perfusionYesPreclinical (stroke)Gi-biased[118]
CCR5Neurons, microgliaCCL5/MaravirocAntagonistGi-cAMPRecovery: neuroplasticity, motor recoveryLimitedApproved (HIV); repurposing exploredGi-biased[88,89]
5-HT1/2VSMCs, neuronsSerotoninAgonist/AntagonistGi-cAMP (5-HT1)/Gq-Ca2+ (5-HT2)Acute: cerebrovascular tone regulationLimitedApproved (migraine)Receptor-specific[60,119,120,121,122]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gu, Q.; Yao, J.; Sheng, J.; Liu, D. G Protein-Coupled Receptors in Cerebrovascular Diseases: Signaling Mechanisms and Therapeutic Opportunities. Int. J. Mol. Sci. 2026, 27, 736. https://doi.org/10.3390/ijms27020736

AMA Style

Gu Q, Yao J, Sheng J, Liu D. G Protein-Coupled Receptors in Cerebrovascular Diseases: Signaling Mechanisms and Therapeutic Opportunities. International Journal of Molecular Sciences. 2026; 27(2):736. https://doi.org/10.3390/ijms27020736

Chicago/Turabian Style

Gu, Qiuxiang, Jia Yao, Jiajing Sheng, and Dong Liu. 2026. "G Protein-Coupled Receptors in Cerebrovascular Diseases: Signaling Mechanisms and Therapeutic Opportunities" International Journal of Molecular Sciences 27, no. 2: 736. https://doi.org/10.3390/ijms27020736

APA Style

Gu, Q., Yao, J., Sheng, J., & Liu, D. (2026). G Protein-Coupled Receptors in Cerebrovascular Diseases: Signaling Mechanisms and Therapeutic Opportunities. International Journal of Molecular Sciences, 27(2), 736. https://doi.org/10.3390/ijms27020736

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop