• All
  • Article Title
  • Abstract
  • Keywords
  • Author
  • Institution
  • Open Access
    Review

    Neuroinflammation—the role of heteroreceptor complexes

    Neelakanta Sarvashiva Kiran
    Senthilkumar Rajagopal *

    Explor Neuroprot Ther. 2025;5:1004105 DOI: https://doi.org/10.37349/ent.2025.1004105

    Received: February 12, 2025 Accepted: April 22, 2025 Published: May 12, 2025

    Academic Editor: Raymond Chuen-Chung Chang, The University of Hong Kong, China

    This article belongs to the special issue GPCR Heteroreceptor Complexes as Key Players in Neuroprotection

    Abstract

    Neuroinflammation is a hallmark of various neurodegenerative and neuropsychiatric disorders, driven by complex interactions between neurotransmitter receptors and immune signaling pathways. Among these, heteroreceptor complexes—functional assemblies formed by the physical interaction of different G protein-coupled or ionotropic receptor subtypes within the same membrane microdomain—play a crucial role in modulating synaptic activity, neuroimmune responses, and inflammatory cascades. For example, the A2A-D2 receptor complex modulates dopaminergic signaling in the striatum and has been implicated in Parkinson’s disease pathology. These receptor-receptor interactions influence key signaling pathways involving dopamine, serotonin, glutamate, adenosine, and cannabinoid systems, thereby contributing to the pathophysiology of Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, schizophrenia, and depression. Dysregulation of heteroreceptor complexes disrupts neuronal homeostasis, exacerbates neuroinflammatory responses, and influences microglial and astrocytic activation. Understanding the molecular mechanisms governing these interactions, including allosteric modulation and biased agonism, offers novel therapeutic avenues for targeting neuroinflammation. Pharmacological strategies, such as selective allosteric modulators, biased agonists, and receptor-specific ligands, aim to restore heteroreceptor function and mitigate neuroinflammatory damage. Emerging clinical trials—such as those evaluating A2A receptor antagonists like istradefylline for Parkinson’s disease and 5-HT2A antagonists for schizophrenia—have shown promising neuroprotective and anti-inflammatory effects, although larger-scale, long-term studies are needed to confirm efficacy. This review highlights the pivotal role of heteroreceptor complexes in neuroinflammation, discusses their therapeutic potential, and underscores the need for further research into their functional dynamics to develop effective interventions for neurodegenerative and neuropsychiatric diseases.

    Keywords

    Astrocytic, inflammatory cascades, neurodegenerative, neuropsychiatric, microglial

    Introduction

    Overview of neuroinflammation

    Neuroinflammation is a complex and dynamic process that plays a crucial role in both neuroprotection and neurodegeneration. It is primarily mediated by glial cells, including microglia and astrocytes, in response to various stimuli such as infection, injury, or neurodegenerative diseases (Figure 1) [1]. While acute neuroinflammation serves as a protective mechanism by clearing pathogens and damaged cells, chronic neuroinflammation contributes to neuronal dysfunction and cell death, leading to the progression of neurodegenerative disorders like Alzheimer’s disease (AD), Parkinson’s disease (PD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS). The sustained release of pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β) not only promotes gliosis but also initiates a cascade of harmful events by enhancing oxidative stress and disrupting neurotransmitter balance [2, 3]. This disruption can impair synaptic signaling and plasticity, further compromising neuronal communication. Additionally, activation of toll-like receptors (TLRs) and nuclear factor kappa B (NF-κB) signaling pathways amplifies inflammatory responses, creating a pathological loop that perpetuates neuronal damage. This self-reinforcing cycle contributes to neuronal death through mechanisms such as excitotoxicity, mitochondrial dysfunction, and impaired autophagy. Understanding the molecular underpinnings of neuroinflammation is essential for identifying novel therapeutic targets and developing effective interventions to mitigate its deleterious effects on brain function [4, 5]. Emerging strategies include the use of anti-inflammatory agents like minocycline, immune modulators targeting microglial activation, and neuroprotective compounds such as cannabinoids [6] and Nrf2 activators, which show promise in preclinical and early clinical studies [7, 8].

    Generalized mechanism of induced neuroinflammation. CNS: central nervous system; TNF-α: tumor necrosis factor-alpha; IL-1β: interleukin-1 beta; IL-6: interleukin-6

    Role of neurotransmitter receptors in brain homeostasis

    Neurotransmitter receptors play a fundamental role in maintaining brain homeostasis by modulating synaptic transmission, neuronal excitability, and neuroplasticity [9]. These receptors, classified into ionotropic and metabotropic types, respond to key neurotransmitters such as glutamate, dopamine, serotonin, gamma-aminobutyric acid (GABA), and acetylcholine [10]. Excitatory neurotransmitters like glutamate activate N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, which are crucial for synaptic plasticity and cognitive functions. Conversely, inhibitory neurotransmitters like GABA bind to GABA_A and GABA_B receptors to regulate neuronal excitability and prevent excitotoxicity [11].

    Neuroinflammation disrupts neurotransmitter receptor signaling, leading to synaptic dysfunction and neuronal loss. Pro-inflammatory cytokines can alter receptor expression, desensitize synaptic responses, and impair receptor trafficking [12]. For instance, increased levels of TNF-α have been shown to enhance AMPA receptor expression, exacerbating excitotoxicity in neurodegenerative conditions [13]. Similarly, dopaminergic dysfunction due to neuroinflammation is a key pathological feature in PD, where microglial activation and cytokine release lead to the progressive loss of dopaminergic neurons in the substantia nigra [14]. The interaction between inflammatory mediators and neurotransmitter receptors underscores the need to explore their crosstalk in the context of neurodegenerative diseases [15].

    Importance of heteroreceptor complexes

    Heteroreceptor complexes are emerging as critical modulators of brain function, serving as integrators of multiple signaling pathways by facilitating receptor crosstalk [16]. These complexes are formed through the physical and functional interaction of different neurotransmitter receptors, altering their pharmacological and signaling properties. Notable examples include dopamine-glutamate receptor complexes (D2-NMDA, D1-NMDA), adenosine-dopamine (A2A-D2) [17], and serotonin-dopamine (5-HT2A-D2) receptor interactions [18, 19]. By modulating neurotransmission, synaptic plasticity, and neuroinflammatory responses, heteroreceptor complexes contribute to brain homeostasis and neuroprotection [20].

    In the context of neuroinflammation, heteroreceptor complexes play a dual role by either exacerbating or mitigating inflammatory responses depending on their configuration and signaling dynamics. However, it is important to note that the existence and stability of these heteromers under pathological conditions remain an active area of investigation. In some disease states, receptor expression may be downregulated or altered, potentially preventing heteromer formation [21]. Further studies using post-mortem tissues, disease models, and advanced imaging techniques are needed to confirm whether these complexes are maintained or disrupted in patients with neuroinflammatory disorders. For instance, the A2A-D2 receptor complex is known to regulate dopaminergic neurotransmission, and its dysfunction has been implicated in neuroinflammatory processes observed in PD [22]. Conversely, activation of A2A receptors within these complexes can also suppress microglial activation and inhibit pro-inflammatory cytokine release, thereby exerting anti-inflammatory effects in certain neurodegenerative conditions. Similarly, alterations in the balance between excitatory and inhibitory receptor complexes contribute to synaptic dysfunction in neurodegenerative diseases [23]. Targeting heteroreceptor complexes offers a promising therapeutic avenue, as modulating their interactions can restore receptor function and attenuate neuroinflammatory damage [24]. Recent studies suggest that allosteric modulators and biased agonists can selectively influence heteroreceptor complex activity, paving the way for precision medicine approaches in neurodegenerative and psychiatric disorders [25]. For instance, several A2A receptor antagonists and 5-HT2A inverse agonists are currently being evaluated in preclinical and early-phase clinical trials for their potential to modulate neuroinflammatory pathways and improve cognitive or motor symptoms in PD and schizophrenia models.

    Understanding the interplay between neuroinflammation and heteroreceptor complexes provides a new dimension in neuropharmacology and disease management [26]. Given their intricate role in regulating neurotransmission and neuroinflammation, further research into heteroreceptor complexes could unlock novel therapeutic strategies aimed at mitigating chronic neuroinflammatory conditions and improving neurodegenerative disease outcomes [27, 28].

    Methodology

    This review was conducted to synthesize current knowledge on the role of heteroreceptor complexes in neuroinflammation and their therapeutic potential in neurodegenerative and neuropsychiatric disorders. Literature was searched across multiple databases, including PubMed, Scopus, and Web of Science, from January 2000 to March 2025. The search strategy employed Boolean operators and keywords such as “heteroreceptor complexes”, “neuroinflammation”, “dopamine receptors”, “glial activation”, “biased agonism”, and “allosteric modulators”. Articles were selected based on relevance, peer-reviewed status, and English language. Reviews, original research articles, and clinical trial reports were included. Studies focusing solely on monoreceptor mechanisms or non-CNS inflammatory models were excluded. Data extraction was performed independently by both authors to ensure consistency and reliability. A PRISMA-based flow diagram summarizing the selection process is included in Figure 2.

    PRISMA-based flow chart diagram representing the methodology of this review

    Heteroreceptor complexes: structure and functions

    Heteroreceptor complexes are specialized molecular assemblies formed by the direct physical interaction of two or more different neurotransmitter receptors. Unlike monomeric receptors that function independently, heteroreceptor complexes allow for dynamic receptor crosstalk, leading to modified pharmacological and functional properties [16]. These complexes can form between receptors of the same family, such as different dopamine receptor subtypes, or between distinct neurotransmitter systems, like dopamine-glutamate or serotonin-dopamine interactions [29]. The presence of these receptor assemblies provides a sophisticated regulatory mechanism for synaptic transmission, neuroplasticity, and neuroinflammatory responses (Figure 3) [30].

    A diverse range of neurological disorders associated with neuroinflammation and their respective heteroreceptors

    The classification of heteroreceptor complexes is based on their structural and functional properties. Homomeric complexes involve identical receptor subunits that enhance function through cooperative interactions [31]. In contrast, heteromeric complexes consist of different receptor subtypes, integrating multiple signaling pathways to regulate neurotransmission. Some heteroreceptor complexes are transient, existing only under specific physiological conditions, whereas others form stable interactions, contributing to synaptic regulation and neuronal function [32]. Several well-characterized heteroreceptor complexes, including dopamine-glutamate (D1-NMDA, D2-NMDA), adenosine-dopamine (A2A-D2), and serotonin-dopamine (5-HT2A-D2), play significant roles in modulating neurotransmission and neuroinflammation, which has been explained in detail in Table 1. For example, dopamine-glutamate receptor complexes are crucial for cognitive function and synaptic plasticity, while adenosine-dopamine receptor interactions regulate motor control and are implicated in PD pathology [20].

    Various types of heteroreceptors and their role in neuroinflammation

    Heteroreceptor complexesStructure and mechanismNeuroinflammatory roleAssociated disordersReferences
    A2A-D2 (adenosine-dopamine)The A2A receptor (GPCR) forms heteromeric complexes with the D2 receptor, leading to an antagonistic interaction where A2A activation inhibits D2 signaling.Enhances neuroinflammation by inhibiting dopaminergic neurotransmission, leading to microglial activation and neurodegeneration.Parkinson’s disease, Huntington’s disease[106]
    5-HT2A-D2 (serotonin-dopamine)Interaction between serotonin 5-HT2A and dopamine D2 receptors modulates dopaminergic neurotransmission and immune signaling.Dysregulation contributes to aberrant dopamine signaling, leading to heightened neuroinflammatory responses in psychiatric disorders.Schizophrenia, depression, and bipolar disorder[107]
    D2-NMDA (dopamine-glutamate)Dopamine D2 receptors modulate NMDA receptor function, affecting excitatory neurotransmission and synaptic plasticity.Dysregulation leads to excessive glutamate excitotoxicity, oxidative stress, and chronic neuroinflammation.Alzheimer’s disease, schizophrenia, stroke[30]
    CB1-D2 (cannabinoid-dopamine)CB1 cannabinoid receptors interact with D2 dopamine receptors to regulate inflammatory and neuroprotective pathways.Impairment of CB1-D2 signaling contributes to excessive immune activation, neuroinflammation, and neurodegenerative processes.Multiple sclerosis, neuroinflammation, anxiety disorders[108]
    mGluR5-D2 (metabotropic glutamate-dopamine)mGluR5 modulates D2 receptor activity, influencing synaptic plasticity and neuroimmune responses.Aberrant crosstalk leads to excitotoxicity, oxidative stress, and neuroinflammatory damage.Autism spectrum disorder, schizophrenia, epilepsy[109]
    TrkB-NMDA (tropomyosin receptor kinase B-glutamate)TrkB, activated by BDNF, interacts with NMDA receptors to regulate synaptic strength and neuroprotection.Dysregulation results in impaired neurotrophic signaling, increased oxidative stress, and inflammatory responses.Major depressive disorder, Alzheimer’s disease[110]
    P2X7-NMDA (Purinoceptor-Glutamate)P2X7 receptor, an ATP-gated ion channel, modulates NMDA receptor activity and glial cell function.Overactivation leads to excessive calcium influx, neuronal death, and chronic neuroinflammation.Multiple sclerosis, Amyotrophic lateral sclerosis (ALS)[111]
    α7nAChR-NMDA (nicotinic acetylcholine-glutamate)Nicotinic α7 receptor modulates NMDA receptor-mediated synaptic transmission and anti-inflammatory responses.Dysregulation results in impaired cholinergic anti-inflammatory signaling and neurotoxicity.Alzheimer’s disease, schizophrenia, Parkinson’s disease[112]
    CXCR4-CCR5 (chemokine receptors)CXCR4 and CCR5 heterodimerization regulate neuroimmune signaling and leukocyte infiltration in the CNS.Hyperactivation promotes neuroinflammatory cascades, contributing to neuronal damage.HIV-associated neurocognitive disorder, multiple sclerosis[113]
    GPR37-D2 (G-protein coupled receptor 37-dopamine)GPR37 interacts with D2 receptors, modulating dopaminergic transmission and neuroinflammation.Dysfunction contributes to dopaminergic neurodegeneration and inflammatory signaling.Parkinson’s disease, schizophrenia[114]
    Display full size

    GPCRs: G-protein-coupled receptors; NMDA: N-methyl-D-aspartate; BDNF: brain-derived neurotrophic factor; CNs: central nervous system

    Heteroreceptor complexes function through various crosstalk mechanisms that influence receptor signaling, trafficking, and neurotransmitter dynamics. One key mechanism is allosteric modulation, where ligand binding to one receptor induces conformational changes in the other, altering its binding affinity and signaling efficiency [33]. For instance, in the A2A-D2 receptor complex, adenosine receptor activation reduces dopamine D2 receptor signaling, which has implications for PD [34]. Another mechanism involves direct protein-protein interactions, where physical contact between receptor domains facilitates cooperative signaling and synaptic stability. This is observed in the dopamine D1-NMDA receptor complex, which enhances NMDA receptor-mediated synaptic plasticity, essential for learning and memory [35].

    G-protein coupling interference is another critical mechanism in heteroreceptor interactions, particularly among G-protein-coupled receptors (GPCRs). When receptors form complexes, their G-protein coupling preferences can shift, leading to altered intracellular signaling [36]. For example, in serotonin-dopamine (5-HT2A-D2) complexes, serotonin receptor activation redirects dopamine D2 receptor signaling from the Gi pathway to alternative intracellular cascades, affecting mood regulation and schizophrenia pathology [37]. Additionally, heteroreceptor complexes influence second messenger modulation by altering intracellular pathways such as cyclic adenosine monophosphate (cAMP) and inositol triphosphate (IP3), leading to changes in neurotransmitter release and synaptic plasticity [38]. Another form of crosstalk occurs through receptor trafficking and internalization, where one receptor modulates the internalization and recycling of another. This is evident in the adenosine A2A-dopamine D2 complex, where A2A receptor activation promotes the internalization of the dopamine D2 receptor, leading to altered dopamine signaling and motor control [39].

    Heteroreceptor complexes regulate multiple signaling pathways that affect neuronal function, synaptic plasticity, and neuroinflammation. One of the key pathways involved is the cAMP-protein kinase A (PKA) pathway, which plays a central role in neurotransmitter signaling [40]. Heteroreceptor complexes often modulate cAMP levels, thereby influencing PKA activity. In the adenosine A2A-dopamine D2 complex, A2A receptor activation increases cAMP levels, leading to PKA activation, which inhibits dopamine D2 receptor signaling [41]. This mechanism is crucial in PD, where excessive A2A receptor activation contributes to motor dysfunction. Similarly, serotonin-dopamine heteroreceptor interactions can modulate cAMP levels, affecting mood regulation and response to antipsychotic medications [42].

    Another key pathway regulated by heteroreceptor complexes is the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway, which is essential for neuronal survival, plasticity, and inflammatory responses. The dopamine D1-NMDA receptor complex enhances NMDA receptor signaling through ERK phosphorylation, promoting synaptic strengthening and memory formation [43]. However, excessive activation of this pathway can also contribute to excitotoxicity in neurodegenerative diseases [44]. The adenosine A2A-dopamine D2 complex, in contrast, can suppress ERK activation, impacting neuronal survival in conditions such as Huntington’s disease and AD [45].

    The phosphoinositide 3-kinase (PI3K)-Akt pathway is another crucial signaling mechanism influenced by heteroreceptor complexes. This pathway is involved in cell survival, neuroprotection, and inflammatory responses [46]. Glutamate-metabotropic receptor complexes, for instance, activate PI3K-Akt signaling, protecting neurons from excitotoxicity and reducing neuroinflammatory responses. GABAergic interactions also modulate Akt activity, maintaining excitatory-inhibitory balance and preventing excessive neuronal excitability, which is particularly relevant in epilepsy and neurodevelopmental disorders [47].

    Calcium-dependent signaling pathways are also significantly regulated by heteroreceptor complexes. Calcium signaling is critical for synaptic function, neurotransmitter release, and plasticity [48]. Dopamine-glutamate receptor complexes, such as D1-NMDA interactions, potentiate calcium influx, enhancing synaptic plasticity but also increasing the risk of excitotoxic damage in neurodegenerative diseases [49]. Similarly, serotonin-glutamate receptor interactions influence calcium-dependent neurotransmission, which plays a role in mood regulation and neuropsychiatric disorders such as depression and schizophrenia [50].

    Heteroreceptor complexes play a crucial role in neuroinflammation, as their dysfunction can lead to excessive cytokine production, oxidative stress, and synaptic abnormalities. In AD, disrupted dopamine-glutamate interactions contribute to cognitive decline and neuroinflammatory responses [51]. In PD, imbalances in adenosine-dopamine heteroreceptor interactions lead to motor dysfunction and neurodegeneration. In schizophrenia, dysregulated serotonin-dopamine receptor interactions play a key role in altered neurotransmission and neuroinflammation, influencing disease progression and therapeutic outcomes [52].

    Targeting heteroreceptor complexes through pharmacological interventions is a promising approach to restoring receptor function and mitigating neuroinflammation-driven diseases. By developing drugs that selectively modulate heteroreceptor interactions, researchers aim to fine-tune neurotransmission and reduce the inflammatory burden in neurodegenerative and neuropsychiatric disorders. Understanding the structure, function, and signaling mechanisms of these complexes is essential for advancing therapeutic strategies that address the complexities of brain disorders [53].

    Heteroreceptor complexes in neuroinflammatory conditions

    AD

    AD is a progressive neurodegenerative disorder characterized by amyloid-beta (Aβ) plaque accumulation, tau protein hyperphosphorylation, synaptic dysfunction, and chronic neuroinflammation. The role of heteroreceptor complexes in AD pathology is increasingly recognized, particularly in relation to neurotransmitter dysregulation and inflammatory responses [54]. Dopamine-glutamate heteroreceptor complexes, such as the D1-NMDA and D2-NMDA receptor interactions, are crucial for synaptic plasticity and cognitive function. In AD, these complexes are disrupted due to excessive glutamate excitotoxicity and reduced dopamine signaling, leading to impaired synaptic transmission and neuronal loss. The dysregulation of the D1-NMDA receptor complex exacerbates synaptic weakening, further contributing to cognitive deficits in AD patients [55].

    Adenosine-dopamine heteroreceptor complexes, particularly the A2A-D2 complex, also play a significant role in AD-associated neuroinflammation. The overactivation of A2A receptors in response to chronic neuroinflammatory stimuli inhibits D2 receptor signaling, leading to impaired cognitive and motor functions [56]. The involvement of these receptor interactions in neuroinflammation suggests that targeting A2A receptors could be a potential therapeutic approach to modulate neuroinflammatory cascades and improve neurotransmitter balance in AD patients. Additionally, serotonin-dopamine heteroreceptor complexes (5-HT2A-D2) contribute to AD pathology by altering neurotransmitter homeostasis and inflammatory responses. Dysregulated 5-HT2A receptor activity is associated with increased tau aggregation and neuroinflammatory processes, making these heteroreceptor interactions a promising target for AD interventions [57].

    PD

    PD is a neurodegenerative disorder characterized by progressive loss of dopaminergic neurons in the substantia nigra and chronic neuroinflammation. The dysregulation of heteroreceptor complexes, particularly those involving dopamine and adenosine receptors, is a key contributor to PD pathophysiology. The A2A-D2 heteroreceptor complex is critically involved in the regulation of motor control, as A2A receptor overactivation suppresses dopamine D2 receptor function, leading to motor deficits [58]. In PD, excessive adenosine A2A receptor activity enhances neuroinflammatory signaling through microglial activation and pro-inflammatory cytokine release, exacerbating neuronal loss and motor impairments. Pharmacological blockade of A2A receptors has been explored as a therapeutic approach to restore D2 receptor function and reduce neuroinflammation [59].

    Dopamine-glutamate heteroreceptor complexes, such as D1-NMDA and D2-NMDA interactions, are also disrupted in PD, contributing to cognitive impairments and excitotoxic damage. The loss of dopaminergic input in PD alters NMDA receptor activity, leading to excessive calcium influx and oxidative stress, further promoting neurodegeneration. Additionally, serotonin-dopamine heteroreceptor complexes (5-HT2A-D2) play a role in PD-related non-motor symptoms, including depression and cognitive decline [60]. The dysregulation of these receptor interactions contributes to altered neurotransmitter signaling and inflammation, making them potential targets for adjunctive therapies in PD treatment. The modulation of heteroreceptor complexes offers a promising strategy to mitigate neuroinflammation and neurodegeneration in PD patients [61].

    MS

    MS is a chronic autoimmune-mediated neuroinflammatory disorder characterized by demyelination, axonal damage, and neurodegeneration. The role of heteroreceptor complexes in MS pathology is gaining attention, particularly regarding neurotransmitter dysregulation and immune system interactions. Glutamate-dopamine heteroreceptor complexes, such as D1-NMDA and D2-NMDA interactions, are implicated in MS-related excitotoxicity and neuroinflammation [62]. The excessive activation of NMDA receptors due to dysregulated glutamate signaling contributes to oligodendrocyte damage and demyelination, leading to disease progression [63].

    Adenosine-dopamine heteroreceptor complexes (A2A-D2) are also involved in MS-related neuroinflammation. A2A receptor activation promotes microglial activation and pro-inflammatory cytokine release, exacerbating neuroimmune responses. The inhibition of A2A receptors has been proposed as a potential therapeutic approach to modulate neuroinflammatory pathways and protect against demyelination in MS patients [64]. Additionally, serotonin-dopamine heteroreceptor interactions (5-HT2A-D2) are linked to mood disturbances and cognitive impairments observed in MS. The dysregulation of these receptor complexes affects neurotransmitter balance, leading to increased neuroinflammatory responses and exacerbation of psychiatric symptoms. Targeting heteroreceptor complexes to modulate neurotransmitter and immune interactions presents a novel approach for mitigating neuroinflammation and neurodegeneration in MS patients [65].

    Depression, schizophrenia, and psychiatric disorders

    Neuroinflammation plays a significant role in psychiatric disorders, including depression and schizophrenia, where heteroreceptor complexes are key regulators of neurotransmitter interactions. In depression, serotonin-dopamine heteroreceptor complexes (5-HT2A-D2) are crucial for mood regulation and cognitive function. The dysregulation of 5-HT2A receptor activity in response to chronic stress and neuroinflammation contributes to altered dopamine signaling, leading to depressive symptoms [66]. The interaction between serotonin and dopamine receptors modulates neuroinflammatory responses, and abnormalities in these complexes are linked to increased levels of pro-inflammatory cytokines, oxidative stress, and neuronal dysfunction. Pharmacological agents targeting these receptor interactions, such as serotonin-dopamine modulators, have shown promise in alleviating depressive symptoms by restoring neurotransmitter balance and reducing inflammation [67].

    In schizophrenia, dopamine-glutamate heteroreceptor complexes, such as D2-NMDA interactions, play a crucial role in synaptic plasticity and cognitive function. The dysfunction of these complexes contributes to altered glutamate signaling, leading to excitotoxic damage and neuroinflammatory processes [68]. The dysregulation of dopamine-serotonin interactions (5-HT2A-D2) is also implicated in schizophrenia-related cognitive deficits and psychotic symptoms. Increased serotonin receptor activity negatively affects dopamine transmission, contributing to impaired cognitive processing and neuroinflammatory responses. Antipsychotic medications targeting these heteroreceptor interactions aim to restore neurotransmitter homeostasis and reduce neuroinflammation, providing therapeutic benefits in schizophrenia treatment [69].

    Heteroreceptor complexes also play a role in bipolar disorder and anxiety-related conditions, where neurotransmitter imbalance and neuroinflammatory mechanisms contribute to disease pathology. The modulation of glutamate-dopamine and serotonin-dopamine receptor interactions influences mood regulation, synaptic plasticity, and inflammatory pathways. Emerging research suggests that targeting heteroreceptor complexes could provide novel therapeutic strategies for psychiatric disorders by reducing neuroinflammation and restoring neurotransmitter function [70]. Understanding the role of these receptor interactions in psychiatric conditions is essential for developing precision medicine approaches that address the underlying neurobiological mechanisms of mental health disorders [71].

    Molecular mechanisms and pathophysiology

    Impact on neuroimmune response

    Neuroinflammation is a complex physiological process involving interactions between neurons, glial cells, and immune signaling pathways. Heteroreceptor complexes play a crucial role in modulating neuroimmune responses by regulating synaptic transmission, neurotransmitter release, and inflammatory signaling cascades [72]. These receptor complexes, consisting of two or more different neurotransmitter receptors physically interacting, influence intracellular signaling pathways that mediate neuroinflammation. Disruptions in heteroreceptor crosstalk can lead to aberrant immune activation, contributing to chronic neuroinflammatory states that underlie various neurological disorders [73].

    In conditions such as AD, PD, and MS, heteroreceptor complexes act as regulators of neuroimmune interactions. Dysregulated heteroreceptor complexes, particularly those involving dopamine (D2), adenosine (A2A), serotonin (5-HT2A), and glutamate (NMDA) receptors, contribute to altered immune responses [74]. These interactions affect the release of pro-inflammatory cytokines, reactive oxygen species, and neurotoxic mediators, leading to neuronal dysfunction and neurodegeneration. Thus, understanding the role of heteroreceptor complexes in neuroimmune signaling provides valuable insights into novel therapeutic approaches targeting neuroinflammatory pathways [75].

    Role of microglia and astrocytes

    Microglia and astrocytes are the primary immune cells of the CNS, playing an integral role in maintaining homeostasis and responding to neuroinflammatory stimuli. Heteroreceptor complexes modulate the activation state of these glial cells, influencing their capacity to regulate synaptic function and neuroinflammatory responses [76]. In pathological conditions, dysregulated receptor crosstalk between neurons and glial cells exacerbates inflammation, leading to progressive neuronal damage [77].

    Microglial activation is influenced by heteroreceptor interactions involving purinergic (P2X7), dopamine (D2), and adenosine (A2A) receptors. Under normal physiological conditions, these complexes maintain microglial homeostasis by balancing pro-inflammatory and anti-inflammatory responses [78]. However, in neurodegenerative diseases, excessive activation of A2A receptors, for instance, leads to heightened microglial reactivity, promoting the release of TNF-α and IL-1β, which contribute to neurotoxicity. Similarly, dopamine-glutamate heteroreceptor complexes influence astrocytic function, modulating glutamate uptake and preventing excitotoxicity. When disrupted, these interactions impair astrocytic glutamate clearance, exacerbating neuroinflammatory damage [79].

    Astrocytes, in particular, contribute to neuroinflammation by releasing pro-inflammatory cytokines and modulating blood-brain barrier integrity. The crosstalk between serotonin (5-HT2A) and glutamate receptors in astrocytes regulates inflammatory responses, influencing cytokine production and gliosis [80]. The involvement of heteroreceptor complexes in glial function highlights their significance in shaping neuroimmune responses and presents potential targets for mitigating neuroinflammation-associated neurodegeneration [81].

    Cross-talk with cytokines and inflammatory markers

    Heteroreceptor complexes influence the expression and release of cytokines and other inflammatory markers, serving as mediators of neuroinflammation [82]. These receptor complexes modulate intracellular signaling pathways that regulate the balance between pro-inflammatory and anti-inflammatory cytokine production. Dysregulation of these interactions leads to excessive inflammation, a hallmark of neurodegenerative and psychiatric disorders [83].

    The activation of heteroreceptor complexes, such as A2A-D2 and 5-HT2A-D2, alters immune responses by modulating the activity of NF-κB, MAPKs, and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways [84]. These intracellular signaling cascades govern the production of cytokines such as IL-6, TNF-α, and IL-1β. Excessive cytokine release in response to dysregulated receptor crosstalk contributes to chronic neuroinflammation, exacerbating neuronal damage and synaptic dysfunction [85].

    Furthermore, heteroreceptor complexes interact with chemokines and other inflammatory mediators that regulate immune cell infiltration into the CNS. In MS, for example, heteroreceptor interactions influence the migration of peripheral immune cells across the blood-brain barrier, contributing to neuroinflammatory demyelination [86]. Similarly, in psychiatric disorders such as depression and schizophrenia, heteroreceptor dysregulation leads to altered cytokine profiles, influencing neurotransmitter function and exacerbating disease pathology [87].

    Therapeutic perspectives

    Drug development

    Heteroreceptor complexes represent a promising target for drug development, particularly in the treatment of neuroinflammatory and neurodegenerative disorders. These complexes, formed by interactions between different neurotransmitter receptors, play a crucial role in modulating synaptic transmission, neuroimmune responses, and inflammatory signaling pathways [88]. Given their influence on neuronal homeostasis and immune activation, pharmacological interventions aimed at restoring the balance of heteroreceptor signaling may provide novel therapeutic avenues for conditions such as AD, PD, MS, and psychiatric disorders [89].

    One of the key advantages of targeting heteroreceptor complexes is the ability to fine-tune neurotransmitter systems with greater specificity than traditional single-receptor-targeting drugs. For instance, the dopamine-glutamate heteroreceptor complexes, including D2-NMDA interactions, are involved in cognitive and motor functions. Dysregulation of these complexes contributes to excitotoxicity and neurodegeneration, making them potential targets for drugs that modulate both dopamine and glutamate signaling simultaneously [90]. Similarly, the adenosine-dopamine heteroreceptor complex (A2A-D2) has been implicated in PD, where excessive adenosine A2A receptor activity inhibits dopamine D2 receptor function, exacerbating motor impairments and neuroinflammation. Drugs that selectively modulate A2A receptor activity, such as istradefylline, have demonstrated therapeutic potential in alleviating symptoms of PD by restoring dopaminergic signaling and reducing neuroinflammation [91].

    Another promising approach is the modulation of serotonin-dopamine heteroreceptor complexes (5-HT2A-D2), which play a role in psychiatric disorders such as schizophrenia and depression. Atypical antipsychotics, such as clozapine and risperidone, target these receptor interactions, leading to improved treatment efficacy compared to traditional dopamine-based antipsychotics. By refining drug design to selectively target heteroreceptor complexes, researchers can develop more effective and less side-effect-prone treatments for neuropsychiatric and neurodegenerative conditions [92].

    Role of allosteric regulators and biased agonism

    The emergence of allosteric modulators and biased agonists has revolutionized the field of receptor pharmacology, offering new strategies to manipulate heteroreceptor complex function. Unlike traditional orthosteric ligands, which directly activate or inhibit receptors, allosteric modulators bind to distinct sites on the receptor complex to enhance or diminish receptor responses in a highly selective manner. This specificity reduces off-target effects and allows for more precise control over receptor signaling pathways [93].

    Positive allosteric modulators (PAMs) and negative allosteric modulators (NAMs) have shown potential in fine-tuning heteroreceptor activity in neuroinflammatory disorders. For example, PAMs targeting metabotropic glutamate receptors (mGluRs) have been explored as potential treatments for neurodegenerative diseases by modulating glutamate-dopamine receptor interactions and reducing excitotoxicity. Similarly, A2A receptor antagonists, acting as NAMs, have demonstrated efficacy in preclinical and clinical models of PD by preventing excessive adenosine-mediated inhibition of D2 receptor activity [94].

    Biased agonism, or ligand-directed signaling, is another promising approach for targeting heteroreceptor complexes. This concept involves designing drugs that preferentially activate specific signaling pathways while avoiding pathways associated with adverse effects. For example, dopamine D2 receptor-biased agonists have been developed to selectively engage beneficial G-protein signaling while minimizing β-arrestin-mediated side effects, which are linked to dyskinesia and cognitive dysfunction [95]. Such approaches could enhance the therapeutic index of drugs targeting heteroreceptor complexes in neuroinflammatory and neuropsychiatric disorders [96].

    In addition to small-molecule modulators, peptide-based therapeutics and monoclonal antibodies are being investigated as potential modulators of heteroreceptor function. These biologics offer high specificity and can be engineered to disrupt or stabilize receptor-receptor interactions, providing novel therapeutic strategies for neuroinflammatory diseases [97].

    Clinical trials and future directions

    The clinical translation of heteroreceptor complex-targeting drugs is still in its early stages, but several promising candidates are being explored in clinical trials. Adenosine A2A receptor antagonists, such as istradefylline, have already been approved for the treatment of PD, providing proof of concept that targeting heteroreceptor interactions can yield therapeutic benefits [98]. Additionally, serotonin-dopamine receptor modulators, including pimavanserin, have been developed for the treatment of PD psychosis and are being investigated for other neuropsychiatric disorders [99].

    Several ongoing clinical trials are evaluating allosteric modulators targeting heteroreceptor interactions in neuroinflammatory conditions. For example, mGluR5 allosteric modulators are being tested for their ability to modulate glutamate-dopamine receptor interactions in schizophrenia and depression. Similarly, biased agonists targeting dopamine and serotonin receptors are being explored for their potential to improve therapeutic outcomes in psychiatric and neurodegenerative diseases while reducing side effects [100].

    Future research in this area will focus on refining our understanding of heteroreceptor complex dynamics and identifying novel targets for therapeutic intervention. Advances in Boolean analysis, structural biology, cryo-electron microscopy, and computational modeling are providing new insights into receptor-receptor interactions, enabling the rational design of drugs with enhanced specificity and efficacy [101, 102]. Additionally, the development of gene editing technologies and RNA-based therapies holds promise for modulating heteroreceptor function at the genetic and epigenetic levels [103].

    Personalized medicine approaches will also play a key role in the future of heteroreceptor-targeted therapies. By utilizing biomarkers and neuroimaging techniques, clinicians can identify patient-specific alterations in heteroreceptor signaling and tailor treatments accordingly [104]. This precision medicine approach has the potential to improve treatment outcomes for patients with neuroinflammatory and neurodegenerative disorders [105].

    Conclusion

    Despite significant advancements, numerous open research questions remain regarding the precise molecular mechanisms underlying heteroreceptor complex formation, function, and dysregulation in neuroinflammatory disorders. The dynamic nature of these complexes, their tissue-specific expression, and the influence of genetic and environmental factors require further elucidation. Future studies must explore how heteroreceptor interactions contribute to disease pathophysiology and whether they can serve as reliable biomarkers for early diagnosis and disease progression. Additionally, translational applications of heteroreceptor-targeted therapies must be optimized through rigorous preclinical validation and well-designed clinical trials. The development of novel pharmacological tools, including high-affinity allosteric modulators and biased agonists, holds promise for precise therapeutic modulation with minimal adverse effects. As the field progresses, integrating cutting-edge technologies such as cryo-electron microscopy, optogenetics, and single-cell transcriptomics will provide deeper insights into heteroreceptor signaling networks, paving the way for innovative drug discovery approaches. This emerging understanding has the potential to revolutionize neuropharmacology by enabling highly targeted, mechanism-based therapies tailored to individual patient profiles. The convergence of basic and clinical research will be instrumental in translating these findings into effective treatments, ultimately improving patient outcomes in neuroinflammatory and neurodegenerative diseases. By bridging molecular neuroscience with clinical innovation, the study of heteroreceptor complexes stands at the forefront of transformative advancements in personalized medicine and neurological care.

    Abbreviations

    AD:

    Alzheimer’s disease

    AMPA:

    α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid

    cAMP:

    cyclic adenosine monophosphate

    CNS:

    central nervous system

    ERK:

    extracellular signal-regulated kinase

    GABA:

    gamma-aminobutyric acid

    GPCR:

    G-protein-coupled receptor

    IL:

    interleukin

    MAPK:

    mitogen-activated protein kinase

    MS:

    multiple sclerosis

    NAMs:

    negative allosteric modulators

    NF-κB:

    nuclear factor kappa B

    NMDA:

    N-methyl-D-aspartate

    PAMs:

    positive allosteric modulators

    PD:

    Parkinson’s disease

    PI3K:

    phosphoinositide 3-kinase

    PKA:

    protein kinase A

    TNF-α:

    tumor necrosis factor-alpha

    Declarations

    Acknowledgements

    We, the authors, sincerely acknowledge REVA University for providing an opportunity and platform for research.

    Author contributions

    NSK: Conceptualization, Investigation, Writing—original draft. SR: Validation, Writing—review & editing, Supervision. Both authors read and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2025.

    Publisher’s note

    Open Exploration maintains a neutral stance on jurisdictional claims in published institutional affiliations and maps. All opinions expressed in this article are the personal views of the author(s) and do not represent the stance of the editorial team or the publisher.

    References

    Kölliker-Frers R, Udovin L, Otero-Losada M, Kobiec T, Herrera MI, Palacios J, et al. Neuroinflammation: An Integrating Overview of Reactive-Neuroimmune Cell Interactions in Health and Disease. Mediators Inflamm. 2021;2021:9999146. [DOI] [PubMed] [PMC]
    Teleanu DM, Niculescu AG, Lungu II, Radu CI, Vladâcenco O, Roza E, et al. An Overview of Oxidative Stress, Neuroinflammation, and Neurodegenerative Diseases. Int J Mol Sci. 2022;23:5938. [DOI] [PubMed] [PMC]
    Hassamal S. Chronic stress, neuroinflammation, and depression: an overview of pathophysiological mechanisms and emerging anti-inflammatories. Front Psychiatry. 2023;14:1130989. [DOI] [PubMed] [PMC]
    Valente M, Dentoni M, Bellizzi F, Kuris F, Gigli GL. Specialized Pro-Resolving Mediators in Neuroinflammation: Overview of Studies and Perspectives of Clinical Applications. Molecules. 2022;27:4836. [DOI] [PubMed] [PMC]
    Alsbrook DL, Di Napoli M, Bhatia K, Biller J, Andalib S, Hinduja A, et al. Neuroinflammation in Acute Ischemic and Hemorrhagic Stroke. Curr Neurol Neurosci Rep. 2023;23:40731. [DOI] [PubMed] [PMC]
    Rivas-Santisteban R, Lillo J, Raïch I, Muñoz A, Lillo A, Rodríguez-Pérez AI, et al. The cannabinoid CB1 receptor interacts with the angiotensin AT2 receptor. Overexpression of AT2-CB1 receptor heteromers in the striatum of 6-hydroxydopamine hemilesioned rats. Exp Neurol. 2023;362:114319. [DOI] [PubMed]
    Kiran NS, Vaishnavi G, Lowkesh G, Yashaswini C, Rajagopal S. Immune System and Obsessive-Compulsive Disorder: Intervention Through Immunotherapy. In: Nutrition and Obsessive-Compulsive Disorder. London: CRC Press; 2023. pp. 236–51.
    Kiran NS, Yashaswini C, Lowkesh G, Range K, Madhu R. Phytochemicals and Herbal Medicines: Potential Drug Candidates for Obsessive-Compulsive Disorder Treatment. In: Nutrition and Obsessive-Compulsive Disorder. London: CRC Press; 2023. pp. 189–200.
    Akyuz E, Polat AK, Eroglu E, Kullu I, Angelopoulou E, Paudel YN. Revisiting the role of neurotransmitters in epilepsy: An updated review. Life Sci. 2021;265:118826. [DOI] [PubMed]
    Zhang H, Wang Z, Wang G, Song X, Qian Y, Liao Z, et al. Understanding the Connection between Gut Homeostasis and Psychological Stress. J Nutr. 2023;153:92439. [DOI] [PubMed]
    Stolero N, Frenkel D. The dialog between neurons and microglia in Alzheimer’s disease: The neurotransmitters view. J Neurochem. 2021;158:141224. [DOI] [PubMed]
    Nimgampalle M, Chakravarthy H, Sharma S, Shree S, Bhat AR, Pradeepkiran JA, et al. Neurotransmitter systems in the etiology of major neurological disorders: Emerging insights and therapeutic implications. Ageing Res Rev. 2023;89:101994. [DOI] [PubMed]
    Ludhiadch A, Sharma R, Muriki A, Munshi A. Role of Calcium Homeostasis in Ischemic Stroke: A Review. CNS & Neurological Disorders-Drug Targets. 2022;21:5261. [DOI]
    Abruzzo PM, Panisi C, Marini M. The Alteration of Chloride Homeostasis/GABAergic Signaling in Brain Disorders: Could Oxidative Stress Play a Role? Antioxidants (Basel). 2021;10:1316. [DOI] [PubMed] [PMC]
    Lim D, Jeong J, Song J. Lipocalin 2 regulates iron homeostasis, neuroinflammation, and insulin resistance in the brains of patients with dementia: Evidence from the current literature. CNS Neurosci Ther. 2021;27:88394. [DOI] [PubMed] [PMC]
    Pérez de la Mora M, Borroto-Escuela DO, Crespo-Ramírez M, Rejón-Orantes JDC, Palacios-Lagunas DA, Martínez-Mata MK, et al. Dysfunctional Heteroreceptor Complexes as Novel Targets for the Treatment of Major Depressive and Anxiety Disorders. Cells. 2022;11:1826. [DOI] [PubMed] [PMC]
    Lillo A, Raïch I, Lillo J, Pérez-Olives C, Navarro G, Franco R. Expression of the Adenosine A2A-A3 Receptor Heteromer in Different Brain Regions and Marked Upregulation in the Microglia of the Transgenic APPSw,Ind Alzheimer’s Disease Model. Biomedicines. 2022;10:214. [DOI] [PubMed] [PMC]
    Borroto-Escuela DO, Ambrogini P, Narvaez M, Di Liberto V, Beggiato S, Ferraro L, et al. Serotonin Heteroreceptor Complexes and Their Integration of Signals in Neurons and Astroglia-Relevance for Mental Diseases. Cells. 2021;10:1902. [DOI] [PubMed] [PMC]
    Rico AJ, Dopeso-Reyes IG, Martínez-Pinilla E, Sucunza D, Pignataro D, Roda E, et al. Neurochemical evidence supporting dopamine D1-D2 receptor heteromers in the striatum of the long-tailed macaque: changes following dopaminergic manipulation. Brain Struct Funct. 2017;222:176784. [DOI] [PubMed] [PMC]
    Narváez M, Crespo-Ramírez M, Fores-Pons R, Pita-Rodríguez M, Ciruela F, Filip M, et al. Study of GPCR Homo- and Heteroreceptor Complexes in Specific Neuronal Cell Populations Using the In Situ Proximity Ligation Assay. Receptor and Ion Channel Detection in the Brain. 2021:11734. [DOI]
    ElNebrisi E, Lozon Y, Oz M. The Role of α7-Nicotinic Acetylcholine Receptors in the Pathophysiology and Treatment of Parkinson’s Disease. Int J Mol Sci. 2025;26:3210. [DOI] [PubMed] [PMC]
    Arrabal-Gómez C, Beltran-Casanueva R, Hernández-García A, Bayolo-Guanche JV, Barbancho-Fernández MA, Serrano-Castro PJ, et al. Enhancing Cognitive Functions and Neuronal Growth through NPY1R Agonist and Ketamine Co-Administration: Evidence for NPY1R-TrkB Heteroreceptor Complexes in Rats. Cells. 2024;13:669. [DOI] [PubMed] [PMC]
    Pinna A, Bonaventura J, Farré D, Sánchez M, Simola N, Mallol J, et al. L-DOPA disrupts adenosine A2A-cannabinoid CB1-dopamine D2 receptor heteromer cross-talk in the striatum of hemiparkinsonian rats: Biochemical and behavioral studies. Exp Neurol. 2014;253:18091. [DOI] [PubMed]
    Ilchibaeva T, Tsybko A, Zeug A, Müller FE, Guseva D, Bischoff S, et al. Serotonin Receptor 5-HT2A Regulates TrkB Receptor Function in Heteroreceptor Complexes. Cells. 2022;11:2384. [DOI] [PubMed] [PMC]
    Borroto-Escuela DO, Ferraro L, Beggiato S, Narváez M, Fores-Pons R, Alvarez-Contino JE, et al. The coming together of allosteric and phosphorylation mechanisms in the molecular integration of A2A heteroreceptor complexes in the dorsal and ventral striatal-pallidal GABA neurons. Pharmacol Rep. 2021;73:1096108. [DOI] [PubMed] [PMC]
    Ren P, Wang JY, Xu MJ, Chen HL, Duan JY, Li YF. Sigma-1 receptor activation produces faster antidepressant-like effect through enhancement of hippocampal neuroplasticity: Focus on sigma-1-5-HT1A heteroreceptor complex. Neurochemistry International. 2025;184:105937. [DOI]
    Ambrogini P, Lattanzi D, Pagliarini M, Di Palma M, Sartini S, Cuppini R, et al. 5HT1AR-FGFR1 Heteroreceptor Complexes Differently Modulate GIRK Currents in the Dorsal Hippocampus and the Dorsal Raphe Serotonin Nucleus of Control Rats and of a Genetic Rat Model of Depression. Int J Mol Sci. 2023;24:7467. [DOI] [PubMed] [PMC]
    Martínez-Pinilla E, Rico AJ, Rivas-Santisteban R, Lillo J, Roda E, Navarro G, et al. Expression of GPR55 and either cannabinoid CB1 or CB2 heteroreceptor complexes in the caudate, putamen, and accumbens nuclei of control, parkinsonian, and dyskinetic non-human primates. Brain Struct Funct. 2020;225:215364. [DOI] [PubMed]
    Bonaventura J, Rico AJ, Moreno E, Sierra S, Sánchez M, Luquin N, et al. L-DOPA-treatment in primates disrupts the expression of A2A adenosine-CB1 cannabinoid-D2 dopamine receptor heteromers in the caudate nucleus. Neuropharmacology. 2014;79:90100. [DOI] [PubMed]
    Guidolin D, Tortorella C, Marcoli M, Cervetto C, De Caro R, Maura G, et al. Possible roles of heteroreceptor complexes in excitotoxic processes. Explor Neuroprot Ther. 2024;4:36691. [DOI]
    Yan Q, Li XJ, Wang QQ, Jia W, Wang SL. FGF21 Exhibits Neuroprotective Effects by Promoting 5-HT1AR-FGFR1 Heteroreceptor Complexes and Triggering MEK1/2-ERK1/2 Signaling Pathway. Mol Neurobiol. 2025;62:636982. [DOI] [PubMed]
    Mirchandani-Duque M, Choucri M, Hernández-Mondragón JC, Crespo-Ramírez M, Pérez-Olives C, Ferraro L, et al. Membrane Heteroreceptor Complexes as Second-Order Protein Modulators: A Novel Integrative Mechanism through Allosteric Receptor-Receptor Interactions. Membranes (Basel). 2024;14:96. [DOI] [PubMed] [PMC]
    Fuxe K, Guidolin D, Agnati LF, Borroto-Escuela DO. Dopamine heteroreceptor complexes as therapeutic targets in Parkinson’s disease. Expert Opin Ther Targets. 2015;19:37798. [DOI] [PubMed]
    Borroto-Escuela DO, Cuesta-Marti C, Lopez-Salas A, Chruścicka-Smaga B, Crespo-Ramírez M, Tesoro-Cruz E, et al. The oxytocin receptor represents a key hub in the GPCR heteroreceptor network: potential relevance for brain and behavior. Front Mol Neurosci. 2022;15:1055344. [DOI] [PubMed] [PMC]
    Borroto-Escuela DO, Fuxe K. Adenosine heteroreceptor complexes in the basal ganglia are implicated in Parkinson’s disease and its treatment. J Neural Transm (Vienna). 2019;126:45571. [DOI] [PubMed] [PMC]
    Porzionato A, Stocco E, Guidolin D, Agnati L, Macchi V, De Caro R. Receptor-Receptor Interactions of G Protein-Coupled Receptors in the Carotid Body: A Working Hypothesis. Front Physiol. 2018;9:697. [DOI] [PubMed] [PMC]
    Borroto-Escuela DO, Wydra K, Filip M, Fuxe K. A2AR-D2R Heteroreceptor Complexes in Cocaine Reward and Addiction. Trends Pharmacol Sci. 2018;39:100820. [DOI] [PubMed]
    Borroto-Escuela DO, Romero-Fernandez W, Garriga P, Ciruela F, Narvaez M, Tarakanov AO, et al. G protein-coupled receptor heterodimerization in the brain. Methods Enzymol. 2013;521:28194. [DOI] [PubMed]
    Rodríguez-Ruiz M, Moreno E, Moreno-Delgado D, Navarro G, Mallol J, Cortés A, et al. Heteroreceptor Complexes Formed by Dopamine D1, Histamine H3, and N-Methyl-D-Aspartate Glutamate Receptors as Targets to Prevent Neuronal Death in Alzheimer’s Disease. Mol Neurobiol. 2017;54:453750. [DOI] [PubMed]
    Feltmann K, Borroto-Escuela DO, Rüegg J, Pinton L, de Oliveira Sergio T, Narváez M, et al. Effects of Long-Term Alcohol Drinking on the Dopamine D2 Receptor: Gene Expression and Heteroreceptor Complexes in the Striatum in Rats. Alcohol Clin Exp Res. 2018;42:33851. [DOI] [PubMed] [PMC]
    Di Liberto V, Borroto-Escuela DO, Frinchi M, Verdi V, Fuxe K, Belluardo N, et al. Existence of muscarinic acetylcholine receptor (mAChR) and fibroblast growth factor receptor (FGFR) heteroreceptor complexes and their enhancement of neurite outgrowth in neural hippocampal cultures. Biochim Biophys Acta Gen Subj. 2017;1861:23545. [DOI] [PubMed]
    Narváez M, Andrade-Talavera Y, Valladolid-Acebes I, Fredriksson M, Siegele P, Hernandez-Sosa A, et al. Existence of FGFR1-5-HT1AR heteroreceptor complexes in hippocampal astrocytes. Putative link to 5-HT and FGF2 modulation of hippocampal gamma oscillations. Neuropharmacology. 2020;170:108070. [DOI] [PubMed]
    Lamirande Ed, Gagnon C. The extracellular signal-regulated kinase (ERK) pathway is involved in human sperm function and modulated by the superoxide anion. Mol Hum Reprod. 2002;8:12435. [DOI] [PubMed]
    Frankland PW, Josselyn SA. Hippocampal Neurogenesis and Memory Clearance. Neuropsychopharmacology. 2016;41:3823. [DOI] [PubMed] [PMC]
    Morén C, deSouza RM, Giraldo DM, Uff C. Antioxidant Therapeutic Strategies in Neurodegenerative Diseases. Int J Mol Sci. 2022;23:9328. [DOI] [PubMed] [PMC]
    Kumari R, Shekhar N, Tyagi S, Thakur AK. Mitochondrial dysfunctions and neurodegenerative diseases: A mini-review. J Anal Pharm Res. 2021;10:1479. [DOI]
    Garcia-Garcia AL, Newman-Tancredi A, Leonardo ED. P5-HT1A receptors in mood and anxiety: recent insights into autoreceptor versus heteroreceptor function. Psychopharmacology (Berl). 2014;231:62336. [DOI] [PubMed] [PMC]
    Batistič O, Kudla J. Analysis of calcium signaling pathways in plants. Biochimica et Biophysica Acta (BBA)-General Subjects. 2012;1820:128393. [DOI]
    Buck SA, Erickson-Oberg M, Logan RW, Freyberg Z. Relevance of interactions between dopamine and glutamate neurotransmission in schizophrenia. Mol Psychiatry. 2022;27:358391. [DOI] [PubMed] [PMC]
    Fedotova EI, Abramov AY, Berezhnov AV. Dopamine Protects Neurons against Glutamate-Induced Excitotoxicity. Biochem. 2023;17:3442. [DOI]
    Solís O, Moratalla R. Dopamine receptors: homomeric and heteromeric complexes in L-DOPA-induced dyskinesia. J Neural Transm (Vienna). 2018;125:118794. [DOI] [PubMed]
    Bloem BR, Okun MS, Klein C. Parkinson’s disease. Lancet. 2021;397:2284303. [DOI] [PubMed]
    Borroto-Escuela DO, Pintsuk J, Schäfer T, Friedland K, Ferraro L, Tanganelli S, et al. Multiple D2 heteroreceptor complexes: new targets for treatment of schizophrenia. Ther Adv Psychopharmacol. 2016;6:7794. [DOI] [PubMed] [PMC]
    Navarro G, Borroto-Escuela D, Angelats E, Etayo Í, Reyes-Resina I, Pulido-Salgado M, et al. Receptor-heteromer mediated regulation of endocannabinoid signaling in activated microglia. Role of CB1 and CB2 receptors and relevance for Alzheimer’s disease and levodopa-induced dyskinesia. Brain, behavior, and immunity. 2018;67:13951. [DOI]
    Ferré S, Agnati LF, Ciruela F, Lluis C, Woods AS, Fuxe K, et al. Neurotransmitter receptor heteromers and their integrative role in ‘local modules’: The striatal spine module. Brain Res Rev. 2007;55:5567. [DOI] [PubMed] [PMC]
    Hoque KE, Indorkar RP, Sammut S, West AR. Impact of dopamine-glutamate interactions on striatal neuronal nitric oxide synthase activity. Psychopharmacology (Berl). 2010;207:57181. [DOI] [PubMed] [PMC]
    Trejo-Lopez JA, Yachnis AT, Prokop S. Neuropathology of Alzheimer’s Disease. Neurotherapeutics. 2022;19:17385. [DOI] [PubMed] [PMC]
    Liu TW, Chen CM, Chang KH. Biomarker of Neuroinflammation in Parkinson’s Disease. Int J Mol Sci. 2022;23:4148. [DOI]
    Araújo B, Caridade-Silva R, Soares-Guedes C, Martins-Macedo J, Gomes ED, Monteiro S, et al. Neuroinflammation and Parkinson’s Disease-From Neurodegeneration to Therapeutic Opportunities. Cells. 2022;11:2908. [DOI] [PubMed] [PMC]
    Badanjak K, Fixemer S, Smajić S, Skupin A, Grünewald A. The Contribution of Microglia to Neuroinflammation in Parkinson’s Disease. Int J Mol Sci. 2021;22:4676. [DOI] [PubMed] [PMC]
    Grotemeyer A, McFleder RL, Wu J, Wischhusen J, Ip CW. Neuroinflammation in Parkinson's Disease - Putative Pathomechanisms and Targets for Disease-Modification. Front Immunol. 2022;13:878771. [DOI] [PubMed] [PMC]
    Nociti V, Romozzi M. The Role of BDNF in Multiple Sclerosis Neuroinflammation. Int J Mol Sci. 2023;24:8447. [DOI] [PubMed] [PMC]
    Azzolini F, Gilio L, Pavone L, Iezzi E, Dolcetti E, Bruno A, et al. Neuroinflammation Is Associated with GFAP and sTREM2 Levels in Multiple Sclerosis. Biomolecules. 2022;12:222. [DOI] [PubMed] [PMC]
    Al-Badri G, Castorina A. Insights into the Role of Neuroinflammation in the Pathogenesis of Multiple Sclerosis. J Funct Morphol Kinesiol. 2018;3:13. [DOI]
    Colasanti A, Guo Q, Giannetti P, Wall MB, Newbould RD, Bishop C, et al. Hippocampal Neuroinflammation, Functional Connectivity, and Depressive Symptoms in Multiple Sclerosis. Biol Psychiatry. 2016;80:6272. [DOI] [PubMed] [PMC]
    Feng T, Tripathi A, Pillai A. Inflammatory Pathways in Psychiatric Disorders: The case of Schizophrenia and Depression. Curr Behav Neurosci Rep. 2020;7:12838. [DOI] [PubMed] [PMC]
    Maier R, Moser G, Chen GB, Ripke S, Coryell W,Potash JB, et al.; Cross-Disorder Working Group of the Psychiatric Genomics Consortium. JJoint Analysis of Psychiatric Disorders Increases Accuracy of Risk Prediction for Schizophrenia, Bipolar Disorder, and Major Depressive Disorder. Am J Hum Genet. 2015;96:28394. [DOI] [PubMed] [PMC]
    Musella A, Gentile A, Rizzo FR, De Vito F, Fresegna D, Bullitta S, et al. Interplay Between Age and Neuroinflammation in Multiple Sclerosis: Effects on Motor and Cognitive Functions. Front Aging Neurosci. 2018;10:238. [DOI] [PubMed] [PMC]
    Baker D, Jackson SJ, Pryce G. Cannabinoid control of neuroinflammation related to multiple sclerosis. Br J Pharmacol. 2007;152:64954. [DOI] [PubMed] [PMC]
    Gilsbach R, Hein L. Are the pharmacology and physiology of α₂ adrenoceptors determined by α₂-heteroreceptors and autoreceptors respectively? Br J Pharmacol. 2012;165:90102. [DOI] [PubMed] [PMC]
    Wydra K, Gawliński D, Frankowska M, Gawlińska K, Borroto-Escuela DO, Fuxe K. Heteroreceptor Complexes in Substance Use Disorders. Handbook of Neurotoxicity. 2022:129. [DOI]
    Morris G, Berk M, Galecki P, Walder K, Maes M. The Neuro-Immune Pathophysiology of Central and Peripheral Fatigue in Systemic Immune-Inflammatory and Neuro-Immune Diseases. Mol Neurobiol. 2016;53:1195219. [DOI] [PubMed]
    Talbot S, Foster SL, Woolf CJ. Neuroimmunity: Physiology and Pathology. Annu Rev Immunol. 2016;34:42147. [DOI] [PubMed]
    Choi YH, Laaker C, Hsu M, Cismaru P, Sandor M, Fabry Z. Molecular Mechanisms of Neuroimmune Crosstalk in the Pathogenesis of Stroke. Int J Mol Sci. 2021;22:9486. [DOI] [PubMed] [PMC]
    Chavan SS, Pavlov VA, Tracey KJ. Mechanisms and Therapeutic Relevance of Neuro-immune Communication. Immunity. 2017;46:92742. [DOI] [PubMed] [PMC]
    Reemst K, Noctor SC, Lucassen PJ, Hol EM. The Indispensable Roles of Microglia and Astrocytes during Brain Development. Front Hum Neurosci. 2016;10:566. [DOI] [PubMed] [PMC]
    Kwon HS, Koh SH. Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes. Transl Neurodegener. 2020;9:42. [DOI] [PubMed] [PMC]
    Sen MK, Mahns DA, Coorssen JR, Shortland PJ. The roles of microglia and astrocytes in phagocytosis and myelination: Insights from the cuprizone model of multiple sclerosis. Glia. 2022;70:121550. [DOI] [PubMed] [PMC]
    Di Benedetto G, Burgaletto C, Bellanca CM, Munafò A, Bernardini R, Cantarella G. Role of Microglia and Astrocytes in Alzheimer’s Disease: From Neuroinflammation to Ca2+ Homeostasis Dysregulation. Cells. 2022;11:2728. [DOI] [PubMed] [PMC]
    Karve IP, Taylor JM, Crack PJ. The contribution of astrocytes and microglia to traumatic brain injury. Br J Pharmacol. 2016;173:692702. [DOI] [PubMed] [PMC]
    Matejuk A, Ransohoff RM. Crosstalk Between Astrocytes and Microglia: An Overview. Front Immunol. 2020;11:1416. [DOI] [PubMed] [PMC]
    Kany S, Vollrath JT, Relja B. Cytokines in Inflammatory Disease. Int J Mol Sci. 2019;20:6008. [DOI] [PubMed] [PMC]
    Elsalhy M, Azizieh F, Raghupathy R. Cytokines as diagnostic markers of pulpal inflammation. Int Endod J. 2013;46:57380. [DOI] [PubMed]
    Ueland T, Gullestad L, Nymo SH, Yndestad A, Aukrust P, Askevold ET. Inflammatory cytokines as biomarkers in heart failure. Clin Chim Acta. 2015;443:717. [DOI] [PubMed]
    Miliopoulos D, Gkouziouta A, Leontiadis E, Adamopoulos S. Cytokines and inflammatory markers. In: Oxford Textbook of Heart Failure. Oxford University Press; 2022. p. 193. [DOI]
    Courtney JM, Ennis M, Elborn JS. Cytokines and inflammatory mediators in cystic fibrosis. J Cyst Fibros. 2004;3:22331. [DOI] [PubMed]
    Rabaan AA, Al-Ahmed SH, Muhammad J, Khan A, Sule AA, Tirupathi R, et al. Role of Inflammatory Cytokines in COVID-19 Patients: A Review on Molecular Mechanisms, Immune Functions, Immunopathology and Immunomodulatory Drugs to Counter Cytokine Storm. Vaccines (Basel). 2021;9:436. [DOI] [PubMed] [PMC]
    Liu C, Chu D, Kalantar-Zadeh K, George J, Young HA, Liu G. Cytokines: From Clinical Significance to Quantification. Adv Sci (Weinh). 2021;8:e2004433. [DOI] [PubMed] [PMC]
    Dawidowski B, Górniak A, Podwalski P, Lebiecka Z, Misiak B, Samochowiec J. The Role of Cytokines in the Pathogenesis of Schizophrenia. J Clin Med. 2021;10:3849. [DOI] [PubMed] [PMC]
    Jagannath B, Lin KC, Pali M, Sankhala D, Muthukumar S, Prasad S. Temporal profiling of cytokines in passively expressed sweat for detection of infection using wearable device. Bioeng Transl Med. 2021;6:e10220. [DOI] [PubMed] [PMC]
    Fontes-Cal TCM, Mattos RT, Medeiros NI, Pinto BF, Belchior-Bezerra M, Roque-Souza B, et al. Crosstalk Between Plasma Cytokines, Inflammation, and Liver Damage as a New Strategy to Monitoring NAFLD Progression. Front Immunol. 2021;12:708959. [DOI] [PubMed] [PMC]
    Bülow Anderberg S, Luther T, Berglund M, Larsson R, Rubertsson S, Lipcsey M, et al. Increased levels of plasma cytokines and correlations to organ failure and 30-day mortality in critically ill Covid-19 patients. Cytokine. 2021;138:155389. [DOI] [PubMed] [PMC]
    Slosky LM, Caron MG, Barak LS. Biased Allosteric Modulators: New Frontiers in GPCR Drug Discovery. Trends Pharmacol Sci. 2021;42:28399. [DOI] [PubMed] [PMC]
    McNeill SM, Baltos JA, White PJ, May LT. Biased agonism at adenosine receptors. Cell Signal. 2021;82:109954. [DOI] [PubMed]
    Kelly E, Conibear A, Henderson G. Biased Agonism: Lessons from Studies of Opioid Receptor Agonists. Annu Rev Pharmacol Toxicol. 2023;63:491515. [DOI] [PubMed]
    Ippolito M, Benovic JL. Biased agonism at β-adrenergic receptors. Cell Signal. 2021;80:109905. [DOI] [PubMed] [PMC]
    Eiger DS, Boldizsar N, Honeycutt CC, Gardner J, Rajagopal S. Biased agonism at chemokine receptors. Cell Signal. 2021;78:109862. [DOI] [PubMed] [PMC]
    Rong L, Li N, Zhang Z. Emerging therapies for glioblastoma: current state and future directions. J Exp Clin Cancer Res. 2022;41:142. [DOI] [PubMed] [PMC]
    Mishra-Kalyani PS, Amiri Kordestani L, Rivera DR, Singh H, Ibrahim A, DeClaro RA, et al. External control arms in oncology: current use and future directions. Ann Oncol. 2022;33:37683. [DOI] [PubMed]
    Smrke A, Anderson PM, Gulia A, Gennatas S, Huang PH, Jones RL. Future Directions in the Treatment of Osteosarcoma. Cells. 2021;10:172. [DOI] [PubMed] [PMC]
    Rivas-Santisteban R, Rico AJ, Muñoz A, Rodríguez-Pérez AI, Reyes-Resina I, Navarro G, et al. Boolean analysis shows a high proportion of dopamine D2 receptors interacting with adenosine A2A receptors in striatal medium spiny neurons of mouse and non-human primate models of Parkinson’s disease. Neurobiol Dis. 2023;188:106341. [DOI] [PubMed]
    Eckerling A, Ricon-Becker I, Sorski L, Sandbank E, Ben-Eliyahu S. Stress and cancer: mechanisms, significance and future directions. Nat Rev Cancer. 2021;21:76785. [DOI] [PubMed]
    Li W, Wang F, Guo R, Bian Z, Song Y. Targeting macrophages in hematological malignancies: recent advances and future directions. J Hematol Oncol. 2022;15:110. [DOI] [PubMed] [PMC]
    Sadee W, Wang D, Hartmann K, Toland AE. Pharmacogenomics: Driving Personalized Medicine. Pharmacol Rev. 2023;75:789814. [DOI] [PubMed] [PMC]
    Ingber DE. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat Rev Genet. 2022;23:46791. [DOI] [PubMed] [PMC]
    Franco R, Navarro G, Martínez-Pinilla E. The adenosine A2A receptor in the basal ganglia: Expression, heteromerization, functional selectivity and signalling. Int Rev Neurobiol. 2023;170:4971. [DOI] [PubMed]
    Feng Y, Lu Y. Immunomodulatory Effects of Dopamine in Inflammatory Diseases. Front Immunol. 2021;12:663102. [DOI] [PubMed] [PMC]
    Margoni M, Preziosa P, Rocca MA, Filippi M. Depressive symptoms, anxiety and cognitive impairment: emerging evidence in multiple sclerosis. Transl Psychiatry. 2023;13:264. [DOI] [PubMed] [PMC]
    Zahra A, Wang YF, Wang Q, Wu J. Shared Etiology in Autism Spectrum Disorder and Epilepsy with Functional Disability. Behav Neurol. 2022;2022:5893519. [DOI] [PubMed] [PMC]
    Correia AS, Cardoso A, Vale N. Highlighting Immune System and Stress in Major Depressive Disorder, Parkinson’s, and Alzheimer’s Diseases, with a Connection with Serotonin. Int J Mol Sci. 2021;22:8525. [DOI] [PubMed] [PMC]
    Pathak N, Vimal SK, Tandon I, Agrawal L, Hongyi C, Bhattacharyya S. Neurodegenerative Disorders of Alzheimer, Parkinsonism, Amyotrophic Lateral Sclerosis and Multiple Sclerosis: An Early Diagnostic Approach for Precision Treatment. Metab Brain Dis. 2022;37:67104. [DOI] [PubMed]
    Kuusimäki T, Al-Abdulrasul H, Kurki S, Hietala J, Hartikainen S, Koponen M, et al. Increased Risk of Parkinson’s Disease in Patients With Schizophrenia Spectrum Disorders. Mov Disord. 2021;36:135361. [DOI] [PubMed]
    Thompson LJ, Genovese J, Hong Z, Singh MV, Singh VB. HIV-Associated Neurocognitive Disorder: A Look into Cellular and Molecular Pathology. Int J Mol Sci. 2024;25:4697. [DOI] [PubMed] [PMC]
    Kim K, Kim S, Myung W, Shim I, Lee H, Kim B, et al. Shared Genetic Background between Parkinson’s Disease and Schizophrenia: A Two-Sample Mendelian Randomization Study. Brain Sci. 2021;11:1042. [DOI] [PubMed] [PMC]