Skip to main content

Beyond the itch: the complex interplay of immune, neurological, and psychological factors in chronic urticaria

Abstract

Chronic urticaria (CU) arises from a multifaceted interplay of immunological, neurological, and psychological components. Immune dysregulation, mediated through both immunoglobulin E (IgE)-dependent and IgE-independent pathways, plays a pivotal role in CU pathogenesis, involving key effector cells such as mast cells (MCs), basophils, and eosinophils. This dysregulation culminates in the release of histamine, prostaglandins, and other mediators, which precipitate pruritus. The chronicity of the disease leads to sustained pruritic symptoms, contributing to both central and peripheral sensitization. The excitation of the itch circuit is augmented, leading to the release of neurotransmitters and neuropeptides, which subsequently interact with immune cells. Psychological factors such as depression, anxiety, and stress exacerbate CU symptoms and diminish quality of life. These factors disrupt the hypothalamic-pituitary-adrenal (HPA) axis and the autonomic nervous system (ANS). Furthermore, the act of scratching activates the reward circuit, resulting in the manifestation of the itch-scratching cycle. Current treatments, such as antihistamines, omalizumab, and cyclosporine, demonstrate variable efficacy and are often associated with adverse effects. A holistic approach addressing both psychological and physiological aspects is advocated. This review highlights the critical importance of understanding neuroimmune interactions and the influence of psychosomatic factors in CU. It aims to enhance diagnostic and therapeutic strategies by integrating psychological, neurological, and immunological perspectives.

Background

CU is a multifactorial skin disorder. The overall lifetime prevalence of CU is 4.4% [1], with a higher prevalence among women, particularly during their youth and middle age [2]. Characterized by recurrent wheals and angioedema lasting for six weeks or more, CU significantly impacts various physiological systems, including the integumentary, cardiovascular, respiratory, gastrointestinal, central nervous, and musculoskeletal systems [3, 4]. This multi-system involvement highlights the complexity of the disease. Up to 85.9% of patients report experiencing CU symptoms during work, with more than half noting a considerable reduction in their productivity due to the condition [5].

CU is typically classified into two main types. Chronic inducible urticaria (CIndU) is triggered by identifiable external factors such as medication, physical stimuli, or stress, while chronic spontaneous urticaria (CSU) lacks a clear external trigger [6, 7]. In patients with CSU, stress may also exacerbate disease activity [8]. CU is closely associated with immune dysregulation via both IgE-dependent and IgE-independent pathways [9,10,11]. The release of histamine and other inflammatory mediators results in intense itching and swelling. Although second-generation antihistamines, omalizumab, and cyclosporine are standard treatments with an efficacy of 60–80%, they are not universally effective and are often accompanied by side effects such as headaches and drowsiness [12,13,14,15].

In addition, CU is heavily influenced by neurological and psychological factors [16]. The interaction between the immune system, nerve system, and psychological states form a complex network of neuroimmune interactions. About 25% of people with skin diseases also exhibit depression, anxiety, or somatoform reactions [17]. Patients with CU often experience psychiatric disorders [18]. Psychological stress is known to exacerbate CU symptoms, creating a vicious cycle where itching leads to increased stress, which, in turn, worsens the itching. This cycle significantly lowers the quality of life and complicates treatment [19,20,21]. Sleep disturbances and heightened anxiety have been shown to correlate with increased disease activity in CU [22], further emphasizing the role of psychological comorbidities in the progression of disease.

Despite significant progress in elucidating the mechanisms underlying CU, our comprehension of the intricate interactions among the immune, neurological, and psychological systems that sustain and exacerbate this condition remains incomplete. Existing animal models offer valuable insights, but they inadequately represent the complexity of CU in humans. The disparities in skin architecture, immune responses, and neuroimmune signaling between humans and animal models further constrain the reliability of these preclinical systems. Consequently, the translation of preclinical findings into clinical practice continues to pose significant challenges, potentially impeding the advancement of effective therapeutic interventions. This highlights the pressing necessity for the development of more sophisticated models that incorporate neuroimmune and psychological factors, thereby more accurately representing the complexities inherent in human CU pathology. Furthermore, in the basis of scrutinizing psychological elements on this crosstalk, a comprehensive approach to diagnosis and treatment is also advocated.

Immune cell involvement and animal models about CU

Activation and recruitment of immune cells

The etiology of CSU and CIndU remains incompletely understood, with distinct immune cell mechanisms implicated in each condition. CIndU comprises several subtypes, including cold urticaria, heat urticaria, delayed pressure urticaria, symptomatic dermographism, solar urticaria, vibratory angioedema, cholinergic urticaria, contact urticaria, and aquagenic urticaria. Current evidence suggests that CSU involves the dysregulation of various immune cells, including MCs, basophils, eosinophils, T cells, B cells, and neutrophils, whereas CIndU is primarily associated with aberrant activation of MCs and basophils. Investigating the activation and regulation of these immune cells in CU is of importance, as depicted in Figs. 1 and 2.

MCs

The activation of MCs is the result from antigen binding to IgE antibodies and cross-linking of the high-affinity IgE receptor (FcεRI) [23]. 25% patients had levels of anti-FcεRI IgE in CSU [24]. MAS-related G protein-coupled receptor X2 (MRGPRX2) exhibits a high level of expression in cutaneous MCs. Its activation releases degranulation products and pro-inflammatory mediators, promoting multicellular signaling and initiating itch signals in sensory neurons [25]. It is also involved in IgE-independent pathways in CU [26]. Subsequently, the manifestation of urticaria is observed [27].

Three factors triggering MC activation in CSU are receptor stimulation, receptor upregulation, and intracellular dysregulation from overexpression of spleen tyrosine kinase or activation of the inhibitory Src homology 2 domain-containing inositol phosphatase-related pathway [28]. Th2 inflammation, B cell autoantibodies, basophil-released histamine, and eosinophil or monocyte initiation of the extrinsic coagulation pathway strongly influence MC regulation in CSU [29]. In CSU, histamine-induced expression of tissue factor (TF) on endothelial cells initiates the extrinsic coagulation pathway, leading to the production of complement 5a (C5a), an anaphylatoxin that subsequently activates both basophils and MCs via the C5a receptor (C5aR) [30]. Additionally, in CSU, upregulated expression of stem-cell factor (SCF) in the skin promotes MC activation through its interaction with the stem-cell factor receptor (KIT) [31]. Certain elements of the coagulation cascade, including thrombin and the complex of activated factor VII, activated factor X, and tissue factor, have the capacity to induce MC degranulation through the activation of protease-activated receptor 1 (PAR1) and protease-activated receptor 2 (PAR2), respectively, in CSU [32,33,34].

CIndU involves MC-triggered wheals in reaction to specific stimuli [35, 36]. Histamine release from dermal MCs is central to CIndU symptoms [37]. In cold urticaria, cold-induced autoallergens lead to IgE production, causing MC degranulation and proinflammatory mediator release [38]. And the activation of MCs via the phosphoinositide 3-kinase (PI3K)/ protein kinase B (Akt)/ nuclear factor kappa-B (NF-κB) pathway is crucial in immunologic contact urticaria [39].

Basophils

Basophils participate in the Th2 immune response and engage in interactions with other skin cells, secreting a range of pruritic mediators such as histamine, interleukin-4 (IL-4), interleukin-13 (IL-13), interleukin-31 (IL-31), and substance P (SP) [40, 41]. Activated basophils exhibit heightened sensitivity to interleukin-3 (IL-3) stimulation, suggesting that deficiencies in signal transduction pathways may be rectified in patients with CU following stimulation of IgE crosslinking [42].

During CSU activity, the expression levels of IgE receptor signaling molecules in blood basophils change, accompanied by altered degranulation functions [43]. The basophil activation phenotype is associated with a longer disease duration in CSU [44,45,46]. Decreased peripheral basophil counts may be due to the recruitment of basophils to the site of skin lesions in CSU [47]. The increased CD63 in CSU patients is related to allergen sensitivity, serum autoreactivity, and basophilic reactivity [48]. Serum basophilic CD203c upregulation and positive autologous serum skin tests were significantly correlated with basophilic histamine release in CSU [49]. Additionly, basophils in the bloodstream of CSU patients have impaired IgE-mediated pathways but retain heightened responsiveness to C5a and histamine [30].

In CIndU, they have shown slight activation in peripheral blood but no abnormalities in responsiveness [50]. The median expression of FcεRI, normalized per density, in blood basophils has demonstrated a significant increase [51].

Eosinophils

Eosinophils are a type of granulocyte containing eosinophilic granules. They inhibit basophil activity by releasing prostaglandin E (PGE) and phagocytosing granules expelled by basophils, as well as releasing histaminase to destroy active substances released by basophils. They also release eosinophil peroxidase (EPO) and eosinophil cationic protein (ECP), and sustained exposure to these proteins may trigger an autoimmune response [52]. It is worth noting that eosinophilic inflammatory infiltrates indicate high clinical activity, which means more severe and vigorous clinical manifestations of CU [53, 54]. Besides that, in patients with metabolic syndrome, urticaria activity scores and serum levels of ECP, tumor necrosis factor-α (TNF-α), and complement were higher [55]. In CSU, vascular endothelial cells and eosinophils may act as TF-expressing cells, thereby activating the exogenous clotting pathways [56]. Blood eosinophilia is associated with positive autoserum skin tests and basophilic histamine release tests, low total IgE, and high levels of C-reactive protein and Immunoglobulin G (IgG)-antithyroid peroxidase in CSU [57].

Neutrophils and other immune cells

In delayed IgE-mediated hypersensitivity reactions, neutrophils are suggested to function as antigen-presenting cells and are linked to the upregulation of calcitonin gene-related peptide (CGRP) and vascular endothelial growth factor (VEGF) in CSU [58]. It’s hypothesized that B-cell receptor (BCR) signaling plays a vital role in CSU by fostering the generation of autoreactive B cells and the production of autoantibodies. And the proportion of immature CD4+ T cells is lower [59]. Mice with CSU exhibited elevated CD8+ expression and an increased CD4+/CD8+ ratio [60]. Th17 cells and cytokines such as interleukin-17 (IL-17) and interleukin-21 (IL-21), are associated with more severe CSU symptoms, while a decrease in regulatory T (Treg) cells and their cytokines, including transforming growth factor-beta 1 (TGF-β1) and interleukin-35 (IL-35), has also been observed [61]. Furthermore, a significant upregulation of bradykinin receptor 1 (BR1) expression has been observed in lymphocyte subsets, including CD3+ T cells, CD4+ T cells, and CD8+ T cells, in patients with CSU [62]. Bruton tyrosine kinase (BTK) is crucial to both FcεRI and BCR signaling pathways [63]. The predominant M2 type macrophage population in CSU skin lesions contributes to the maintenance of a Th2-mediated inflammatory environment, thus fostering the development of CSU [64].

Fig. 1
figure 1

Aberrant activation and interaction of immune cells in CSU. The figure was created using Figdraw (ID: WPASA0bd00). MCs are activated, leading to the release of inflammatory mediators that initiate pruritus. Basophils contribute to pruritus via Th2 responses and the release of pruritic mediators. Eosinophils, linked to severe urticaria, work to suppress basophils. Additionally, neutrophils, T cells and other immune cells, including Th17 cells and B cells, are implicated in the pathophysiology of CSU. The condition is characterized by elevated inflammatory markers and dysregulation of immune cells, with M2 macrophages fostering a Th2-dominant environment

Fig. 2
figure 2

Abnormal activation of immune cells in CIndU. The figure was created using Figdraw (ID: WUUTA43559). MC degranulation and highly active FcεRI on the surface of basophils are involved in the development of CIndU

Immunity-related animal models of CU might apply or differ in human conditions

The current CU models are summarized in Tables 1 and 2. Ovalbumin (OVA)-induced CSU models are widely used. Some are designed to mimic key CSU features like wheal formation, MC degranulation, and inflammatory cytokine release. CIndU models are still limited, with reports primarily focusing on contact urticaria models induced by trimellitic anhydride (TMA), while models for other subtypes remain scarce.

Animal model of CSU

A CSU mouse model, created through intradermal OVA IgE antiserum injection, replicates CSU symptoms like itching and wheals, confirmed by hematoxylin-eosin staining [65]. Another model uses intraperitoneal OVA and aluminum hydroxide to induce an IgE response, causing wheals and pruritus similar to human CSU [66]. However, not all models exhibit wheals, which may limit their broader application. For instance, Janus kinase (JAK)/ signal transducer and activator of transcription (STAT) pathway gene-silencing reduces inflammation but lacks wheal formation, limiting its relevance​ [67]. The chronic autoimmune urticaria (CAU) mouse model, a CSU subset, uses OVA sensitization and anti-OVA antiserum to mimic chronic inflammation and MC activation [68].

A common model uses subcutaneous histamine injection to trigger wheal-and-flare reactions, simulating MC degranulation and increased vascular permeability [69]. It also shows eosinophil infiltration, itching, and the roles of interleukin-9 (IL-9) and interleukin-10 (IL-10) through the JAK/STAT pathway, mainly emphasizing cytokine regulation [60]. Since wheal formation is crucial for diagnosing CSU, models lacking transient edema as a main feature should be carefully evaluated for CSU research.

Table 1 The immunity-related animal models of CSU

Animal model of CIndU

Applying TMA to mice skin induces an IgE response, leading to MC degranulation and ear swelling, mimicking contact urticaria [70, 71]. Immunologic contact urticaria (ICU) mice develop transient wheals on their skin [72]. The ICU mouse model is also created by injecting 2,4-Dinitrophenol (DNP)-IgE antibody and applying 2,4-dinitrofluorobenzene (DNFB) to trigger an IgE-mediated allergic reaction, activating MCs and causing wheals and itching [39]. These reports only mention information about wheals in about half of the cases. Due to the limited number of reports currently available, it is difficult to assess whether this model can effectively reflect clinical manifestations.

Table 2 The immunity-related animal models of CIndU

Neurological mechanisms and sensitization

Association between neuronal excitation and immune cell in CU

Recent studies have increasingly demonstrated that CU, similar to other type 2 inflammatory skin diseases like atopic dermatitis, involves an interaction between immune cells and sensory neurons that worsens the condition. In CU, immune cells including MCs, eosinophils, and T/B lymphocytes release cytokines such as IL-4, IL-13, interleukin-33 (IL-33), and IL-31, which regulate chronic itch by acting on sensory neurons or modifying their sensitivity to pruritogens [73, 74]. MCs release IL-33 upon activation, leading to the amplification of histamine-induced itch through an IL-13-dependent mechanism in CU [75]. CU may also involve central sensitization. Chronic itching and skin inflammation in CU can alter the spinal cord and brain, increasing excitability and weakening inhibitory circuits, thus greatly intensifying itch perception.

Central sensitization and peripheral sensitization

Wheals and/or angioedema represent cutaneous responses elicited by inflammatory mediators, whereas pruritus is induced when these mediators interact with sensory nerve endings. Itch signals are conveyed by unmyelinated C-fibers to the dorsal root ganglia, subsequently relayed to the dorsal horn of the spinal cord, and ultimately transmitted to the brain [76]. Persistent itching triggers alterations in the spinal cord and brain. Specifically, within the spinal cord, the constant bombardment of itching stimuli can heighten the excitability of dorsal horn neuron [77, 78]. This encompasses increased activity within itch-inducing neural pathways, impairment of inhibitory circuits within the spinal cord, and diminished effectiveness of inhibitory pathways originating from higher brain regions [79, 80]. The phenomenon known as central sensitization significantly amplifies and extends the responses to pruritic stimuli. Additionally, pruritic stimuli can sensitize peripheral nerve fibers, particularly the C-fibers responsible for transmitting itch sensations, a process termed peripheral sensitization. Sensory nerve endings exhibit increased responsiveness to pruritogenic mediators such as histamine and proteases, leading to enhanced neuronal excitability and intensified local inflammation [81,82,83]. This, in turn, amplifies and prolongs the sensation of itch. The worsening of itch leads to increased scratching behavior, resulting in mechanical damage to the skin. The degranulation of immune cells and the subsequent release of inflammatory mediators are facilitated, leading to capillary dilation and increased permeability. These changes exacerbate the swelling associated with wheals and angioedema.

Itch transmission and modulation in the central nervous system

The substances released by immune cells and sensory nerve endings are the primary contributors to the chemical itch in CU, while scratching further induces mechanical itching. In chemical pruritus, keratinocytes communicate directly with sensory neurons through synaptic-like contacts via soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-dependent vesicle release mechanisms, thereby activating sensory neurons [84]. Activation of D1/D5 dopamine receptors and metabotropic glutamate receptor 5 (mGluR5) in the spinal cord can induce non-Hebbian long-term potentiation at sensory synapses without the strict requirement of synchronized presynaptic and postsynaptic activity, amplifying pain and itch signals transmitted to the brain [85]. Mas-related G protein-coupled receptor A3 (MrgprA3)+ neurons are a specialized subpopulation of sensory neurons located in the dorsal root ganglia and trigeminal ganglia by responding to various pruritogens, including both histamine-dependent and histamine-independent stimuli. In chronic itch conditions, MrgprA3+ neurons become more sensitive and excitable, with upregulated ion channels and receptors driving the persistence and severity of the itch [86]. Gastrin-releasing peptide (GRP) neurons receive direct input from MrgprA3+ pruritoceptors [87]. Gastrin-releasing peptide receptor (GRPR)-expressing neurons in the spinal cord form disynaptic connections with glutamatergic spinal projection neurons, mediating itch [88]. Spinal GRPR+ neurons receive inhibitory synaptic inputs from local galanin + gamma-aminobutyric acid (GABA)ergic neurons and long-range neurons from the rostral ventromedial medulla (RVM), which together regulate the transmission of itch signals and dynamically modulate itch perception at the spinal level [89]. Microglia promote chronic itch by producing interleukin-1β (IL-1β) through the activation of the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome and subsequently activating GRPR+ neurons via the type 1 IL-1 receptor (IL-1R1) [90]. Hypothalamic orexin neurons alleviate pain and exacerbate itch by activating periaqueductal gay (PAG) neurons through orexin-dependent and independent pathways, respectively [91]. Activating cannabinoid receptor type 1 (CB1) expressed on glutamatergic neurons and downregulating CB1 on GABAergic neurons in the ventrolateral PAG to alleviate chronic itch leads to the release of 5-hydroxytryptamine (5-HT) in the rostral ventromedial medulla and a reduction in GRPR signaling in the spinal cord [92]. Locus coeruleus (LC) noradrenergic (NAergic) neurons facilitate inhibitory synaptic inputs through activation of α1A-adrenergic receptors (α1AAR) on inhibitory interneurons that project onto GRPR+ neurons [93].

In addition to chemical pruritus, CU also involves mechanical pruritus. In the spinal cord, urocortin 3 (Ucn3)+ excitatory interneurons specifically mediate and regulate mechanical itch by receiving inputs from Toll-like receptor 5+ Aβ low-threshold mechanoreceptors (LTMRs). These Ucn3 + neurons are modulated by feedforward inhibition from neuropeptide Y (NPY)+ inhibitory interneurons [94]. The inhibitory effect of NPY+ interneurons may involve the activation of the neuropeptide Y1 receptor (NPY1R) [95]. In chronic itch, the downregulation of Nav1.6 channels in NPY+ neurons reduces inhibition of Ucn3+ neurons, leading to excessive transmission of mechanical itch signals, while the excitability of NPY-expressing spinal neurons is significantly diminished during persistent itch [96]. Ucn3+ neurons transmit mechanical itch signals to sensory brain regions, particularly the parabrachial nucleus (PBN), via spinoparabrachial projection neurons that express calcitonin receptor like receptor (Calcrl), making synapses with PBN neurons which express forkhead box protein P2 (FoxP2) [97]. The transmission of itch signal from skin to brain is shown in Fig. 3.

Fig. 3
figure 3

Itch Transmission and Modulation in the Nervous System from Peripheral to Central Pathways. The figure was created using Figdraw (ID: PUSUA074f4). In chronic itch, keratinocytes communicate directly with sensory neurons through SNARE-dependent vesicle release, triggering itch signals. MrgprA3+ neurons respond to pruritogens, while GRPR+ neurons mediate itch via disynaptic connections. Ucn3+ neurons specifically transmit mechanical itch signals from TLR5+ Aβ mechanoreceptors to the PBN, regulated by NPY+ inhibitory neurons. Chronic itch sensitization involves reduced inhibition of Ucn3+ neurons due to downregulated Nav1.6 channels in NPY+ neurons, leading to excessive itch signal transmission to sensory brain regions

Neurons interact bidirectionally with immune cells in chronic itch

The skin functions as both a physical barrier and an active player in neuro-immunological interactions. Neuropeptides and neurotransmitters from neurons regulate immune cells, which in turn activate neurons through inflammatory mediators [98, 99]. Sensory neurons, including those expressing transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential ankyrin 1 (TRPA1), participate in a bidirectional interaction with immune cells through the expression of immune receptors and the release of neuropeptides [100]. SP, released by sensory nerves in the skin, causes wheals and itching by promoting vasodilation, increasing vascular permeability, transmitting itch signals, and triggering MCs to release histamine and prostaglandin D2 (PGD2) [101]. Activation of MRGPRX2 by the neuropeptide SP is implicated in CU [102, 103]. Upon binding to the H1 receptor (H1R), histamine activates the TRPV1 channel on sensory neurons, resulting in nerve discharge signals that produce the sensation of itching. PGD2, through activation of the chemoattractant receptor-homologous molecule expressed on Th2 cells receptor (CRTH2R), directly influences sensory neurons, augmenting their excitability and consequently enhancing pain sensitivity. They forms a feedback loop, further amplifying neurogenic inflammation and itch in chronic skin conditions [104]. In CSU, there is a significant increase in circulating levels of SP related to the severity of the disease, activating MCs and stimulating the degranulation of basophilic granulocytes [105]. These effects are mainly achieved through the neurokinin-1 receptor (NK1R). However, there are also reports that SP values were not correlated with the severity of urticaria or angioedema [106]. In addition, sensory nerves release CGRP, contributing to vasodilation and inflammation. Lesional skin in CSU contained significantly more CGRP+ cells than non-lesional skin [107]. Although the neuroimmune mechanisms in CU require further exploration, the interaction between immune cells and the nervous system has been demonstrated in certain dermatosis models. interleukin-27 (IL-27) promotes neuroimmune interactions and exacerbates chronic itch by increasing the expression of bone marrow stromal antigen 2 (BST2) in sensory neurons and keratinocytes and upregulating the activity of PAR2 [108]. Eosinophils promote increased neurite branching in sensory neurons by releasing non- nerve growth factor (NGF)-dependent factors, thereby enhancing nerve density [109]. Basophils mediate acute itch by releasing leukotriene C4 (LTC4), which activates cysteinyl leukotriene receptor 2 (CysLTR2) on sensory neurons, triggering the itch sensation via TRPV1 and TRPA1 calcium channels [110, 111]. Additionally, basophils upregulate tetrahydrobiopterin (BH4), enhancing the release of itch mediators like histamine and serotonin from MCs, while directly activating TRPA1 channels, further intensifying the sensation of itch [112]. These communications are indicated in Fig. 4.

Fig. 4
figure 4

Bidirectional Interaction Between Sensory Neurons and Immune Cells in CU. The figure was created using Figdraw (ID: SYAPT61556). Sensory neurons interact bidirectionally with immune cells, releasing neuropeptides and activating receptors such as TRPV1 and TRPA1, thereby enhancing neurogenic inflammation and itch responses

Neuro-immuity-related animal models of CU might apply or differ in human conditions

Injecting SP subcutaneously into mice induces MC degranulation, histamine release, and an inflammatory response, effectively simulating CU symptoms in humans [113]. The strength lies in replicating key CU processes, especially MC-mediated inflammation, while also revealing the connection between SP and the Src kinase signaling pathway. It further demonstrated the potential of paeonol, a natural compound, in inhibiting MC activation, offering clinical application potential. However, the model focuses heavily on the MRGPRX2 pathway, overlooking other immune mechanisms that may contribute to CU. Additionally, it induces symptoms like wheals and itching through SP-induced MC degranulation in Balb/c mice [114]. Another similar model mimics CSU by triggering both IgE-mediated and pseudo-allergic reactions, leading to MC degranulation and inflammatory responses [66]. These highlight the interaction between SP and MCs, but its effects on eosinophils, basophils, and mesenchymal cells in animal models need further study. These SP-related neuroimmune CU models are listed in Table 3.

Table 3 Neuroimmuity-related animal models of CU

Psychological factors related to CU

Mind-body influences are bidirectional [115]. CU patients frequently experience elevated levels of psychological disorders [116,117,118]. The repeated attacks of skin diseases also bring huge pressure to patients, leading to more depression, impatience, tension, and anxiety. Numerous mediators enhance sensory innervation, stimulate the production of additional pruritogenic agents, sustain neurogenic inflammation, and reduce the threshold for itch perception [119]. However, the application of relevant animal models has not been reported.

Current states of psychological factors linked to CU

Psychological factors are internal mental processes affecting emotions, thoughts, and behaviors, including stress and emotional responses. While stress causes CIndU, CSU is also associated with psychiatric comorbidities [120]. Psychological and emotional stress may also precede the onset of CSU [121]. Nearly 16% of CU patients suffered from mental disorders [122]. They exhibit significantly poorer mental and physical health, with increased depression, anxiety, and sleep issues [123, 124]. Patients with CU, especially CSU, have a higher prevalence of psychiatric disorders and medication use, with increased risk and severity of depression and anxiety correlating with the severity of urticaria [117, 125,126,127]. CSU is more strongly associated with anxiety in individuals aged 18–29 and those with high socioeconomic status, while the link between CSU and depression is strongest in the 50–69 age group and among those with low socioeconomic status [128]. The urticaria control test moderately negatively correlates with quality-of-life scores, the Patient Health Questionnaire-9, and the Beirut Distress Scale-22. Patients with the lowest scores experience the greatest impact on quality of life and depression [129]. Therefore, psychological support and patient education are as important as traditional medication in controlling disease activity and prolonging remission [130].

Current state of psychological factors in CU models

There are no widely reported CU models that specifically incorporate psychological stress. Instead, existing studies primarily focus on psychological stress models in general. The Chronic Mild Stress model subjects animals to mild stressors, mimicking human chronic stress, and is useful for examining behavioral and neurochemical changes. It reveals stress-induced dysfunctions similar to those in humans, such as altered reward processing and stress hormone regulation [131]. However, translating these findings to humans is limited, as the models often use simplified stimuli. In humans, psychological issues like anxiety, depression, and stress in CU are tied to cognitive and emotional aspects such as self-esteem, peer interactions, and emotional regulation, which are hard to replicate in animal models.

Some stress-related skin disease models may offer valuable insights for developing stress-related CU models. Chronic allergic contact dermatitis was induced through repeated sensitizing agent applications and social isolation to simulate the interaction between psychological stress and skin inflammation [132]. Repeated immobilization stress and social isolation stress have been used in atopic dermatitis research to explore the link between stress and its development [133, 134].

Interaction between immune, neurological, and psychological factors

During embryonic development, both the skin and the nervous system originate from the ectoderm, underscoring their interconnected roles in responding to environmental stimuli. The interaction between the nervous and immune systems is implicated in dermatological conditions, particularly CU, where neural factors are increasingly associated with depression, anxiety, and pruritus. These are shown in Fig. 5.

Neural mechanisms of depression, anxiety, and itch in CU

In CU, the prefrontal cortex, cerebellum, and thalamus are implicated in depression and anxiety. The intensity of serotonin transporter protein expression in CSU patients was not significantly associated with the severity of depression, but was significantly associated with the severity of anxiety [135]. The dorsolateral prefrontal cortex is crucial for emotional regulation and cognitive control, while the cerebellum and thalamus, beyond motor functions, influence emotional processing and mood disorders in CSU [136,137,138,139].

Research on the neural mechanisms of depression, anxiety, and itching in CU is still limited. CU is also a pruritic skin disease, and other pruritic mechanisms may provide valuable insights for exploring the neural mechanisms underlying depression, anxiety, and itching in CU. Itch signals move from the PBN to the thalamus and somatosensory cortex, mapping the location of itch and linking it to emotional and reward systems [140]. Neurons expressing CGRP in the lateral external subdivision of the PBN are activated by various threat stimuli and transmit this information to the laterocapsular subdivision of the central amygdala (CeA), modulating affective itch and scratching behavior [141]. Chronic itch and addiction share neural circuits, with both activating the dopamine system in brain reward and motivation pathways, such as the ventral tegmental area (VTA) and nucleus accumbents (NAc), making scratching behavior similar to addictive behavior and forming a hard-to-control vicious cycle [142]. Brain structures related to emotion, such as the limbic system and periaqueductal gray, which modulate itch through descending facilitation, are crucial in stress-induced itch [143]. Activation of the amygdala and hippocampus often occurs concurrently in itch studies, suggesting the role of past itch experiences in itch-related anxiety [144].

Psychosomatic factors affect immune cells through the nervous system

Under stress, various central nervous system loci exhibit plasticity and undergo remodeling, mediated by increased glutamatergic and noradrenergic activity, cytokines, and glucocorticoids, regulated by the HPA axis and the brainstem LC-norepinephrine (NE)/ssympathetic nervous/sympatho-adrenomedullary systems (SNS-SAM) systems [145]. Interleukin-6 (IL-6), interleukin-1 (IL-1) and interferon-γ are activated, and corticosteroid-releasing hormone (CRH)-proopiomelanocortin-adrenocorticotropic hormone (ACTH)-corticosteroid axis is excited [146]. Furthermore, chronic stress appears to increase nerve fiber density in the dermis, MCs, NGF, and CGRP [147]. This interaction exacerbates symptoms and promoting the persistence of CU [148].

HPA axis and hormonal regulation in stress and inflammation

The hypothalamus releases CRH, stimulating the anterior pituitary to secrete ACTH. Then the adrenal glands are prompted to produce cortisol, a primary stress hormone. While cortisol regulates the immune system and has anti-inflammatory effects under normal conditions, the body may develop a tolerance to high cortisol levels, leading to an overactivation of the immune system and increased inflammatory responses [149,150,151]. Both anxiety and depressive disorders influence cortisol responses to stress [152]. Inhibiting cellular retinoic acid-binding protein 1 within the hypothalamus and pituitary glands attenuates acute stress-induced anxiety-like behaviors and markedly decreases corticosterone concentrations [153]. Adrenaline and NE are released from the adrenal medulla, causing characteristic vasoconstriction and MC degranulation [154]. Stress-induced MC degranulation depends on CRH but may also involve the role of SP and neurotensin [155]. CRH also causes blood vessel dilation in human skin through an MC-dependent pathway [156]. Moreover, both corticotropin-releasing factor (CRF) and ACTH have been confirmed to activate basophils [157]. In addition, anxious depression was associated with a decreased basophil subfraction [158].

Autonomic nervous system in emotion regulation, immune response, and CU

The activity of the ANS is fundamental in the regulation of emotion and motivated behavior [159]. The potential facilitative role of the SNS is promoting hostile reactivity and emotion-driven impulsivity in individuals with higher levels of neuroticism [160]. Immune function may be impacted in various conditions marked by heightened sympathetic activity, with these changes potentially linked to dysregulation of CRH in the brain [161]. Additionally, autonomic nerves release neuromediators such as adrenaline and norepinephrine that communicate with both innate and adaptive immune cells, activating specific receptors on numerous target cells in the skin [162, 163]. In CU, activating the SNS stimulates immune cells, especially MCs, releasing inflammatory mediators [164, 165]. The parasympathetic nervous system, particularly through the vagus nerve, influences immune responses by releasing neurotransmitters like acetylcholine. Cholinergic urticaria is a common type of CIndU that presents with pruritic wheals and angioedema triggered by perspiration. The diminished expression of acetylcholine receptor M3 and acetylcholinesterase in individuals with impaired sweating may be either the etiology or a resultant factor of cholinergic urticaria [166].

In addition, the dysregulation of the stress axis in the presence of heightened sympathetic tone and diminished parasympathetic activity, may exacerbate inflammation through direct impacts on brain regions essential for fear and anxiety regulation [167]. Anxiety and depression also cause dysfunction of the blood-brain barrier, which is maked by increased permeability related to inflammation [168].

Neuro-cutaneous interactions of embryonic origins and implications for skin disease modeling

The embryonic origins of the skin and nervous system are intricately linked, tracing back to the early stages of embryonic development. Both the skin and the nervous system originate from the ectoderm, the outermost of the three primary germ layers formed during early embryogenesis [169]. This common origin underlies their integrated roles in sensing and responding to environmental stimuli. The intrinsic transcription factors, such as Neurogenin 1 and Runx1, and extrinsic neurotrophic factors including NGF and glial cell derived neurotrophic factor, work together to drive the differentiation of pain and itch receptors from common embryonic precursor cells into distinct functional subtypes [170]. The immune system, while primarily arising from the mesoderm, interacts extensively with both the skin and nervous system to modulate responses to injury, infection, and inflammation [171]. Sensory neurons can activate reflex arcs to induce anticipatory immune responses in adjacent tissues, priming them to better defend against potential infections [172]. As these balances are disrupted, the likelihood of itching, edema, and hives increases.

By utilizing advanced 3D imaging techniques, an unprecedented and detailed cellular map of early human development is provided [173]. Integrating components of the neuro-immune-cutaneous system into the design of human skin equivalents allows for more accurate modeling of physiological skin responses, particularly in the study of skin diseases involving neuro-immune interactions [174]. The development of tissue-engineered innervated skin models offers a more precise and physiologically relevant platform for studying neuro-cutaneous disorders, improving drug screening, disease mechanism understanding, and the creation of therapeutic interventions [175].

Fig. 5
figure 5

Emotional abnormalities interact with skin itching. The figure was created using Figdraw (ID: RSSTAad4de). Anxiety, depression, and stress are correlated with the development of CU. They overstimulate the HPA axis and the brainstem LC-NE/ SNS-SAM systems, resulting in the release of CRH, ACTH, cortisol, LC, adrenaline, NE, acetylcholine, and other substances. These secretions activate immune cells to produce cytokines, which can trigger itching. Moreover, the act of scratching elicits pleasure that is stored in memory by various regions of the brain, including the hippocampus, VTA, and amygdala, ultimately impacting behavior by reinforcing the habit of scratching

Discussion and future directions

Limitations of current research and the need for enhanced translational models in CU

In human CU, MCs and basophils are crucial in releasing histamines and other pro-inflammatory mediators, contributing to the characteristic symptoms of wheals, angioedema, and pruritus. However, the immune responses observed in animal models, particularly murine systems, diverge significantly from those in humans. The distribution and density of MCs differ between human and murine skin, resulting in different activation thresholds and responses to stimuli like IgE cross-linking.

Anxiety, depression, and stress are common in human CU, worsening the condition and making it chronic by causing neuroimmune dysregulation that leads to persistent itching and inflammation. Although stress can be simulated in animal models, they fail to capture the complex interplay between psychological stress and immune function seen in humans. Stress-induced activation of the HPA axis and ANS has downstream effects on immune cells, particularly MCs and basophils. This neuroimmune connection is difficult to model in animals, where the emotional and cognitive components of stress differ significantly from humans. Thus, future models should employ human-derived tissues, advanced in vitro systems like organoids, or humanized animal models to replicate human immune responses in CU.

Diagnosis and treatment strategy of CU under the guidance of mind-body model

During the diagnostic process of CU, a comprehensive assessment incorporating neurological, immunological, and psychological biomarkers is essential. Psychological status and stress levels can be evaluated using tools such as the Symptom Checklist-90 and the Perceived Social Support Scale. For patients with high stress or psychological issues, consider using detailed assessments like the Eysenck Personality Questionnaire, the Chronic Urticaria Quality of Life Questionnaire, and stress-specific questionnaires. This comprehensive approach improves diagnostic accuracy and aids in personalized treatment.

Current treatments for CU, such as antihistamines, omalizumab, and cyclosporine, show variable efficacy under psychological stress. Therefore, integrating psychological and physical treatments, such as cognitive-behavioral therapy, relaxation training, and stress management, could be more effective. In addition, considering individual differences in psychological stress responses, treatment should emphasize personalized approaches. Combining different therapeutic agents based on the patient’s specific disease characteristics and comorbidities is emerging as a strategy to improve treatment outcomes and address the complex nature of CU [176].

Complementary and alternative therapies may also be an effective means of alleviating CU symptoms and regulating negative emotions. The patients went through psychological counseling, improving these disorders [177]. Acupuncture is an external therapy in traditional Chinese medicine. Randomized clinical trials has shown that it can produce a greater improvement in the Weekly Urticaria Activity Score, and regulate humoral immunity markers and serum total IgE levels [178, 179]. Its antidepressant-like effects appear to be involved in the inhibition of NLRP3 inflammasome activation and apoptosis in the prefrontal cortex [180]. Moreover, it can partly suppress the neuroinflammation induced by the Toll-like receptor 4 signaling pathway [181]. The combination of acupuncture, pricking, and cupping therapy has shown great efficacy in treating CSU by reducing symptoms and negative emotions, enhancing quality of life and sleep, and balancing Th1/Th2 cytokines [182].

Conclusion

CU is a multifactorial condition involving complex neuroimmune and psychological interactions. Current treatments, while effective for some, often fail to address the full spectrum of contributing factors, particularly psychological comorbidities like anxiety and stress, which exacerbate CU symptoms. The limitations of existing animal models, which cannot fully replicate human immune and neuropsychological responses, hinder the development of more targeted therapies. Future research should focus on creating advanced models that integrate neuroimmune mechanisms and psychological factors, while incorporating complementary therapies such as acupuncture to provide more holistic and effective treatment strategies.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

CU:

Chronic urticaria

IgE:

Immunoglobulin E

MCs:

Mast cells

HPA:

Hypothalamic-pituitary-adrenal

ANS:

Autonomic nervous system

CIndU:

Chronic inducible urticaria

CSU:

Chronic spontaneous urticaria

FcεRI:

High-affinity IgE receptor

MRGPRX2:

MAS-related G protein-coupled receptor X2

TF:

Tissue factor

C5a:

Complement 5a

C5aR:

C5a receptor

KIT:

Stem-cell factor receptor

PAR1:

Protease-activated receptor 1

PAR2:

Protease-activated receptor 2

PI3K:

Phosphoinositide 3-kinase

Akt:

Protein kinase B

NF-κB:

Nuclear factor kappa-B

IL-4:

Interleukin-4

IL-13:

Interleukin-13

IL-31:

Interleukin-31

SP:

Substance P

IL-3:

Interleukin-3

PGE:

Prostaglandin E

EPO:

Eosinophil peroxidase

ECP:

Eosinophil cationic protein

TNF-α:

Tumor necrosis factor-α

CGRP:

Calcitonin gene-related peptide

IgG:

Immunoglobulin G

VEGF:

Vascular endothelial growth factor

BCR:

B-cell receptor

IL-17:

Interleukin-17

IL-21:

Interleukin-21

Treg:

Regulatory T

TGF-β1:

Transforming growth factor-β1

IL-35:

Interleukin-35

BR1:

Bradykinin receptor 1

BTK:

Bruton tyrosine kinase

OVA:

Ovalbumin

TMA:

Trimellitic anhydride

JAK:

Janus kinase

STAT:

Signal transducer and activator of transcription

CAU:

Chronic autoimmune urticaria

IL-9:

Interleukin-9

IL-10:

Interleukin-10

DNP:

2,4-Dinitrophenol

DNFB:

2,4-dinitrofluorobenzene

IL-33:

Interleukin-33

SNARE:

Soluble N-ethylmaleimide-sensitive factor attachment protein receptor

mGluR5:

Metabotropic glutamate receptor 5

MrgprA3:

Mas-related G protein-coupled receptor A3

GRP:

Gastrin-releasing peptide

GRPR:

Gastrin-releasing peptide receptor

GABA:

Gamma-aminobutyric acid

RVM:

Rostral ventromedial medulla

IL-1β:

Interleukin-1β

NLRP3:

NOD-like receptor thermal protein domain associated protein 3

IL-1R1:

Type 1 IL-1 receptor

PAG:

Periaqueductal gray

CB1:

Cannabinoid receptor type 1

5-HT:

5-hydroxytryptamine

LC:

Locus coeruleus

NAergic:

Noradrenergic

α1AAR:

α1A-adrenergic receptors

Ucn3:

Urocortin 3

LTMRs:

Low-threshold mechanoreceptors

NPY:

neuropeptide Y

NPY1R:

Neuropeptide Y1 receptor

PBN:

Parabrachial nucleus

Calcrl:

Calcitonin receptor like receptor

FoxP2:

Forkhead box protein P2

TRPV1:

Transient receptor potential vanilloid 1

TRPA1:

Transient receptor potential ankyrin 1

PGD2:

Prostaglandin D2

H1R:

H1 receptor

CRTH2R:

Chemoattractant receptor-homologous molecule expressed on Th2 cells receptor

NK1R:

Neurokinin-1 receptor

IL-27:

Interleukin-27

BST2:

Bone marrow stromal antigen 2

NGF:

Nerve growth factor

LTC4:

Leukotriene C4

CysLTR2:

Cysteinyl leukotriene receptor 2

BH4:

Tetrahydrobiopterin

NGF:

Nerve growth factor

CeA:

Central amygdala

VTA:

Ventral tegmental area

NAc:

Nucleus accumbens

NE:

Norepinephrine

SNS-SAM:

sympathetic nervous/sympatho-adrenomedullary systems

IL-6:

Interleukin-6

IL-1:

Interleukin-1

CRH:

Corticosteroid-releasing hormone

ACTH:

Adrenocorticotropic hormone

CRF:

Corticotropin-releasing factor

References

  1. Fricke J, Ávila G, Keller T, Weller K, Lau S, Maurer M, et al. Prevalence of chronic urticaria in children and adults across the Globe: systematic review with meta-analysis. Allergy. 2020;75:423–32.

    Article  PubMed  Google Scholar 

  2. Gonçalo M, Gimenéz-Arnau A, Al-Ahmad M, Ben-Shoshan M, Bernstein JA, Ensina LF, et al. The global burden of chronic urticaria for the patient and society. Br J Dermatol. 2021;184:226–36.

    Article  PubMed  Google Scholar 

  3. Kocatürk E, Grattan C. Is chronic urticaria more than skin deep? Clin Transl Allergy. 2019;9:48.

    Article  PubMed  PubMed Central  Google Scholar 

  4. He L, Yi W, Huang X, Long H, Lu Q. Chronic urticaria: advances in Understanding of the disease and clinical management. Clin Rev Allergy Immunol. 2021;61:424–48.

    Article  PubMed  Google Scholar 

  5. Baudy A, Raison-Peyron N, Serrand C, Crépy M-N, Du-Thanh A. Impact of chronic spontaneous or inducible urticaria on occupational activity. Acta Derm Venereol. 2024;104:adv36122.

    Article  PubMed  Google Scholar 

  6. Zuberbier T, Abdul Latiff AH, Abuzakouk M, Aquilina S, Asero R, Baker D, et al. The international EAACI/GA2LEN/EuroGuiDerm/APAAACI guideline for the definition, classification, diagnosis, and management of urticaria. Allergy. 2022;77:734–66.

    Article  PubMed  Google Scholar 

  7. Hon KL, Leung AKC, Ng WGG, Loo SK. Chronic urticaria: an overview of treatment and recent patents. Recent Pat Inflamm Allergy Drug Discov. 2019;13:27–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kolkhir P, Giménez-Arnau AM, Kulthanan K, Peter J, Metz M, Maurer M, Urticaria. Nat Rev Dis Primers. 2022;8:61.

  9. Szymanski K, Schaefer P. Urticaria and angioedema. Prim Care. 2023;50:237–52.

    Article  PubMed  Google Scholar 

  10. Church MK, Kolkhir P, Metz M, Maurer M. The role and relevance of mast cells in urticaria. Immunol Rev. 2018;282:232–47.

    Article  CAS  PubMed  Google Scholar 

  11. Schaefer P. Acute and chronic urticaria: evaluation and treatment. Am Fam Physician. 2017;95:717–24.

    PubMed  Google Scholar 

  12. Iriarte Sotés P, Armisén M, Usero-Bárcena T, Rodriguez Fernández A, Otero Rivas MM, Gonzalez MT, et al. Efficacy and safety of Up-dosing antihistamines in chronic spontaneous urticaria: A systematic review of the literature. J Investig Allergol Clin Immunol. 2021;31:282–91.

    Article  PubMed  Google Scholar 

  13. Giménez-Arnau AM, Manzanares N, Podder I. Recent updates in urticaria. Med Clin (Barc). 2023;161:435–44.

    Article  PubMed  Google Scholar 

  14. Altrichter S, Staubach P, Pasha M, Singh B, Chang AT, Bernstein JA, et al. An open-label, proof-of-concept study of lirentelimab for antihistamine-resistant chronic spontaneous and inducible urticaria. J Allergy Clin Immunol. 2022;149:1683–e16907.

    Article  CAS  PubMed  Google Scholar 

  15. Wedi B. Emerging treatments for chronic urticaria. Expert Opin Investig Drugs. 2022;31:281–90.

    Article  CAS  PubMed  Google Scholar 

  16. Jafferany M, Franca K, Psychodermatology. Basics concepts. Acta Derm Venereol. 2016;96:35–7.

  17. Gieler U, Gieler T, Peters EMJ, Linder D. Skin and Psychosomatics - Psychodermatology today. J Dtsch Dermatol Ges. 2020;18:1280–98.

    PubMed  PubMed Central  Google Scholar 

  18. Konstantinou GN, Konstantinou GN. Psychiatric comorbidity in chronic urticaria patients: a systematic review and meta-analysis. Clin Transl Allergy. 2019;9:42.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Staubach P, Dechene M, Metz M, Magerl M, Siebenhaar F, Weller K, et al. High prevalence of mental disorders and emotional distress in patients with chronic spontaneous urticaria. Acta Derm Venereol. 2011;91:557–61.

    Article  PubMed  Google Scholar 

  20. Bangera A, Singh M, Godse K, Patil S. The incidence of depression and anxiety disorders in spontaneous chronic urticaria patients. Qatar Med J. 2023;2023:16.

    PubMed  PubMed Central  Google Scholar 

  21. Dortas Junior SD, Azizi GG, Moret RN, Bastos Junior RM, Valle SOR. Spiritual well-being and quality of life are impaired in chronic urticaria. Eur Ann Allergy Clin Immunol. 2021;53:221–7.

    Article  CAS  PubMed  Google Scholar 

  22. Pyatilova P, Hackler Y, Aulenbacher F, Asero R, Bauer A, Bizjak M, et al. Non-Skin related symptoms are common in chronic spontaneous urticaria and linked to active and uncontrolled disease: results from the chronic urticaria registry. J Allergy Clin Immunol Pract. 2024;12:1890–e18993.

    Article  PubMed  Google Scholar 

  23. Meixiong J, Anderson M, Limjunyawong N, Sabbagh MF, Hu E, Mack MR, et al. Activation of Mast-Cell-Expressed Mas-Related G-Protein-Coupled receptors drives Non-histaminergic itch. Immunity. 2019;50:1163–e11715.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Asero R, Marzano AV, Ferrucci S, Lorini M, Carbonelli V, Cugno M. Co-occurrence of IgE and IgG autoantibodies in patients with chronic spontaneous urticaria. Clin Exp Immunol. 2020;200:242–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kühn H, Kolkhir P, Babina M, Düll M, Frischbutter S, Fok JS, et al. Mas-related G protein-coupled receptor X2 and its activators in dermatologic allergies. J Allergy Clin Immunol. 2021;147:456–69.

    Article  PubMed  Google Scholar 

  26. Lerner L, Babina M, Zuberbier T, Stevanovic K. Beyond Allergies-Updates on the role of Mas-Related G-Protein-Coupled receptor X2 in chronic urticaria and atopic dermatitis. Cells. 2024;13:220.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Elieh-Ali-Komi D, Metz M, Kolkhir P, Kocatürk E, Scheffel J, Frischbutter S, et al. Chronic urticaria and the pathogenic role of mast cells. Allergol Int. 2023;72:359–68.

    Article  CAS  PubMed  Google Scholar 

  28. Larenas-Linnemann D. Biomarkers of autoimmune chronic spontaneous urticaria. Curr Allergy Asthma Rep. 2023;23:655–64.

    Article  CAS  PubMed  Google Scholar 

  29. Zhou B, Li J, Liu R, Zhu L, Peng C. The role of crosstalk of immune cells in pathogenesis of chronic spontaneous urticaria. Front Immunol. 2022;13:879754.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Matsubara D, Yanase Y, Ishii K, Takahagi S, Tanaka A, Ozawa K, et al. Basophils activation of patients with chronic spontaneous urticaria in response to C5a despite failure to respond to IgE-mediated stimuli. Front Immunol. 2022;13:994823.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mostmans Y, De Smedt K, Feoli F, Waelput W, De Maertelaer V, Olemans C, et al. Elevated cutaneous expression of stem cell factor in chronic spontaneous urticaria: a prospective cohort study. Clin Exp Dermatol. 2024;49:1659–67.

    Article  PubMed  Google Scholar 

  32. Yanase Y, Takahagi S, Ozawa K, Hide M. The role of coagulation and complement factors for mast cell activation in the pathogenesis of chronic spontaneous urticaria. Cells. 2021;10:1759.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Asero R. Mechanisms of Histamine release from mast cells beyond the high affinity IgE receptor in severe chronic spontaneous urticaria. Immunol Lett. 2024;265:1–4.

    Article  CAS  PubMed  Google Scholar 

  34. Carvalho RFda, Nilsson S, Harvima G. Increased mast cell expression of PAR-2 in skin inflammatory diseases and release of IL-8 upon PAR-2 activation. Exp Dermatol. 2010;19:117–22.

    Article  CAS  PubMed  Google Scholar 

  35. Muñoz M, Kiefer LA, Pereira MP, Bizjak M, Maurer M. New insights into chronic inducible urticaria. Curr Allergy Asthma Rep. 2024;24:457–69.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Terhorst-Molawi D, Hawro T, Grekowitz E, Kiefer L, Merchant K, Alvarado D, et al. Anti-KIT antibody, barzolvolimab, reduces skin mast cells and disease activity in chronic inducible urticaria. Allergy. 2023;78:1269–79.

    Article  CAS  PubMed  Google Scholar 

  37. Kulthanan K, Church MK, Grekowitz EM, Hawro T, Kiefer LA, Munprom K, et al. Evidence for Histamine release in chronic inducible urticaria - A systematic review. Front Immunol. 2022;13:901851.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Maltseva N, Borzova E, Fomina D, Bizjak M, Terhorst-Molawi D, Košnik M, et al. Cold urticaria - What we know and what we do not know. Allergy. 2021;76:1077–94.

    Article  PubMed  Google Scholar 

  39. Hu S, Zhang Y, Dang B, Wang Y, Zheng G, Zhang T, et al. Myricetin alleviated Immunologic contact urticaria and mast cell degranulation via the PI3K/Akt/NF-κB pathway. Phytother Res. 2023;37:2024–35.

    Article  CAS  PubMed  Google Scholar 

  40. Hashimoto T, Rosen JD, Sanders KM, Yosipovitch G. Possible roles of basophils in chronic itch. Exp Dermatol. 2019;28:1373–9.

    Article  CAS  PubMed  Google Scholar 

  41. Wiebe D, Limberg MM, Gray N, Raap U. Basophils in pruritic skin diseases. Front Immunol. 2023;14:1213138.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lourenço FD, Azor MH, Santos JC, Prearo E, Maruta CW, Rivitti EA, et al. Activated status of basophils in chronic urticaria leads to interleukin-3 hyper-responsiveness and enhancement of Histamine release induced by anti-IgE stimulus. Br J Dermatol. 2008;158:979–86.

    Article  PubMed  Google Scholar 

  43. Saini SS. Urticaria and basophils. Allergol Int. 2023;72:369–74.

    Article  CAS  PubMed  Google Scholar 

  44. Oda Y, Fukunaga A, Washio K, Imamura S, Hatakeyama M, Ogura K, et al. Low responsiveness of basophils via FcεRI reflects disease activity in chronic spontaneous urticaria. J Allergy Clin Immunol Pract. 2019;7:2835–e28447.

    Article  PubMed  Google Scholar 

  45. Huang AH, Chichester KL, Saini SS. Association of basophil parameters with disease severity and duration in chronic spontaneous urticaria (CSU). J Allergy Clin Immunol Pract. 2020;8:793–e7956.

    Article  PubMed  Google Scholar 

  46. de Montjoye L, Darrigade AS, Giménez-Arnau A, Herman A, Dumoutier L, Baeck M. Correlations between disease activity, autoimmunity and biological parameters in patients with chronic spontaneous urticaria. Eur Ann Allergy Clin Immunol. 2021;53:55–66.

    Article  PubMed  Google Scholar 

  47. Kishimoto I, Ma N, Takimoto-Ito R, Nakashima C, Otsuka A, Walls AF, et al. Decreased peripheral basophil counts in urticaria and mouse model of oxazolone-induced hypersensitivity, the latter suggesting basopenia reflecting migration to skin. Front Immunol. 2022;13:1014924.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen Q, Zhai Z, Xu J, Chen W, Chen S, Zhong H, et al. Basophil CD63 expression in chronic spontaneous urticaria: correlation with allergic sensitization, serum autoreactivity and basophil reactivity. J Eur Acad Dermatol Venereol. 2017;31:463–8.

    Article  PubMed  Google Scholar 

  49. Yasnowsky KM, Dreskin SC, Efaw B, Schoen D, Vedanthan PK, Alam R, et al. Chronic urticaria Sera increase basophil CD203c expression. J Allergy Clin Immunol. 2006;117:1430–4.

    Article  CAS  PubMed  Google Scholar 

  50. Mizuno M, Oda Y, Imamura S, Washio K, Fukumoto T, Fukunaga A. IgE receptor responsiveness of basophils in chronic inducible urticaria. Front Immunol. 2022;13:995596.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Giménez-Arnau AM, Ribas-Llauradó C, Mohammad-Porras N, Deza G, Pujol RM, Gimeno R. IgE and high-affinity IgE receptor in chronic inducible urticaria, pathogenic, and management relevance. Clin Transl Allergy. 2022;12:e12117.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Sánchez J, Sánchez A, Munera M, Garcia E, Lopez J-F, Velásquez-Lopera M, et al. Presence of IgE autoantibodies against eosinophil peroxidase and eosinophil cationic protein in severe chronic spontaneous urticaria and atopic dermatitis. Allergy Asthma Immunol Res. 2021;13:746–61.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Martins CF, Morais KL, Figueroa P, Dias NF, Valente NS, Maruta CW, et al. Histopathological and clinical evaluation of chronic spontaneous urticaria patients with neutrophilic and non-neutrophilic cutaneous infiltrate. Allergol Int. 2018;67:114–8.

    Article  PubMed  Google Scholar 

  54. Marques RZS, Criado RFJ, Machado CDS, Tamanini JM, Mello C, van Speyer B. Correlation between the histopathology of chronic urticaria and its clinical picture. Bras Dermatol. 2016;91:760–3.

    Article  Google Scholar 

  55. Ye Y-M, Jin H-J, Hwang E-K, Nam Y-H, Kim J-H, Shin Y-S, et al. Co-existence of chronic urticaria and metabolic syndrome: clinical implications. Acta Derm Venereol. 2013;93:156–60.

    Article  CAS  PubMed  Google Scholar 

  56. Yanase Y, Takahagi S, Hide M. Chronic spontaneous urticaria and the extrinsic coagulation system. Allergol Int. 2018;67:191–4.

    Article  CAS  PubMed  Google Scholar 

  57. Kolkhir P, Church MK, Altrichter S, Skov PS, Hawro T, Frischbutter S, et al. Eosinopenia, in chronic spontaneous urticaria, is associated with high disease activity, autoimmunity, and poor response to treatment. J Allergy Clin Immunol Pract. 2020;8:318–e3255.

    Article  PubMed  Google Scholar 

  58. Kulthanan K, Chularojanamontri L, Tuchinda P, Buranaporn P, Karoopongse E. Unveiling the role of neutrophils in chronic spontaneous urticaria: beyond mast cells. Asian Pac J Allergy Immunol. 2023;41:179–85.

    CAS  PubMed  Google Scholar 

  59. Peng S, Zhang T, Zhang S, Tang Q, Yan Y, Feng H. Integrated bioinformatics and validation reveal IL1B and its related molecules as potential biomarkers in chronic spontaneous urticaria. Front Immunol. 2022;13:850993.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Feng H, Feng J, Zhang Z, Xu Q, Hu M, Wu Y, et al. Role of IL-9 and IL-10 in the pathogenesis of chronic spontaneous urticaria through the JAK/STAT signalling pathway. Cell Biochem Funct. 2020;38:480–9.

    Article  CAS  PubMed  Google Scholar 

  61. Yang X, Chen L, Wang S, Wu Y, Zhou X, Meng Z. The correlation between Th17/Treg immune dysregulation and the disease severity in chronic spontaneous urticaria patients. Immun Inflamm Dis. 2023;11:e920.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Obtulowicz A, Dubiela P, Dyga W, Migacz-Gruszka K, Mikolajczyk T, Wojas-Pelc A, et al. The role of Bradykinin receptors in the etiopathogenesis of chronic spontaneous urticaria. Med (Kaunas). 2021;57:1133.

    Google Scholar 

  63. Bernstein JA, Maurer M, Saini SS. BTK signaling-a crucial link in the pathophysiology of chronic spontaneous urticaria. J Allergy Clin Immunol. 2024;153:1229–40.

    Article  PubMed  Google Scholar 

  64. Criado RFJ, Criado PR, Pagliari C, Sotto MN, Machado Filho CD, Bianco B. M2 macrophage polarization in chronic spontaneous urticaria refractory to antihistamine treatment. Allergol Int. 2021;70:504–6.

    Article  CAS  PubMed  Google Scholar 

  65. Wang H, Xu Y, Jin M, Yuan W. SELE downregulation suppresses mast cell accumulation to protect against inflammatory response in chronic idiopathic urticaria. Int Arch Allergy Immunol. 2021;182:83–93.

    Article  CAS  PubMed  Google Scholar 

  66. Che D, Zhang T, Zhang T, Zheng Y, Hou Y, Geng S, et al. Clarithromycin-treated chronic spontaneous urticaria with the negative regulation of FcεRΙ and MRGPRX2 activation via CD300f. Int Immunopharmacol. 2022;110:109063.

    Article  CAS  PubMed  Google Scholar 

  67. An Y-P, Yuan R, Wang S-S, Yang S-Q, Zhang Q. Knockdown of miR-155 alleviates skin damage in rats with chronic spontaneous urticaria by modulating the JAK/STAT signaling pathway. Allergy Asthma Clin Immunol. 2024;20:38.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Luo X-Y, Liu Q, Yang H, Tan Q, Gan L-Q, Ren F-L, et al. OSMR gene effect on the pathogenesis of chronic autoimmune urticaria via the JAK/STAT3 pathway. Mol Med. 2018;24:28.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Qu S, Liu Z, Wang B. Down-regulation of Gremlin1 inhibits inflammatory response and vascular permeability in chronic idiopathic urticaria through suppression of TGF-β signaling pathway. Gene. 2020;756:144916.

    Article  CAS  PubMed  Google Scholar 

  70. Hyun SY, Kim E-Y, Kang M, Park JW, Hong K-S, Chung H-M, et al. Embryonic-stem-cell-derived mesenchymal stem cells relieve experimental contact urticaria by regulating the functions of mast cells and T cells. Sci Rep. 2023;13:22694.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Lauerma AI, Fenn B, Maibach HI. Trimellitic anhydride-sensitive mouse as an animal model for contact urticaria. J Appl Toxicol. 1997;17:357–60.

    Article  CAS  PubMed  Google Scholar 

  72. Lv G, Fan J. Silencing ICAM-1 reduces the adhesion of vascular endothelial cells in mice with Immunologic contact urticaria. Gene. 2020;760:144965.

    Article  CAS  PubMed  Google Scholar 

  73. Garcovich S, Maurelli M, Gisondi P, Peris K, Yosipovitch G, Girolomoni G. Pruritus as a distinctive feature of type 2 inflammation. Vaccines (Basel). 2021;9:303.

    Article  CAS  PubMed  Google Scholar 

  74. Kolkhir P, Akdis CA, Akdis M, Bachert C, Bieber T, Canonica GW, et al. Type 2 chronic inflammatory diseases: targets, therapies and unmet needs. Nat Rev Drug Discov. 2023;22:743–67.

    Article  CAS  PubMed  Google Scholar 

  75. Trier AM, Ver Heul AM, Fredman A, Le V, Wang Z, Auyeung K, et al. IL-33 potentiates histaminergic itch. J Allergy Clin Immunol. 2024;153:852–e8593.

    Article  CAS  PubMed  Google Scholar 

  76. Yang T-LB, Kim BS. Pruritus in allergy and immunology. J Allergy Clin Immunol. 2019;144:353–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Vikman KS, Kristensson K, Hill RH. Sensitization of dorsal Horn neurons in a two-compartment cell culture model: wind-up and long-term potentiation-like responses. J Neurosci. 2001;21:RC169.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Fatima M, Ren X, Pan H, Slade HFE, Asmar AJ, Xiong CM, et al. Spinal somatostatin-positive interneurons transmit chemical itch. Pain. 2019;160:1166–74.

    Article  CAS  PubMed  Google Scholar 

  79. Mahmoud O, Oladipo O, Mahmoud RH, Yosipovitch G. Itch: from the skin to the brain - peripheral and central neural sensitization in chronic itch. Front Mol Neurosci. 2023;16:1272230.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Pereira MP, Agelopoulos K, Köllner J, Neufang G, Schmelz M, Ständer S. Selective nerve fibre activation in patients with generalized chronic pruritus: hint for central sensitization?? Acta Derm Venereol. 2019;99:1009–15.

    Article  PubMed  Google Scholar 

  81. Li C, Kim HJ, Back SK, Na HS. Common and discrete mechanisms underlying chronic pain and itch: peripheral and central sensitization. Pflugers Arch. 2021;473:1603–15.

    Article  CAS  PubMed  Google Scholar 

  82. Misery L, Pierre O, Le Gall-Ianotto C, Lebonvallet N, Chernyshov PV, Le Garrec R, et al. Basic mechanisms of itch. J Allergy Clin Immunol. 2023;152:11–23.

    Article  CAS  PubMed  Google Scholar 

  83. Pogatzki-Zahn EM, Pereira MP, Cremer A, Zeidler C, Dreyer T, Riepe C, et al. Peripheral sensitization and loss of descending Inhibition is a hallmark of chronic pruritus. J Invest Dermatol. 2020;140:203–e2114.

    Article  CAS  PubMed  Google Scholar 

  84. Talagas M, Lebonvallet N, Leschiera R, Sinquin G, Elies P, Haftek M, et al. Keratinocytes communicate with sensory neurons via Synaptic-like contacts. Ann Neurol. 2020;88:1205–19.

    Article  CAS  PubMed  Google Scholar 

  85. Li J, Price TJ, Baccei ML. D1/D5 dopamine receptors and mGluR5 jointly enable Non-Hebbian Long-Term potentiation at sensory synapses onto Lamina I spinoparabrachial neurons. J Neurosci. 2022;42:350–61.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Xing Y, Chen J, Hilley H, Steele H, Yang J, Han L. Molecular signature of pruriceptive MrgprA3 + Neurons. J Invest Dermatol. 2020;140:2041–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Albisetti GW, Pagani M, Platonova E, Hösli L, Johannssen HC, Fritschy J-M, et al. Dorsal Horn Gastrin-Releasing peptide expressing neurons transmit spinal itch but not pain signals. J Neurosci. 2019;39:2238–50.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Mu D, Deng J, Liu K-F, Wu Z-Y, Shi Y-F, Guo W-M, et al. A central neural circuit for itch sensation. Science. 2017;357:695–9.

    Article  CAS  PubMed  Google Scholar 

  89. Liu M-Z, Chen X-J, Liang T-Y, Li Q, Wang M, Zhang X-Y, et al. Synaptic control of spinal GRPR + neurons by local and long-range inhibitory inputs. Proc Natl Acad Sci U S A. 2019;116:27011–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Liu X, Wang Y, Zeng Y, Wang D, Wen Y, Fan L, et al. Microglia-neuron interactions promote chronic itch via the NLRP3-IL-1β-GRPR axis. Allergy. 2023;78:1570–84.

    Article  CAS  PubMed  Google Scholar 

  91. Kaneko T, Kuwaki T, Kashiwadani H. Hypothalamic orexinergic neurons modulate pain and itch in an opposite way: pain relief and itch exacerbation. J Physiol Sci. 2022;72:21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Ge W-Q, Zhan-Mu O-Y, Chen C, Zhang H, Wang X-Y, Liu X, et al. Electroacupuncture reduces chronic itch via cannabinoid CB1 receptors in the ventrolateral periaqueductal Gray. Front Pharmacol. 2022;13:931600.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Koga K, Shiraishi Y, Yamagata R, Tozaki-Saitoh H, Shiratori-Hayashi M, Tsuda M. Intrinsic braking role of descending locus coeruleus noradrenergic neurons in acute and chronic itch in mice. Mol Brain. 2020;13:144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Wang Z, Donnelly CR, Ji R-R. Scratching after stroking and poking: A spinal circuit underlying mechanical itch. Neuron. 2019;103:952–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Jakobsson JET, Ma H, Lagerström MC. Neuropeptide Y in itch regulation. Neuropeptides. 2019;78:101976.

    Article  CAS  PubMed  Google Scholar 

  96. Lee H, Graham RD, Melikyan D, Smith B, Mirzakhalili E, Lempka SF, et al. Molecular determinants of mechanical itch sensitization in chronic itch. Front Mol Neurosci. 2022;15:937890.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Follansbee T, Dong X. A tactile twist: decoding the phenomena of mechanical itch and alloknesis. Front Mol Neurosci. 2023;16:1278151.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Wu W, Li J, Chen S, Ouyang S. The airway neuro-immune axis as a therapeutic target in allergic airway diseases. Respir Res. 2024;25:83.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Kabata H, Artis D. Neuro-immune crosstalk and allergic inflammation. J Clin Invest. 2019;129:1475–82.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Tauber M, Wang F, Kim B, Gaudenzio N. Bidirectional sensory neuron-immune interactions: a new vision in the Understanding of allergic inflammation. Curr Opin Immunol. 2021;72:79–86.

    Article  CAS  PubMed  Google Scholar 

  101. Chong AC, Chwa WJ, Ong PY. Aeroallergens in atopic dermatitis and chronic urticaria. Curr Allergy Asthma Rep. 2022;22:67–75.

    Article  CAS  PubMed  Google Scholar 

  102. Ten Voorde W, Akinseye C, Abdisalaam I, Wind S, Klarenbeek N, Bergmans M, et al. Intradermal substance P as a challenge agent in healthy individuals. Clin Transl Sci. 2023;16:1856–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Chaki S, Alkanfari I, Roy S, Amponnawarat A, Hui Y, Oskeritzian CA, et al. Inhibition of Orai Channel Function Regulates Mas-Related G Protein-Coupled Receptor-Mediated Responses in Mast Cells. Front Immunol. 2021;12:803335.Ca SCIASRAAYH et al. O,. Inhibition of Orai Channel Function Regulates Mas-Related G Protein-Coupled Receptor-Mediated Responses in Mast Cells. Frontiers in immunology [Internet]. 2022 [cited 2025 Jan 28];12. Available from: https://pubmed.ncbi.nlm.nih.gov/35126366/

  104. Siiskonen H, Harvima I. Mast cells and sensory nerves contribute to neurogenic inflammation and pruritus in chronic skin inflammation. Front Cell Neurosci. 2019;13:422.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Vena GA, Cassano N, Di Leo E, Calogiuri GF, Nettis E. Focus on the role of substance P in chronic urticaria. Clin Mol Allergy. 2018;16:24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Fadaee J, Khoshkhui M, Emadzadeh M, Hashemy SI, Farid Hosseini R, Jabbari Azad F, et al. Evaluation of serum substance P level in chronic urticaria and correlation with disease severity. Iran J Allergy Asthma Immunol. 2020;19:18–26.

    PubMed  Google Scholar 

  107. Kay AB, Ying S, Ardelean E, Mlynek A, Kita H, Clark P, et al. Calcitonin gene-related peptide and vascular endothelial growth factor are expressed in lesional but not uninvolved skin in chronic spontaneous urticaria. Clin Exp Allergy. 2014;44:1053–60.

    Article  CAS  PubMed  Google Scholar 

  108. Li Y, Chen W, Zhu X, Mei H, Steinhoff M, Buddenkotte J, et al. Neuronal BST2: A pruritic mediator alongside Protease-Activated receptor 2 in the IL-27-Driven itch pathway. J Invest Dermatol. 2024;144:1829–e18424.

    Article  CAS  PubMed  Google Scholar 

  109. Foster EL, Simpson EL, Fredrikson LJ, Lee JJ, Lee NA, Fryer AD, et al. Eosinophils increase neuron branching in human and murine skin and in vitro. PLoS ONE. 2011;6:e22029.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Wang F, Trier AM, Li F, Kim S, Chen Z, Chai JN, et al. A basophil-neuronal axis promotes itch. Cell. 2021;184:422–e44017.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Das M, Leyva-Castillo J-M, Geha RS. Basophil: the cell that itches. J Allergy Clin Immunol. 2021;148:708–9.

    Article  PubMed  Google Scholar 

  112. Zschiebsch K, Fischer C, Wilken-Schmitz A, Geisslinger G, Channon K, Watschinger K, et al. Mast cell tetrahydrobiopterin contributes to itch in mice. J Cell Mol Med. 2019;23:985–1000.

    Article  CAS  PubMed  Google Scholar 

  113. Ding Y, Dang B, Zhang Y, Hu S, Wang Y, Zhao C, et al. Paeonol attenuates substance P-induced urticaria by inhibiting Src kinase phosphorylation in mast cells. Cell Immunol. 2023;388–389:104728.

    Article  PubMed  Google Scholar 

  114. Ding Y, Dang B, Wang Y, Zhao C, An H. Artemisinic acid attenuated symptoms of substance P-induced chronic urticaria in a mice model and mast cell degranulation via Lyn/PLC-p38 signal pathway. Int Immunopharmacol. 2022;113:109437.

    Article  CAS  PubMed  Google Scholar 

  115. Lotti T, Bianchi B, Ghersetich I, Brazzini B, Hercogova J. Can the brain inhibit inflammation generated in the skin? The lesson of gamma-melanocyte-stimulating hormone. Int J Dermatol. 2002;41:311–8.

    CAS  PubMed  Google Scholar 

  116. Zysk W, Trzeciak M. Characterization of chronic urticaria and associated Conditions - A Web-Based survey. Dermatol Pract Concept. 2023;13:e2023056.

    Article  PubMed  PubMed Central  Google Scholar 

  117. Chu CY, Cho YT, Jiang JH, Chang CC, Liao SC, Tang CH. Patients with chronic urticaria have a higher risk of psychiatric disorders: a population-based study. Br J Dermatol. 2020;182:335–41.

    Article  CAS  PubMed  Google Scholar 

  118. Cornillier H, Giraudeau B, Munck S, Hacard F, Jonville-Bera A-P, d’Acremont G, et al. Chronic spontaneous urticaria in children - a systematic review on interventions and comorbidities. Pediatr Allergy Immunol. 2018;29:303–10.

    Article  PubMed  Google Scholar 

  119. Arck P, Paus R. From the brain-skin connection: the neuroendocrine-immune misalliance of stress and itch. Neuroimmunomodulation. 2006;13:347–56.

    Article  CAS  PubMed  Google Scholar 

  120. Huang Y, Xiao Y, Jing D, Li J, Zhang J, Chen X, et al. Association of chronic spontaneous urticaria with anxiety and depression in adolescents: A mediation analysis. Front Psychiatry. 2021;12:655802.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Bansal CJ, Bansal AS. Stress, Pseudoallergens, autoimmunity, infection and inflammation in chronic spontaneous urticaria. Allergy Asthma Clin Immunol. 2019;15:56.

    Article  PubMed  PubMed Central  Google Scholar 

  122. Ghazanfar MN, Sørensen JA, Zhang D, Holgersen NK, Vestergaard C, Thomsen SF. Occurrence and risk factors of mental disorders in patients with chronic urticaria. World Allergy Organ J. 2023;16:100835.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Altınöz AE, Taşkıntuna N, Altınöz ST, Ceran S. A cohort study of the relationship between anger and chronic spontaneous urticaria. Adv Ther. 2014;31:1000–7.

    Article  PubMed  Google Scholar 

  124. Vietri J, Turner SJ, Tian H, Isherwood G, Balp M-M, Gabriel S. Effect of chronic urticaria on US patients: analysis of the National health and wellness survey. Ann Allergy Asthma Immunol. 2015;115:306–11.

    Article  PubMed  Google Scholar 

  125. Choi G-S, Nam Y-H, Park C-S, Kim M-Y, Jo E-J, Park H-K, et al. Anxiety, depression, and stress in Korean patients with chronic urticaria. Korean J Intern Med. 2020;35:1507–16.

    Article  PubMed  PubMed Central  Google Scholar 

  126. Tat TS. Higher levels of depression and anxiety in patients with chronic urticaria. Med Sci Monit. 2019;25:115–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Huang Y, Xiao Y, Zhang X, Li J, Chen X, Shen M. A Meta-Analysis of observational studies on the association of chronic urticaria with symptoms of depression and anxiety. Front Med (Lausanne). 2020;7:39.

    Article  PubMed  Google Scholar 

  128. Tzur Bitan D, Berzin D, Cohen A. The association of chronic spontaneous urticaria (CSU) with anxiety and depression: a nationwide cohort study. Arch Dermatol Res. 2021;313:33–9.

    Article  PubMed  Google Scholar 

  129. Tawil S, Irani C, Kfoury R, Abramian S, Salameh P, Weller K, et al. Association of chronic urticaria with psychological distress: A multicentre Cross-sectional study. Acta Derm Venereol. 2023;103:adv00865.

    Article  PubMed  Google Scholar 

  130. Tomaszewska K, Słodka A, Tarkowski B, Zalewska-Janowska A. Neuro-Immuno-Psychological aspects of chronic urticaria. J Clin Med. 2023;12:3134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Tian X, Russo SJ, Li L. Behavioral animal models and Neural-Circuit framework of depressive disorder. Neurosci Bull. 2024.

  132. Kitagaki H, Hiyama H, Kitazawa T, Shiohara T. Psychological stress with long-standing allergic dermatitis causes psychodermatological conditions in mice. J Invest Dermatol. 2014;134:1561–9.

    Article  CAS  PubMed  Google Scholar 

  133. Huong Nguyen LT, Choi M-J, Shin H-M, Yang I-J. Effect of Sopoongsan on skin inflammation and hyperlocomotion in socially isolated mice with atopic dermatitis. Evid Based Complement Alternat Med. 2022;2022:3323201.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Cho D-E, Hong J-P, Kim Y, Sim JY, Kim HS, Kim S-R, et al. Role of gut-derived bacterial lipopolysaccharide and peripheral TLR4 in immobilization stress-induced itch aggravation in a mouse model of atopic dermatitis. Sci Rep. 2024;14:6263.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Zabolinejad N, Molkara S, Bakhshodeh B, Ghaffari-Nazari H, Khoshkhui M. The expression of serotonin transporter protein in the skin of patients with chronic spontaneous urticaria and its relation with depression and anxiety. Arch Dermatol Res. 2019;311:825–31.

    Article  CAS  PubMed  Google Scholar 

  136. Tupker RA, Rustemeyer T, Frölich M, Babri S, Soliman M, de Haan W, et al. Functional brain alterations in symptomatic dermographism patients-An exploratory magnetoencephalography study. Exp Dermatol. 2024;33:e15023.

    Article  CAS  PubMed  Google Scholar 

  137. Wang Y, Gao D, Cui B, Yu B, Fang J, Wang Z, et al. Increased grey matter volume and associated resting-state functional connectivity in chronic spontaneous urticaria: A structural and functional MRI study. J Neuroradiol. 2021;48:236–42.

    Article  PubMed  Google Scholar 

  138. Wang Y, Fang J, Song P, Bao Y, Song W, Liu J, et al. The dysfunction of the cerebellum and its cerebellum-Reward-Sensorimotor loops in chronic spontaneous urticaria. Cerebellum. 2018;17:507–16.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Zhang L, Zou Z, Yu S, Xiao X, Shi Y, Cao W, et al. Functional connectivity impairment of thalamus-cerebellum-scratching neural circuits in pruritus of chronic spontaneous urticaria. Front Neurosci. 2022;16:1026200.

    Article  PubMed  PubMed Central  Google Scholar 

  140. Dong X, Dong X. Peripheral and central mechanisms of itch. Neuron. 2018;98:482–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Sanders KM, Sakai K, Henry TD, Hashimoto T, Akiyama T. A subpopulation of amygdala neurons mediates the affective component of itch. J Neurosci. 2019;39:3345–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Ishiuji Y. Addiction and the itch-scratch cycle. What Do They Have Common?? Exp Dermatol. 2019;28:1448–54.

    PubMed  Google Scholar 

  143. Golpanian RS, Kim HS, Yosipovitch G. Effects of stress on itch. Clin Ther. 2020;42:745–56.

    Article  CAS  PubMed  Google Scholar 

  144. Sanders KM, Akiyama T. The vicious cycle of itch and anxiety. Neurosci Biobehav Rev. 2018;87:17–26.

    Article  PubMed  PubMed Central  Google Scholar 

  145. Makrygianni EA, Chrousos GP. Extracellular vesicles and the stress system. Neuroendocrinology. 2023;113:120–67.

    Article  CAS  PubMed  Google Scholar 

  146. Pondeljak N, Lugović-Mihić L. Stress-induced interaction of skin immune cells, hormones, and neurotransmitters. Clin Ther. 2020;42:757–70.

    Article  CAS  PubMed  Google Scholar 

  147. Keller JJ. Cutaneous neuropeptides: the missing link between psychological stress and chronic inflammatory skin disease? Arch Dermatol Res. 2023;315:1875–81.

    Article  CAS  PubMed  Google Scholar 

  148. Konstantinou GN, Konstantinou GN. Psychological stress and chronic urticaria: A Neuro-immuno-cutaneous crosstalk. A systematic review of the existing evidence. Clin Ther. 2020;42:771–82.

    Article  CAS  PubMed  Google Scholar 

  149. Hassamal S. Chronic stress, neuroinflammation, and depression: an overview of pathophysiological mechanisms and emerging anti-inflammatories. Front Psychiatry. 2023;14:1130989.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Miller GE, Cohen S, Ritchey AK. Chronic psychological stress and the regulation of pro-inflammatory cytokines: a glucocorticoid-resistance model. Health Psychol. 2002;21:531–41.

    Article  PubMed  Google Scholar 

  151. Hall JMF, Cruser D, Podawiltz A, Mummert DI, Jones H, Mummert ME. Psychological stress and the cutaneous immune response: roles of the HPA Axis and the sympathetic nervous system in atopic dermatitis and psoriasis. Dermatol Res Pract. 2012;2012:403908.

    Article  PubMed  PubMed Central  Google Scholar 

  152. Fiksdal A, Hanlin L, Kuras Y, Gianferante D, Chen X, Thoma MV, et al. Associations between symptoms of depression and anxiety and cortisol responses to and recovery from acute stress. Psychoneuroendocrinology. 2019;102:44–52.

    Article  CAS  PubMed  Google Scholar 

  153. Lin Y-L, Wei C-W, Lerdall TA, Nhieu J, Wei L-N. Crabp1 modulates HPA Axis homeostasis and Anxiety-like behaviors by altering FKBP5 expression. Int J Mol Sci. 2021;22:12240.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Slater KN, Abu-Zahra A, Kartono F. Adrenergic Urticaria: Updated Rev Cureus. 2024;16:e62171.

  155. Singh LK, Pang X, Alexacos N, Letourneau R, Theoharides TC. Acute immobilization stress triggers skin mast cell degranulation via Corticotropin releasing hormone, neurotensin, and substance P: A link to neurogenic skin disorders. Brain Behav Immun. 1999;13:225–39.

    Article  CAS  PubMed  Google Scholar 

  156. Crompton R, Clifton VL, Bisits AT, Read MA, Smith R, Wright IMR. Corticotropin-releasing hormone causes vasodilation in human skin via mast cell-dependent pathways. J Clin Endocrinol Metab. 2003;88:5427–32.

    Article  CAS  PubMed  Google Scholar 

  157. Dyke SM, Carey BS, Kaminski ER. Effect of stress on basophil function in chronic idiopathic urticaria. Clin Exp Allergy. 2008;38:86–92.

    Article  CAS  PubMed  Google Scholar 

  158. Baek JH, Kim H-J, Fava M, Mischoulon D, Papakostas GI, Nierenberg A, et al. Reduced venous blood basophil count and anxious depression in patients with major depressive disorder. Psychiatry Investig. 2016;13:321–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Weissman DG, Mendes WB. Correlation of sympathetic and parasympathetic nervous system activity during rest and acute stress tasks. Int J Psychophysiol. 2021;162:60–8.

    Article  PubMed  PubMed Central  Google Scholar 

  160. Peters JR, Eisenlohr-Moul TA, Walsh EC, Derefinko KJ. Exploring the pathophysiology of emotion-based impulsivity: the roles of the sympathetic nervous system and hostile reactivity. Psychiatry Res. 2018;267:368–75.

    Article  PubMed  PubMed Central  Google Scholar 

  161. Friedman EM, Irwin MR. A role for CRH and the sympathetic nervous system in stress-induced immunosuppression. Ann N Y Acad Sci. 1995;771:396–418.

    Article  CAS  PubMed  Google Scholar 

  162. Voisin T, Bouvier A, Chiu IM. Neuro-immune interactions in allergic diseases: novel targets for therapeutics. Int Immunol. 2017;29:247–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Roosterman D, Goerge T, Schneider SW, Bunnett NW, Steinhoff M. Neuronal control of skin function: the skin as a neuroimmunoendocrine organ. Physiol Rev. 2006;86:1309–79.

    Article  CAS  PubMed  Google Scholar 

  164. Hide M, Francis DM, Grattan CE, Hakimi J, Kochan JP, Greaves MW. Autoantibodies against the high-affinity IgE receptor as a cause of Histamine release in chronic urticaria. N Engl J Med. 1993;328:1599–604.

    Article  CAS  PubMed  Google Scholar 

  165. Kenney MJ, Ganta CK. Autonomic nervous system and immune system interactions. Compr Physiol. 2014;4:1177–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Wang Y, Scheffel J, Vera CA, Liu W, Günzel D, Terhorst-Molawi D, et al. Impaired sweating in patients with cholinergic urticaria is linked to low expression of acetylcholine receptor CHRM3 and acetylcholine esterase in sweat glands. Front Immunol. 2022;13:955161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Michopoulos V, Powers A, Gillespie CF, Ressler KJ, Jovanovic T. Inflammation in Fear- and Anxiety-Based disorders: PTSD, GAD, and beyond. Neuropsychopharmacology. 2017;42:254–70.

    Article  CAS  PubMed  Google Scholar 

  168. Welcome MO. Cellular mechanisms and molecular signaling pathways in stress-induced anxiety, depression, and blood-brain barrier inflammation and leakage. Inflammopharmacology. 2020;28:643–65.

    Article  PubMed  Google Scholar 

  169. Quan X-J, Hassan BA. From skin to nerve: flies, vertebrates and the first helix. Cell Mol Life Sci. 2005;62:2036–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Cranfill SL, Luo W. The development of somatosensory neurons: insights into pain and itch. Curr Top Dev Biol. 2021;142:443–75.

    Article  PubMed  Google Scholar 

  171. Morimoto K, Nakajima K. Role of the immune system in the development of the central nervous system. Front Neurosci. 2019;13:916.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Trier AM, Kim BS. Sensory neurons drive anticipatory immunity. Cell. 2019;178:771–3.

    Article  CAS  PubMed  Google Scholar 

  173. Belle M, Godefroy D, Couly G, Malone SA, Collier F, Giacobini P, et al. Tridimensional visualization and analysis of early human development. Cell. 2017;169:161–e17312.

    Article  CAS  PubMed  Google Scholar 

  174. Vidal Yucha SE, Tamamoto KA, Kaplan DL. The importance of the neuro-immuno-cutaneous system on human skin equivalent design. Cell Prolif. 2019;52:e12677.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Schutte SC, Kadakia F, Davidson S. Skin-Nerve Co-Culture systems for disease modeling and drug discovery. Tissue Eng Part C Methods. 2021;27:89–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Zuberbier T, Ensina LF, Giménez-Arnau A, Grattan C, Kocatürk E, Kulthanan K, et al. Chronic urticaria: unmet needs, emerging drugs, and new perspectives on personalised treatment. Lancet. 2024;404:393–404.

    Article  CAS  PubMed  Google Scholar 

  177. Patella V, Zunno R, Florio G, Palmieri M, Palmieri S, Brancaccio R. Omalizumab improves perceived stress, anxiety, and depression in chronic spontaneous urticaria. J Allergy Clin Immunol Pract. 2021;9:1402–4.

    Article  CAS  PubMed  Google Scholar 

  178. Shi Y, Zheng H, Zhou S, Zheng Q, Zhang L, Xiao X, et al. Efficacy and safety of acupuncture for patients with chronic urticaria: study protocol of a randomized, sham-controlled pilot trial. Trials. 2019;20:326.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Zheng H, Xiao X-J, Shi Y-Z, Zhang L-X, Cao W, Zheng Q-H, et al. Efficacy of acupuncture for chronic spontaneous urticaria: A randomized controlled trial. Ann Intern Med. 2023;176:1617–24.

    Article  PubMed  Google Scholar 

  180. Li X, Wang H, Li C, Wu J, Lu J, Guo J-Y, et al. Acupuncture inhibits NLRP3 inflammasome activation in the prefrontal cortex of a chronic stress rat model of depression. Anat Rec (Hoboken). 2021;304:2470–9.

    Article  CAS  PubMed  Google Scholar 

  181. Jiang H, Long X, Wang Y, Zhang X, Chen L, Yang X, et al. Acupuncture ameliorates Depression-Like behaviors through modulating the neuroinflammation mediated by TLR4 signaling pathway in rats exposed to chronic restraint stress. Mol Neurobiol. 2024;61:2606–19.

    Article  CAS  PubMed  Google Scholar 

  182. Shi Y, Guo H, Du Y, Wang J, Shang Y, Wang Y. Acupuncture combined with pricking and cupping therapy is effective in patients with chronic spontaneous urticaria. Am J Transl Res. 2023;15:1195–203.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Figdraw (www.figdraw.com) for the assistance in creating figures.

Funding

This study was supported by the National Natural Science Foundation of China (Nos. 82274634, 82105009).

Author information

Authors and Affiliations

Authors

Contributions

Shurui Yang and Li Chen made equal contributions and are listed as co-first authors. Shurui Yang and Li Chen: conceptualization. Haiming Zhang, Yanjuan Song, Wenyan Wang, Zhengbo Hu, Siyu Wang, Liuyang Huang, Yayuan Wang and Song Wu: literature search. Shurui Yang, Li Chen, Haiming Zhang, Yanjuan Song, Wenyan Wang, Zhengbo Hu, Siyu Wang, Liuyang Huang, Yayuan Wang and Song Wu: writing—original draft. Rui Chen and Fengxia Liang: writing—review and editing.

Corresponding authors

Correspondence to Rui Chen or Fengxia Liang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, S., Chen, L., Zhang, H. et al. Beyond the itch: the complex interplay of immune, neurological, and psychological factors in chronic urticaria. J Neuroinflammation 22, 75 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-025-03397-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-025-03397-4

Keywords