Next Article in Journal
A Novel PDK1/MEK Dual Inhibitor Induces Cytoprotective Autophagy via the PDK1/Akt Signaling Pathway in Non-Small Cell Lung Cancer
Previous Article in Journal
Solvent System-Guided Extraction of Centaurium spicatum (L.) Fritch Provides Optimized Conditions for the Biological and Chemical Characteristics of the Herbal Extracts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neural Regulation of Innate Immunity in Inflammatory Skin Diseases

1
Department of Dermatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
2
Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2023, 16(2), 246; https://doi.org/10.3390/ph16020246
Submission received: 8 December 2022 / Revised: 22 January 2023 / Accepted: 31 January 2023 / Published: 6 February 2023
(This article belongs to the Section Pharmacology)

Abstract

:
As the largest barrier organ of the body, the skin is highly innervated by peripheral sensory neurons. The major function of these sensory neurons is to transmit sensations of temperature, pain, and itch to elicit protective responses. Inflammatory skin diseases are triggered by the aberrant activation of immune responses. Recently, increasing evidence has shown that the skin peripheral nervous system also acts as a regulator of immune responses, particularly innate immunity, in various skin inflammatory processes. Meanwhile, immune cells in the skin can express receptors that respond to neuropeptides/neurotransmitters, leading to crosstalk between the immune system and nervous system. Herein, we highlight recent advances of such bidirectional neuroimmune interactions in certain inflammatory skin conditions.

1. Introduction

The immune system and the nervous system collaborate to protect the body from environmental threats. As the largest organ in the human body, the skin barrier is the first line to detect and combat various environmental stimuli such as toxins and pathogens [1]. In addition to the physical barrier function, the skin is also an important immune organ. The immune system is composed of two parts: the innate immune system and the adaptive immune system [2]. While the main purpose of innate immune responses is to immediately prevent the spread and movement of foreign pathogens throughout the body, the adaptive immune system builds the second line to defend against non-self elements. Although the skin is usually involved in infectious diseases, inflammatory disorders that are preceded by aberrant immune responses also occur and can significantly impair the quality of life of patients. The nervous system includes the peripheral nervous system (PNS) and central nervous system (CNS). The skin is highly innervated by the PNS, which includes sensory nerves and autonomic nerves [3]. Recently, seminal advances demonstrated that the nervous system interacting with the immune system—emerging as neuroimmunology—plays an important role in inflammatory processes. In this review, we aim to depict the classical themes and highlight updates of neuroimmunology underlying common inflammatory skin disorders.

2. Neuroimmune Crosstalk: A Novel Concept in Skin Inflammation

Inflammation is either caused by infections or aberrant immune responses. Regarding the characteristics of immune cells and mediators, immune responses are mainly classified into type 1, type 2, or type 3 [4]. Type 1 immunity, which is mediated by adaptive T helper (Th) type 1 cells, cytotoxic T cells, group 1 innate lymphoid cells (ILC1s), and natural killer cells, defends against intracellular pathogens and tumor cells through interferon (IFN)-γ production [2]. Adaptive Th2 cells, as well as innate basophils, eosinophils, ILC2s, and mast cells, mediate the type 2 immune responses by producing type 2 effector cytokines, such as interleukin (IL)-4, IL-5, and IL-13 [2]. The type 3 immune response protects against extracellular bacteria and fungi with the production of IL-17 and IL-22 from adaptive Th17 cells and gamma-delta T cells, as well as ILC3s and neutrophils [5,6]. Emerging evidence has demonstrated that immune cells in the skin are capable of releasing various cytokines that could directly act on skin nerve terminals [7,8]. Meanwhile, the skin PNS senses the stimulation and transmits the information to the CNS or nearby efferent neurons, leading to various neural symptoms such as itch and pain [9].
The skin comprises three major layers: epidermis, dermis, and subcutis [10]. The cutaneous immune cells mainly reside in the epidermis and dermis, where they are more likely to encounter environmental invaders. Antigen-presenting cells, including Langerhans cells and dermal dendritic cells (DCs), serve as part of the first line of defense against pathogens. Langerhans cells are often restricted to the epidermis, while dermal DCs stay deeper in the skin, and both of them are found in close proximity to nerve fibers [10,11]. Unlike mast cells that are usually skin-resident in the steady state, other myeloid cells, such as basophils, eosinophils, and neutrophils, are commonly found in the dermis in skin inflammatory conditions [12,13]. There are also ample T cells in the epidermis (mostly cytotoxic CD8+ T cells) and dermis (mostly CD4+ Th cells) [14].
Cutaneous sensory nerves, which derive from cell bodies in the dorsal root ganglia (DRG) and trigeminal ganglia, can be divided into three subtypes of nerve endings, which include Aβ, Aδ, and C nerve fibers based on the diameter and speed of transmission [15]. These sensory nerves are densely distributed throughout the dermis and epidermis. They mediate various sensations by encoding signals of pain, itch, temperature, pressure, position, and vibration [3]. Furthermore, these nerve fibers are anatomically close to the functional immune cells in the skin, providing the basis for cutaneous neuroimmune interactions [16,17] (Figure 1). It has been reported that pruritic diseases are closely related to immune cells [18]. For instance, mast cells have been implicated in various types of pruritus due to their capacity to release pruritogens [19]. However, neuropeptides and neurotransmitters released from activated neurons can act on the microvascular cells and resident mast cells to induce mast cell degranulation, which subsequently leads to a physiological reaction such as vasodilation and extravasation of plasma and leukocytes [20,21,22]. Therefore, the skin is a systemic organ that requires a neuroimmune interaction network to maintain its homeostasis.

3. Atopic Dermatitis (AD): A Paradigm of Itch Neuroimmune Mechanisms

AD is a chronic and relapsing skin disorder that is characterized by extensive itch and inflammatory skin lesions [23]. It is preceded by type 2 immune responses [24]. Although the role of the Th2 inflammatory axis, which leads to the immunoglobulin (Ig) E reactivity towards environmental allergens (e.g., pollen, house dust mites), is well established in its mechanisms [25], recent studies have demonstrated the importance of innate immune reactions. Moreover, the advances of itch mechanisms identify an important role of neuroimmune reactions in AD [26].
Histopathology examination has found that the skin lesions in patients with AD contain various innate immune cells that include mast cells, basophils, eosinophils, ILC2s, and DCs, leading to a possible role of innate immunity in AD [23]. Among these cells, mast cells represent a classic neuroimmune paradigm due to their close proximity to sensory nerves. Indeed, antigen recognition by IgE bound to FcεRI on mast cells results in IgE crosslinking and triggers the release of a variety of effector molecules, such as histamine, serotonin, proteases, and various cytokines [19]. These mediators are sufficient to activate sensory neurons and cause itch sensation [27]. Importantly, activated sensory neurons, in turn, could release neuropeptides or transmitters that act on mast cells to form a neuroimmune feedback loop. It has been demonstrated that mast cells have several receptors for neuropeptides, such as substance P (SP), calcitonin-gene-related peptide (CGRP), neuropeptide Y (NPY), and vasoactive intestinal peptide (VIP), expressed on their surface [28]. Decades ago, mast cells were discovered to degranulate in response to nerve growth factor (NGF) by the TrkA tyrosine receptor [29]. Interestingly, mast cells are also a source of NGF [30]. These data suggest that these mast cells and the sensory nervous system have a mutual interaction and thus form a skin–immune–nerve circuit.
More recently, Dong group found that a member of the Mas-related G protein-coupled receptor (MRGPR) family, MRGPRX2 (its mouse orthologue is MrgprB2), is expressed on the human mast cell surface [31,32]. Via binding to cationic molecules such as neuropeptides and host defense peptides, MRGPRX2 encourages mast cells to degranulate and release tryptase, rather than histamine [31]. Since tryptase is also a pruritogen that could evoke itch scratching, these new findings indicate the presence of heterogeneous neuroimmune pathways underlying innate immunity. Despite this, studies on MRGPRX2 in the context AD are still limited. Although mast cells are increased in skin lesions and show signs of degranulation in AD [33], the mechanisms of their activation modes are surprisingly ill-defined. Interestingly, nerve fibers in AD skin lesions show greater positive staining for SP [34], provoking the hypothesis of an intense neural–mast cell communication in AD. Due to the expression of MRGPRX2 in connective tissue mast cells of the skin [35], Wang et al. proposed a possible role of MRGPRX2 in AD: while MRGPRX2-activated mast cells release inflammatory cellular contents that act on sensory nerves, neuropeptides from sensory fibers promote mast cell activity via MRGPRX2 in the skin. In an allergic skin inflammation model induced by house dust mites with cysteine protease activity, Serhan et al. found that transient receptor potential vanilloid 1+ neurons were able to promote mast cell activation via the release of SP, which causes mast cell degranulation by binding to MrgprB2 [36]. Moreover, patients with AD often have Staphylococcus aureus isolated from their skin [37]. Staphylococcus aureus can also directly activate sensory neurons to release SP [38]. Given the activation of MrgprB2 on mast cells is essential for host defense against Staphylococcus aureus [39], these data indicate that mast cells and sensory nerves form an intrinsic and cyclical relationship [40].
Basophils share multiple similar features with tissue-resident mast cells. Increasing discoveries have shown that basophil recruitment to tissues enables them to exhibit unique functions in skin inflammatory disorders. Similar to mast cells, basophils respond rapidly to the IgE-mediated activation by degranulating a variety of pre-stored effector molecules such as histamine, and later leukotrienes, IL-4, and IL-13 [41]. Interestingly, Oetjen et al. discovered that the orchestration of sensory neurons and type 2 cytokines could be the key mechanism of chronic pruritus [7]. They demonstrated that the type 2 cytokines IL-4 and IL-13 could directly activate sensory neurons in both mice and humans. Therefore, basophils may play an important role in chronic itch due to their capacity to release IL-4 and IL-13. Apart from chronic itch, we found that basophils also contribute to acute itch flares in a mast-cell-independent manner in the context of AD [42]. Specifically, the basophil-associated leukotriene C4 and its receptor CysLTR2 on sensory neurons are critically required to transmit this form of IgE-mediated itch. Similar to mast cells, functional MRGPRX2 is also significantly expressed by basophils, and the upregulation of MRGPRX2 is associated with basophil degranulation and CD63 expression [43]. Collectively, these data indicate that basophils may be an important modulator of AD-associated itch.
ILCs are immune cells that belong to the lymphoid lineage but do not express antigen-specific T cell receptors. ILC2s serve crucial functions in various different tissues, but are especially enriched in barrier tissues, such as the lungs, gut, and skin. Due to the specific receptor expression on ILC2s, epithelial-cell-derived cytokines such as IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) are sufficient to activate ILC2s [44,45]. Notably, it has been demonstrated that human AD skin has elevated expression of IL-33, IL-25, and TSLP [46,47,48], indicating a role of ILC2s in AD. Indeed, Kim et al. showed that ILC2s significantly accumulate in skin lesions of patients with AD [49]. Furthermore, those ILC2s stay in close proximity to basophils in the inflammatory dermis. Utilizing an AD murine model mediated by topical MC903 (a calcipotriol analog), authors have demonstrated that basophil-derived IL-4 plays a significant role in promoting ILC2s in the setting of AD-like disease. Following activation, skin ILC2s could constitutively produce IL-5 and IL-13, leading to AD-like pathology and Th2 responses [48,50,51,52]. Collectively, these studies demonstrate that skin ILC2s promote type 2 skin inflammation and coordinately interact with adaptive cells in the process of AD.
The crosstalk between ILC2s and the PNS has been demonstrated in the context of allergic lung inflammation. Cardoso et al. found that murine mucosal ILC2s selectively express neuromedin U (NMU) receptor 1 and co-localize with cholinergic neurons that express NMU [53,54]. Following stimulation with IL-25 and IL-33, NMU induces ILC2 proliferation and activation to produce type 2 cytokines and promote lung inflammation. Nagashima et al. employed a lung helminth infection model and discovered that α-CGRP limits ILC2 proliferation and IL-13 production by antagonizing the action of NMU and IL-33 [55]. However, the production of IL-5 in ILC2s is promoted in the same context. The Locksley group showed that mucosal ILC2s also have mRNA expression of both VIP receptor type 1 and type 2 [56]. Moreover, ILC2s isolated from murine intestine had an elevated IL-5 release with the addition of VIP or activation of VIP receptor type 2 [56], providing another neuroimmune pathway of ILC2 regulation.
Due to the tight bond between ILC2s and basophils, Inclan-Rico et al. re-explored the role of ILC2 in the murine model of lung Nippostrongylus brasiliensis infection [57]. They found that helminth-induced ILC2 responses are exaggerated in the setting of basophil depletion, resulting in increased inflammation and diminished lung function. More importantly, a neuropeptide, neuromedin B (NMB), has similar effects on ILC2s to basophil deficiency. Authors have further shown that the role of basophils in this setting is to enhance ILC2s to increase the expression of the NMB receptor. Although the major source of NMB in this setting remains undefined, these findings suggest that basophils have the capacity to prime ILC2s to respond to neuropeptides, and that NMB might be a potent inhibitor of type 2 inflammation. To date, neuroimmune studies based on ILC2s in the context of AD remain quite few. However, given those previous findings on neuropeptides and ILC2s in other settings of type 2 inflammation (Figure 2), we are optimistic that novel neuroimmune mechanisms of AD will be disclosed in the near future.
DCs are a subset of innate immune cells and have unique functions of antigen uptake and presentation, which are essential for the production of allergen-specific Igs. The distribution of DC subsets depends on the different phases of inflammation [58]. In skin lesions from AD patients, inflammatory epidermal DCs (positive for CD11c and CD206) were markedly shown in central areas of the spongiotic epidermis [59]. These DCs are believed to induce T cell responses and provide a potential target for therapeutics. Interestingly, a recent study conducted by Perner et al. employed an allergen recognition model in which papain was directly injected into murine skin [60]. They further found that SP and its receptor MrgprA1 are critically required for the migration of CD301b+ DCs from the skin to the draining lymph node where they initiate Th2 cell differentiation. In human studies, NPY has been shown to induce the migration of human monocyte-derived immature DCs through its engagement of the NPY Y1 receptor in vitro, resulting in Th2 polarization [61]. Unlike NPY, the effects of VIP on DCs are multifaceted. Immature DCs following the treatment of VIP, both in vivo and in vitro, have increased CD86 expression to promote CD4+ T cell proliferation and to exhibit a Th2 phenotype [62]. In contrast, in the setting of lipopolysaccharide stimulation, VIP reduces CD86 and CD80 expression on DCs and thus leads to an inhibitory effect on T cell proliferation [62]. Brain natriuretic peptide transmits the itch signal in the nervous system. In AD, brain natriuretic peptide was found to be released from sensory nerves that are activated by IL-31 [63]. In turn, brain natriuretic peptide stimulates DCs to release inflammatory factors, which contributes to a positive feedback loop of neuroinflammation [63,64].
Langerhans cells are epidermal-resident DCs and play a key role in the initiation of cutaneous immune responses. Due to the close association between Langerhans cells and nerves, several studies have investigated the neural effects on Langerhans cells. In a murine model of ovalbumin-specific presentation, Ding et al. showed that CGRP enhances Langerhans cell function and Th2 responses by increasing IL-4, CCL17, and CCL22 production and decreasing IFN-γ production [65]. These studies indicate that neuropeptides are regulators in the process of allergen presentation and may open new avenues to help regulate the pathologic process of AD.

4. Psoriasis: Neurogenic Skin Inflammation

Psoriasis is a chronic inflammatory skin disease shaped by genetics, environmental factors, and psychological stress [66]. Both clinical and experimental studies have suggested an involvement of neurogenic components in its pathogenesis [67]. The aberrant type 1/17 immune responses represent a predominant mechanism in psoriasis due to the abundant release of type 1/17 cytokines such as IFN-γ, IL-17A, IL-23, and tumor necrosis factor (TNF)-α [68]. These cytokines lead to keratinocyte proliferation as well as sustained skin inflammation.
Chronic itch is a symptom occurring not only in patients with AD, but also in patients with psoriasis. Although mast cells may not be key to AD-related itch, they partially contribute to the itch mechanisms in psoriasis [67]. Histopathologic examination of skin lesions in patients with psoriasis revealed that, compared with non-pruritic skin tissues, the pruritic areas had more mast cells and degranulated mast cells [69]. Additionally, a rich innervation of nerves and an increase in SP-, CGRP-, and VIP-positive nerve fibers were also shown in the psoriatic skin [10]. In addition, there is a greater frequency of morphological contacts between neurofilament+ nerves and tryptase+ mast cells in psoriatic lesions than in non-lesional skin or normal-looking skin, suggesting a morphologic basis for mast cell–neural interactions in this disease [70]. Notwithstanding this, the effects of antihistamines on psoriasis are often limited, provoking the hypothesis that other inflammatory mediators are involved in this process. Indeed, itch-associated cytokines or pruritogens such as IL-31, TSLP, and tryptase have been found to be elevated in skin lesions at the transcriptomic level [69,71,72], indicating a complex of itch mechanisms in psoriasis.
Neurogenic inflammation is believed to play an important role in psoriasis. The direct evidence is that SP-immunoreactive fibers and SP receptors (neurokinin 1 receptor (NK1R) and MRGPRX2) in the skin of psoriatic patients with itch have a higher expression than those in patients without itch [69,73]. Meanwhile, elevated plasma CGRP levels were observed in psoriatic patients [74]. The receptor of CGRP is also detected in psoriatic skin lesions [75]. It is well established that SP and CGRP are sufficient to induce mast cells to release various cytokines such as IL-1β and TNF-α, which can attract neutrophils to accumulate in the skin [69,76]. Neutrophils can produce antimicrobial peptides such as α-defensins, thus leading to persistent inflammation [77]. Smith et al. conducted an interesting experiment by intradermally injecting CGRP into normal human skin and found a significant infiltration of neutrophils [78]. In addition to SP and CGRP, galanin is another bioactive neuropeptide. One of its receptors, galanin-R3, is expressed on the vascular endothelium. In a murine model of psoriasis-like disease mediated by imiquimod, the lack of galanin-R3 led to a less severe disease phenotype that included delayed neo-vascularization, reduced neutrophil infiltration, and significantly lower levels of proinflammatory cytokines in contrast to the controls [79]. An interesting phenomenon in psoriasis is called the Koebner phenomenon, in which a trauma or wound can cause psoriatic lesions in normal-looking skin. However, its mechanisms remain unclear. NGF appears to be an initial neuropeptide in this process since it is capable of causing the release of other neuropeptides, including SP and CGRP [67]. Meanwhile, elevated expression of NGF and its receptor TrkA was also exhibited in psoriatic lesions [80]. Taken together, these findings suggest that the nervous system likely contributes to skin inflammation in psoriasis (Figure 3).
The direct evidence that sensory nerves, particularly nociceptors, are involved in the pathogenesis of psoriasis-like disease was published in 2014 [81]. In this study, by employing the imiquimod-induced psoriasis murine model, the authors found that the activated transient receptor potential V1+ nerves innervating the skin are required to promote IL-23 production in dermal DCs, which, in turn, can stimulate dermal gamma-delta T cells to secrete IL-17A, IL-17F, and IL-22, resulting in the recruitment of more neutrophils to the skin and excessive keratinocyte proliferation. These findings shed light on how the nervous system affects the reaction of local innate immune cells and thereby contribute to the mechanisms of psoriasis.
ILC3s are defined by their capacity to produce IL-17A and/or IL-22. Similar to Th17 cells, ILC3s depend on the transcription factor RORγt for their development and function [82]. ILC3s are considered to contribute to the pathogenesis of psoriasis [82]. Natural cytotoxicity receptor-positive ILC3s have been found to be increased in psoriatic lesions [83]. Following stimulation with IL-23 and IL-1β, these ILC3s isolated from psoriatic skin lesions can produce IL-22 ex vivo. Although studies on the effects of neuropeptides on ILC3 regulation in the context of psoriasis remain few, some studies have explored neural–ILC3 interactions in the gut. A recent study demonstrated that intestinal ILC3 responses depend on the food-induced expression of the neuropeptide VIP [84]. Intestinal ILC3 had a high expression of VIP receptor 2. Enteric neuron-derived VIP binds to this receptor on ILC3s and causes the production of IL-22, which is critical for the maintenance of homeostasis in the intestinal tract. The deficiency in signaling through VIP receptor 2 increased susceptibility to inflammation-induced gut injury by reducing IL-22 production from ILC3. In contrast, Talbot et al. discovered that VIPergic neurons reduced IL-22 production by CCR6+ILC3s through VIP receptor 2 [85]. They found that the presence of commensal microorganisms upregulates ILC3s, which, in turn, can enhance IL-22 production, and this phenomenon is inhibited by the engagement of VIP receptor 2. Moreover, Yu et al. have discovered that VIP promotes ILC3 recruitment to the intestine through VIP receptor 1 [86]. Although discrepancy exists among different studies, these findings establish scientific clues for future research on the neural–ILC3 axis in psoriasis (Figure 3).
The complex network of DCs in the skin tissue is composed of Langerhans cells, conventional bone-marrow-derived dermal DCs, plasmacytoid DCs, and inflammatory DCs. Via the activation of Toll-like receptors, Langerhans cells purified from normal healthy skin produce high levels of IL-23, which is crucial in the pathogenesis of psoriasis [87,88]. Meanwhile, activated conventional DCs secrete a range of inflammatory cytokines such as IL-12 and IL-23 [89]. Intriguingly, it has been shown that CGRP is required for the infiltration of DCs and T cells into the skin in a psoriasiform murine model [90]. The authors utilized a small molecule to inhibit CGRP and found a reduction in the CD4+ T cell number as well as skin acanthosis. Although α-melanocyte-stimulating hormone has been found to stimulate melanogenesis, which is responsible for the pigmentation of the hair and skin, a study by Matteo et al. found that α-melanocyte-stimulating hormone, binding to MC1R, is sufficient to induce tolerogenic DCs and leads to the proliferation of regulatory T cells and inhibition of Th17 activities [91]. Another study by Ding et al. found that VIP could promote the process of antigen presentation and the secretion of IL-17A and IL-6 from Langerhans cells, indicating a role of VIP in psoriasis-related mechanisms [92].

5. Other Inflammatory Skin Diseases: Neuroimmune Responses Likely Involved

In addition to AD and psoriasis, other cutaneous disorders that may involve neuroimmune interactions at least include chronic spontaneous urticaria (CSU) and bullous pemphigoid. Previously published studies showed that the intradermal injection of SP and VIP induces exaggerated responses in the skin of patients with CSU [93,94]. Moreover, the SP receptor MRGPRX2 has elevated expression levels on mast cells in the context of CSU [95]. Additionally, Zheng et al. found that circulating SP+ and NK1R+ basophils were markedly elevated in CSU in comparison with healthy controls, and SP is capable of causing basophil degranulation and tissue accumulation [96]. In patients with bullous pemphigoid, both NK1R+ cells and SP+ cells were exhibited in the skin [97]. More importantly, both NK1R+ cells and SP+ cells have a positive correlation with the itch severity. Given that most NK1R+ cells are identified as eosinophils, it would be interesting to explore the neuro–eosinophil interactions in bullous pemphigoid in future studies.

6. Conclusions

Emerging evidence has shown that the neuroimmune crosstalk plays a significant role in the regulation of skin inflammation. We highlighted the updates of the neuroimmune interactions underlying AD and psoriasis (Table 1). Although several groups including us have identified novel neuroimmune mechanisms in skin inflammation, a lot of questions remain unclear. For instance, the heterogenous functions of neuropeptides on various immune cells are not fully investigated. Due to the small amount of production of neuron-derived molecules and their rapid action within nerve ending microenvironments, the detection and quantification of these molecules in skin tissues are technically challenging. Beyond neurons, immune cells and keratinocytes could also be a source of neuropeptides, indicating that an autocrine or paracrine mechanism might contribute to the immune–nerve circuit. Therefore, it is necessary to clarify the cellular source and function of various neuropeptides in the skin and other peripheral organs to deeply explore the complex of neuroimmune responses.
In recent decades, an increasing number of novel targeted therapies, especially biologics, have been brought into clinical use for inflammatory skin diseases. Here, we summarize the therapeutic targets and medications that are associated with neuroimmune interactions for skin inflammatory disorders (Table 2). In the future, with more neuroimmune interactions unveiled, more effective treatment options will be introduced to skin inflammatory disorders.

Author Contributions

Writing—original draft preparation, X.H. and F.L.; writing—review and editing, F.W. All authors have read and agreed to the published version of the manuscript.

Funding

The work was funded by the Ministry of Science and Technology of the People’s Republic of China (STI2030-Major Projects 2022ZD0206200), the National Natural Science Foundation of China (82073427 and 82273511), the Fundamental Research Funds for the Central Universities, Sun Yat-sen University (22ykqb03), and Bethune Foundation and National Center for Clinical Medical Research on Skin and Immune Diseases (J202102E015).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank all members of the Wang lab for the helpful comments and discussion.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADatopic dermatitis
CGRPcalcitonin-gene-related peptide
CNScentral nervous system
CSUchronic spontaneous urticaria
DCdendritic cell
IFN-γinterferon-γ
Igimmunoglobulin
ILCinnate lymphoid cell
ILinterleukin
MRGPRMas-related G protein-coupled receptor
NGFnerve growth factor
NK1Rneurokinin 1 receptor
NMBneuromedin B
NMUneuromedin U
NPYneuropeptide Y
PNSperipheral nervous system
SPsubstance P
ThT helper
TNF-αtumor necrosis factor α
TSLPthymic stromal lymphopoietin
VIPvasoactive intestinal peptide

References

  1. Trier, A.M.; Mack, M.R.; Kim, B.S. The Neuroimmune Axis in Skin Sensation, Inflammation, and Immunity. J. Immunol. 2019, 202, 2829–2835. [Google Scholar] [CrossRef] [PubMed]
  2. Abreu, D.; Kim, B.S. Innate Immune Regulation of Dermatitis. Immunol. Allergy Clin. N. Am. 2021, 41, 347–359. [Google Scholar] [CrossRef] [PubMed]
  3. Wang, F.; Kim, B.S. Itch: A Paradigm of Neuroimmune Crosstalk. Immunity 2020, 52, 753–766. [Google Scholar] [CrossRef] [PubMed]
  4. Annunziato, F.; Romagnani, C.; Romagnani, S. The 3 major types of innate and adaptive cell-mediated effector immunity. J. Allergy Clin. Immunol. 2015, 135, 626–635. [Google Scholar] [CrossRef]
  5. Rutz, S.; Eidenschenk, C.; Ouyang, W. IL-22, not simply a Th17 cytokine. Immunol. Rev. 2013, 252, 116–132. [Google Scholar] [CrossRef]
  6. Harrington, L.E.; Hatton, R.D.; Mangan, P.R.; Turner, H.; Murphy, T.L.; Murphy, K.M.; Weaver, C.T. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat. Immunol. 2005, 6, 1123–1132. [Google Scholar] [CrossRef]
  7. Oetjen, L.K.; Mack, M.R.; Feng, J.; Whelan, T.M.; Niu, H.; Guo, C.J.; Chen, S.; Trier, A.M.; Xu, A.Z.; Tripathi, S.V.; et al. Sensory Neurons Co-opt Classical Immune Signaling Pathways to Mediate Chronic Itch. Cell 2017, 171, 217–228.e213. [Google Scholar] [CrossRef]
  8. Cevikbas, F.; Wang, X.; Akiyama, T.; Kempkes, C.; Savinko, T.; Antal, A.; Kukova, G.; Buhl, T.; Ikoma, A.; Buddenkotte, J.; et al. A sensory neuron-expressed IL-31 receptor mediates T helper cell-dependent itch: Involvement of TRPV1 and TRPA1. J. Allergy Clin. Immunol. 2014, 133, 448–460. [Google Scholar] [CrossRef]
  9. Lay, M.; Dong, X. Neural Mechanisms of Itch. Annu. Rev. Neurosci. 2020, 43, 187–205. [Google Scholar] [CrossRef]
  10. Oleszycka, E.; Kwiecien, K.; Kwiecinska, P.; Morytko, A.; Pocalun, N.; Camacho, M.; Brzoza, P.; Zabel, B.A.; Cichy, J. Soluble mediators in the function of the epidermal-immune-neuro unit in the skin. Front. Immunol. 2022, 13, 1003970. [Google Scholar] [CrossRef]
  11. Zhang, S.; Edwards, T.N.; Chaudhri, V.K.; Wu, J.; Cohen, J.A.; Hirai, T.; Rittenhouse, N.; Schmitz, E.G.; Zhou, P.Y.; McNeil, B.D.; et al. Nonpeptidergic neurons suppress mast cells via glutamate to maintain skin homeostasis. Cell 2021, 184, 2151–2166.e16. [Google Scholar] [CrossRef] [PubMed]
  12. Yazdi, A.S.; Rocken, M.; Ghoreschi, K. Cutaneous immunology: Basics and new concepts. Semin. Immunopathol. 2016, 38, 3–10. [Google Scholar] [CrossRef]
  13. Pasparakis, M.; Haase, I.; Nestle, F.O. Mechanisms regulating skin immunity and inflammation. Nat. Rev. Immunol. 2014, 14, 289–301. [Google Scholar] [CrossRef] [PubMed]
  14. Kwiecien, K.; Zegar, A.; Jung, J.; Brzoza, P.; Kwitniewski, M.; Godlewska, U.; Grygier, B.; Kwiecinska, P.; Morytko, A.; Cichy, J. Architecture of antimicrobial skin defense. Cytokine Growth Factor Rev. 2019, 49, 70–84. [Google Scholar] [CrossRef]
  15. Abraira, V.E.; Ginty, D.D. The sensory neurons of touch. Neuron 2013, 79, 618–639. [Google Scholar] [CrossRef]
  16. Peters, E.M.; Liezmann, C.; Klapp, B.F.; Kruse, J. The neuroimmune connection interferes with tissue regeneration and chronic inflammatory disease in the skin. Ann. N. Y. Acad. Sci. 2012, 1262, 118–126. [Google Scholar] [CrossRef]
  17. Kabata, H.; Artis, D. Neuro-immune crosstalk and allergic inflammation. J. Clin. Investig. 2019, 129, 1475–1482. [Google Scholar] [CrossRef]
  18. Agelopoulos, K.; Pereira, M.P.; Wiegmann, H.; Stander, S. Cutaneous neuroimmune crosstalk in pruritus. Trends Mol. Med. 2022, 28, 452–462. [Google Scholar] [CrossRef]
  19. Steinhoff, M.; Buddenkotte, J.; Lerner, E.A. Role of mast cells and basophils in pruritus. Immunol. Rev. 2018, 282, 248–264. [Google Scholar] [CrossRef] [PubMed]
  20. Corbière, A.; Loste, A.; Gaudenzio, N. MRGPRX2 sensing of cationic compounds-A bridge between nociception and skin diseases? Exp. Dermatol. 2021, 30, 193–200. [Google Scholar] [CrossRef]
  21. Alysandratos, K.D.; Asadi, S.; Angelidou, A.; Zhang, B.; Sismanopoulos, N.; Yang, H.; Critchfield, A.; Theoharides, T.C. Neurotensin and CRH interactions augment human mast cell activation. PLoS ONE 2012, 7, e48934. [Google Scholar] [CrossRef]
  22. Quinlan, K.L.; Song, I.S.; Bunnett, N.W.; Letran, E.; Steinhoff, M.; Harten, B.; Olerud, J.E.; Armstrong, C.A.; Wright Caughman, S.; Ansel, J.C. Neuropeptide regulation of human dermal microvascular endothelial cell ICAM-1 expression and function. Am. J. Physiol. 1998, 275, C1580–C1590. [Google Scholar] [CrossRef] [PubMed]
  23. Langan, S.M.; Irvine, A.D.; Weidinger, S. Atopic dermatitis. Lancet 2020, 396, 345–360. [Google Scholar] [CrossRef]
  24. Kuo, I.H.; Yoshida, T.; De Benedetto, A.; Beck, L.A. The cutaneous innate immune response in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2013, 131, 266–278. [Google Scholar] [CrossRef] [PubMed]
  25. Honda, T.; Kabashima, K. Reconciling innate and acquired immunity in atopic dermatitis. J. Allergy Clin. Immunol. 2020, 145, 1136–1137. [Google Scholar] [CrossRef] [PubMed]
  26. Steinhoff, M.; Ahmad, F.; Pandey, A.; Datsi, A.; AlHammadi, A.; Al-Khawaga, S.; Al-Malki, A.; Meng, J.; Alam, M.; Buddenkotte, J. Neuroimmune communication regulating pruritus in atopic dermatitis. J. Allergy Clin. Immunol. 2022, 149, 1875–1898. [Google Scholar] [CrossRef]
  27. Wang, F.; Yang, T.B.; Kim, B.S. The Return of the Mast Cell: New Roles in Neuroimmune Itch Biology. J. Investig. Dermatol. 2020, 140, 945–951. [Google Scholar] [CrossRef]
  28. Zhang, Y.; Zhang, H.; Jiang, B.; Tong, X.; Yan, S.; Lu, J. Current views on neuropeptides in atopic dermatitis. Exp. Dermatol. 2021, 30, 1588–1597. [Google Scholar] [CrossRef]
  29. Horigome, K.; Pryor, J.C.; Bullock, E.D.; Johnson, E.M., Jr. Mediator release from mast cells by nerve growth factor. Neurotrophin specificity and receptor mediation. J. Biol. Chem. 1993, 268, 14881–14887. [Google Scholar] [CrossRef]
  30. Nilsson, G.; Forsberg-Nilsson, K.; Xiang, Z.; Hallbook, F.; Nilsson, K.; Metcalfe, D.D. Human mast cells express functional TrkA and are a source of nerve growth factor. Eur. J. Immunol. 1997, 27, 2295–2301. [Google Scholar] [CrossRef]
  31. Meixiong, J.; Anderson, M.; Limjunyawong, N.; Sabbagh, M.F.; Hu, E.; Mack, M.R.; Oetjen, L.K.; Wang, F.; Kim, B.S.; Dong, X. Activation of Mast-Cell-Expressed Mas-Related G-Protein-Coupled Receptors Drives Non-histaminergic Itch. Immunity 2019, 50, 1163–1171.e1165. [Google Scholar] [CrossRef] [PubMed]
  32. McNeil, B.D.; Pundir, P.; Meeker, S.; Han, L.; Undem, B.J.; Kulka, M.; Dong, X. Identification of a mast-cell-specific receptor crucial for pseudo-allergic drug reactions. Nature 2015, 519, 237–241. [Google Scholar] [CrossRef]
  33. Kawakami, T.; Ando, T.; Kimura, M.; Wilson, B.S.; Kawakami, Y. Mast cells in atopic dermatitis. Curr. Opin. Immunol. 2009, 21, 666–678. [Google Scholar] [CrossRef] [PubMed]
  34. Yamaoka, J.; Di, Z.H.; Sun, W.; Kawana, S. Changes in cutaneous sensory nerve fibers induced by skin-scratching in mice. J. Dermatol. Sci. 2007, 46, 41–51. [Google Scholar] [CrossRef]
  35. Plum, T.; Wang, X.; Rettel, M.; Krijgsveld, J.; Feyerabend, T.B.; Rodewald, H.R. Human Mast Cell Proteome Reveals Unique Lineage, Putative Functions, and Structural Basis for Cell Ablation. Immunity 2020, 52, 404–416.e405. [Google Scholar] [CrossRef]
  36. Serhan, N.; Basso, L.; Sibilano, R.; Petitfils, C.; Meixiong, J.; Bonnart, C.; Reber, L.L.; Marichal, T.; Starkl, P.; Cenac, N.; et al. House dust mites activate nociceptor-mast cell clusters to drive type 2 skin inflammation. Nat. Immunol. 2019, 20, 1435–1443. [Google Scholar] [CrossRef]
  37. Geoghegan, J.A.; Irvine, A.D.; Foster, T.J. Staphylococcus aureus and Atopic Dermatitis: A Complex and Evolving Relationship. Trends Microbiol. 2018, 26, 484–497. [Google Scholar] [CrossRef] [PubMed]
  38. Blake, K.J.; Baral, P.; Voisin, T.; Lubkin, A.; Pinho-Ribeiro, F.A.; Adams, K.L.; Roberson, D.P.; Ma, Y.X.C.; Otto, M.; Woolf, C.J.; et al. Staphylococcus aureus produces pain through pore-forming toxins and neuronal TRPV1 that is silenced by QX-314. Nat. Commun. 2018, 9, 37. [Google Scholar] [CrossRef]
  39. Arifuzzaman, M.; Mobley, Y.R.; Choi, H.W.; Bist, P.; Salinas, C.A.; Brown, Z.D.; Chen, S.L.; Staats, H.F.; Abraham, S.N. MRGPR-mediated activation of local mast cells clears cutaneous bacterial infection and protects against reinfection. Sci. Adv. 2019, 5, eaav0216. [Google Scholar] [CrossRef]
  40. Wang, Z.; Babina, M. MRGPRX2 signals its importance in cutaneous mast cell biology: Does MRGPRX2 connect mast cells and atopic dermatitis? Exp. Dermatol. 2020, 29, 1104–1111. [Google Scholar] [CrossRef]
  41. Miyake, K.; Karasuyama, H. Emerging roles of basophils in allergic inflammation. Allergol. Int. 2017, 66, 382–391. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, F.; Trier, A.M.; Li, F.; Kim, S.; Chen, Z.; Chai, J.N.; Mack, M.R.; Morrison, S.A.; Hamilton, J.D.; Baek, J.; et al. A basophil-neuronal axis promotes itch. Cell 2021, 184, 422–440.e417. [Google Scholar] [CrossRef] [PubMed]
  43. Wedi, B.; Gehring, M.; Kapp, A. The pseudoallergen receptor MRGPRX2 on peripheral blood basophils and eosinophils: Expression and function. Allergy 2020, 75, 2229–2242. [Google Scholar] [CrossRef] [PubMed]
  44. Sonnenberg, G.F.; Artis, D. Innate lymphoid cell interactions with microbiota: Implications for intestinal health and disease. Immunity 2012, 37, 601–610. [Google Scholar] [CrossRef] [PubMed]
  45. Monticelli, L.A.; Sonnenberg, G.F.; Artis, D. Innate lymphoid cells: Critical regulators of allergic inflammation and tissue repair in the lung. Curr. Opin. Immunol. 2012, 24, 284–289. [Google Scholar] [CrossRef]
  46. Soumelis, V.; Reche, P.A.; Kanzler, H.; Yuan, W.; Edward, G.; Homey, B.; Gilliet, M.; Ho, S.; Antonenko, S.; Lauerma, A.; et al. Human epithelial cells trigger dendritic cell mediated allergic inflammation by producing TSLP. Nat. Immunol. 2002, 3, 673–680. [Google Scholar] [CrossRef]
  47. Deleuran, M.; Hvid, M.; Kemp, K.; Christensen, G.B.; Deleuran, B.; Vestergaard, C. IL-25 induces both inflammation and skin barrier dysfunction in atopic dermatitis. Chem. Immunol. Allergy 2012, 96, 45–49. [Google Scholar]
  48. Savinko, T.; Matikainen, S.; Saarialho-Kere, U.; Lehto, M.; Wang, G.; Lehtimaki, S.; Karisola, P.; Reunala, T.; Wolff, H.; Lauerma, A.; et al. IL-33 and ST2 in atopic dermatitis: Expression profiles and modulation by triggering factors. J. Investig. Dermatol. 2012, 132, 1392–1400. [Google Scholar] [CrossRef]
  49. Kim, B.S.; Wang, K.; Siracusa, M.C.; Saenz, S.A.; Brestoff, J.R.; Monticelli, L.A.; Noti, M.; Tait Wojno, E.D.; Fung, T.C.; Kubo, M.; et al. Basophils promote innate lymphoid cell responses in inflamed skin. J. Immunol. 2014, 193, 3717–3725. [Google Scholar] [CrossRef] [PubMed]
  50. Kim, B.S.; Siracusa, M.C.; Saenz, S.A.; Noti, M.; Monticelli, L.A.; Sonnenberg, G.F.; Hepworth, M.R.; Van Voorhees, A.S.; Comeau, M.R.; Artis, D. TSLP elicits IL-33-independent innate lymphoid cell responses to promote skin inflammation. Sci. Transl. Med. 2013, 5, 170ra16. [Google Scholar] [CrossRef]
  51. Salimi, M.; Barlow, J.L.; Saunders, S.P.; Xue, L.; Gutowska-Owsiak, D.; Wang, X.; Huang, L.C.; Johnson, D.; Scanlon, S.T.; McKenzie, A.N.; et al. A role for IL-25 and IL-33-driven type-2 innate lymphoid cells in atopic dermatitis. J. Exp. Med. 2013, 210, 2939–2950. [Google Scholar] [CrossRef] [PubMed]
  52. Leyva-Castillo, J.M.; Galand, C.; Mashiko, S.; Bissonnette, R.; McGurk, A.; Ziegler, S.F.; Dong, C.; McKenzie, A.N.J.; Sarfati, M.; Geha, R.S. ILC2 activation by keratinocyte-derived IL-25 drives IL-13 production at sites of allergic skin inflammation. J. Allergy Clin. Immunol. 2020, 145, 1606–1614.e1604. [Google Scholar] [CrossRef] [PubMed]
  53. Cardoso, V.; Chesne, J.; Ribeiro, H.; Garcia-Cassani, B.; Carvalho, T.; Bouchery, T.; Shah, K.; Barbosa-Morais, N.L.; Harris, N.; Veiga-Fernandes, H. Neuronal regulation of type 2 innate lymphoid cells via neuromedin U. Nature 2017, 549, 277–281. [Google Scholar] [CrossRef]
  54. Klose, C.S.N.; Mahlakoiv, T.; Moeller, J.B.; Rankin, L.C.; Flamar, A.L.; Kabata, H.; Monticelli, L.A.; Moriyama, S.; Putzel, G.G.; Rakhilin, N.; et al. The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammation. Nature 2017, 549, 282–286. [Google Scholar] [CrossRef] [PubMed]
  55. Nagashima, H.; Mahlakoiv, T.; Shih, H.Y.; Davis, F.P.; Meylan, F.; Huang, Y.; Harrison, O.J.; Yao, C.; Mikami, Y.; Urban, J.F., Jr.; et al. Neuropeptide CGRP Limits Group 2 Innate Lymphoid Cell Responses and Constrains Type 2 Inflammation. Immunity 2019, 51, 682–695.e686. [Google Scholar] [CrossRef]
  56. Nussbaum, J.C.; Van Dyken, S.J.; von Moltke, J.; Cheng, L.E.; Mohapatra, A.; Molofsky, A.B.; Thornton, E.E.; Krummel, M.F.; Chawla, A.; Liang, H.E.; et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 2013, 502, 245–248. [Google Scholar] [CrossRef]
  57. Inclan-Rico, J.M.; Ponessa, J.J.; Valero-Pacheco, N.; Hernandez, C.M.; Sy, C.B.; Lemenze, A.D.; Beaulieu, A.M.; Siracusa, M.C. Basophils prime group 2 innate lymphoid cells for neuropeptide-mediated inhibition. Nat. Immunol. 2020, 21, 1181–1193. [Google Scholar] [CrossRef]
  58. Novak, N. An update on the role of human dendritic cells in patients with atopic dermatitis. J. Allergy Clin. Immunol. 2012, 129, 879–886. [Google Scholar] [CrossRef]
  59. Tanei, R.; Hasegawa, Y. Immunohistopathological Analysis of Immunoglobulin E-Positive Epidermal Dendritic Cells with House Dust Mite Antigens in Naturally Occurring Skin Lesions of Adult and Elderly Patients with Atopic Dermatitis. Dermatopathology 2021, 8, 426–441. [Google Scholar] [CrossRef]
  60. Perner, C.; Flayer, C.H.; Zhu, X.; Aderhold, P.A.; Dewan, Z.N.A.; Voisin, T.; Camire, R.B.; Chow, O.A.; Chiu, I.M.; Sokol, C.L. Substance P Release by Sensory Neurons Triggers Dendritic Cell Migration and Initiates the Type-2 Immune Response to Allergens. Immunity 2020, 53, 1063–1077.e1067. [Google Scholar] [CrossRef]
  61. Buttari, B.; Profumo, E.; Domenici, G.; Tagliani, A.; Ippoliti, F.; Bonini, S.; Businaro, R.; Elenkov, I.; Rigano, R. Neuropeptide Y induces potent migration of human immature dendritic cells and promotes a Th2 polarization. FASEB J. 2014, 28, 3038–3049. [Google Scholar] [CrossRef] [PubMed]
  62. Chorny, A.; Gonzalez-Rey, E.; Delgado, M. Regulation of dendritic cell differentiation by vasoactive intestinal peptide: Therapeutic applications on autoimmunity and transplantation. Ann. N. Y. Acad. Sci. 2006, 1088, 187–194. [Google Scholar] [CrossRef] [PubMed]
  63. Meng, J.; Moriyama, M.; Feld, M.; Buddenkotte, J.; Buhl, T.; Szollosi, A.; Zhang, J.; Miller, P.; Ghetti, A.; Fischer, M.; et al. New mechanism underlying IL-31-induced atopic dermatitis. J. Allergy Clin. Immunol. 2018, 141, 1677–1689.e1678. [Google Scholar] [CrossRef] [PubMed]
  64. Meng, J.H.; Chen, W.W.; Wang, J.F. Interventions in the B-type natriuretic peptide signalling pathway as a means of controlling chronic itch. Br. J. Pharmacol. 2020, 177, 1025–1040. [Google Scholar] [CrossRef] [PubMed]
  65. Ding, W.; Stohl, L.L.; Wagner, J.A.; Granstein, R.D. Calcitonin gene-related peptide biases Langerhans cells toward Th2-type immunity. J. Immunol. 2008, 181, 6020–6026. [Google Scholar] [CrossRef]
  66. Griffiths, C.E.M.; Armstrong, A.W.; Gudjonsson, J.E.; Barker, J.N.W.N. Psoriasis. Lancet 2021, 397, 1301–1315. [Google Scholar] [CrossRef]
  67. Ayasse, M.T.; Buddenkotte, J.; Alam, M.; Steinhoff, M. Role of neuroimmune circuits and pruritus in psoriasis. Exp. Dermatol. 2020, 29, 414–426. [Google Scholar] [CrossRef]
  68. Sato, Y.; Ogawa, E.; Okuyama, R. Role of Innate Immune Cells in Psoriasis. Int. J. Mol. Sci. 2020, 21, 6604. [Google Scholar] [CrossRef]
  69. Nakamura, M.; Toyoda, M.; Morohashi, M. Pruritogenic mediators in psoriasis vulgaris: Comparative evaluation of itch-associated cutaneous factors. Br. J. Dermatol. 2003, 149, 718–730. [Google Scholar] [CrossRef]
  70. 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. [Google Scholar] [CrossRef]
  71. Volpe, E.; Pattarini, L.; Martinez-Cingolani, C.; Meller, S.; Donnadieu, M.H.; Bogiatzi, S.I.; Fernandez, M.I.; Touzot, M.; Bichet, J.C.; Reyal, F.; et al. Thymic stromal lymphopoietin links keratinocytes and dendritic cell-derived IL-23 in patients with psoriasis. J. Allergy Clin. Immunol. 2014, 134, 373–381. [Google Scholar] [CrossRef] [PubMed]
  72. Narbutt, J.; Olejniczak, I.; Sobolewska-Sztychny, D.; Sysa-Jedrzejowska, A.; Slowik-Kwiatkowska, I.; Hawro, T.; Lesiak, A. Narrow band ultraviolet B irradiations cause alteration in interleukin-31 serum level in psoriatic patients. Arch. Dermatol. Res. 2013, 305, 191–195. [Google Scholar] [CrossRef] [PubMed]
  73. Nattkemper, L.A.; Tey, H.L.; Valdes-Rodriguez, R.; Lee, H.; Mollanazar, N.K.; Albornoz, C.; Sanders, K.M.; Yosipovitch, G. The Genetics of Chronic Itch: Gene Expression in the Skin of Patients with Atopic Dermatitis and Psoriasis with Severe Itch. J. Investig. Dermatol. 2018, 138, 1311–1317. [Google Scholar] [CrossRef] [PubMed]
  74. Szepietowski, J.C.; Reich, A.; Wisnicka, B. Pruritus and psoriasis. Br. J. Dermatol. 2004, 151, 1284. [Google Scholar] [CrossRef]
  75. Chang, S.E.; Han, S.S.; Jung, H.J.; Choi, J.H. Neuropeptides and their receptors in psoriatic skin in relation to pruritus. Br. J. Dermatol. 2007, 156, 1272–1277. [Google Scholar] [CrossRef] [PubMed]
  76. Sandoval-Talamantes, A.K.; Gomez-Gonzalez, B.A.; Uriarte-Mayorga, D.F.; Martinez-Guzman, M.A.; Wheber-Hidalgo, K.A.; Alvarado-Navarro, A. Neurotransmitters, neuropeptides and their receptors interact with immune response in healthy and psoriatic skin. Neuropeptides 2020, 79, 102004. [Google Scholar] [CrossRef]
  77. Takahashi, T.; Yamasaki, K. Psoriasis and Antimicrobial Peptides. Int. J. Mol. Sci. 2020, 21, 6791. [Google Scholar] [CrossRef]
  78. Smith, C.H.; Barker, J.N.; Morris, R.W.; MacDonald, D.M.; Lee, T.H. Neuropeptides induce rapid expression of endothelial cell adhesion molecules and elicit granulocytic infiltration in human skin. J. Immunol. 1993, 151, 3274–3282. [Google Scholar] [CrossRef]
  79. Locker, F.; Vidali, S.; Holub, B.S.; Stockinger, J.; Brunner, S.M.; Ebner, S.; Koller, A.; Trost, A.; Reitsamer, H.A.; Schwarzenbacher, D.; et al. Lack of Galanin Receptor 3 Alleviates Psoriasis by Altering Vascularization, Immune Cell Infiltration, and Cytokine Expression. J. Investig. Dermatol. 2018, 138, 199–207. [Google Scholar] [CrossRef]
  80. Raychaudhuri, S.K.; Raychaudhuri, S.P. NGF and its receptor system: A new dimension in the pathogenesis of psoriasis and psoriatic arthritis. Ann. N. Y. Acad. Sci. 2009, 1173, 470–477. [Google Scholar] [CrossRef]
  81. Riol-Blanco, L.; Ordovas-Montanes, J.; Perro, M.; Naval, E.; Thiriot, A.; Alvarez, D.; Paust, S.; Wood, J.N.; von Andrian, U.H. Nociceptive sensory neurons drive interleukin-23-mediated psoriasiform skin inflammation. Nature 2014, 510, 157–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Spits, H.; Artis, D.; Colonna, M.; Diefenbach, A.; Di Santo, J.P.; Eberl, G.; Koyasu, S.; Locksley, R.M.; McKenzie, A.N.; Mebius, R.E.; et al. Innate lymphoid cells—A proposal for uniform nomenclature. Nat. Rev. Immunol. 2013, 13, 145–149. [Google Scholar] [CrossRef]
  83. Teunissen, M.B.M.; Munneke, J.M.; Bernink, J.H.; Spuls, P.I.; Res, P.C.M.; Te Velde, A.; Cheuk, S.; Brouwer, M.W.D.; Menting, S.P.; Eidsmo, L.; et al. Composition of innate lymphoid cell subsets in the human skin: Enrichment of NCR(+) ILC3 in lesional skin and blood of psoriasis patients. J. Investig. Dermatol. 2014, 134, 2351–2360. [Google Scholar] [CrossRef] [PubMed]
  84. Seillet, C.; Luong, K.; Tellier, J.; Jacquelot, N.; Shen, R.D.; Hickey, P.; Wimmer, V.C.; Whitehead, L.; Rogers, K.; Smyth, G.K.; et al. The neuropeptide VIP confers anticipatory mucosal immunity by regulating ILC3 activity. Nat. Immunol. 2020, 21, 168–177. [Google Scholar] [CrossRef] [PubMed]
  85. Talbot, J.; Hahn, P.; Kroehling, L.; Nguyen, H.; Li, D.; Littman, D.R. Feeding-dependent VIP neuron-ILC3 circuit regulates the intestinal barrier. Nature 2020, 579, 575–580. [Google Scholar] [CrossRef]
  86. Yu, H.B.; Yang, H.; Allaire, J.M.; Ma, C.; Graef, F.A.; Mortha, A.; Liang, Q.; Bosman, E.S.; Reid, G.S.; Waschek, J.A.; et al. Vasoactive intestinal peptide promotes host defense against enteric pathogens by modulating the recruitment of group 3 innate lymphoid cells. Proc. Natl. Acad. Sci. USA 2021, 118, e2106634118. [Google Scholar] [CrossRef]
  87. Nakajima, K.; Kataoka, S.; Sato, K.; Takaishi, M.; Yamamoto, M.; Nakajima, H.; Sano, S. Stat3 activation in epidermal keratinocytes induces Langerhans cell activation to form an essential circuit for psoriasis via IL-23 production. J. Dermatol. Sci. 2019, 93, 82–91. [Google Scholar] [CrossRef]
  88. Martini, E.; Wiken, M.; Cheuk, S.; Gallais Serezal, I.; Baharom, F.; Stahle, M.; Smed-Sorensen, A.; Eidsmo, L. Dynamic Changes in Resident and Infiltrating Epidermal Dendritic Cells in Active and Resolved Psoriasis. J. Investig. Dermatol. 2017, 137, 865–873. [Google Scholar] [CrossRef]
  89. Chiricozzi, A.; Romanelli, P.; Volpe, E.; Borsellino, G.; Romanelli, M. Scanning the Immunopathogenesis of Psoriasis. Int. J. Mol. Sci. 2018, 19, 179. [Google Scholar] [CrossRef]
  90. Ostrowski, S.M.; Belkadi, A.; Loyd, C.M.; Diaconu, D.; Ward, N.L. Cutaneous denervation of psoriasiform mouse skin improves acanthosis and inflammation in a sensory neuropeptide-dependent manner. J. Investig. Dermatol. 2011, 131, 1530–1538. [Google Scholar] [CrossRef]
  91. Auriemma, M.; Brzoska, T.; Klenner, L.; Kupas, V.; Goerge, T.; Voskort, M.; Zhao, Z.; Sparwasser, T.; Luger, T.A.; Loser, K. α-MSH-stimulated tolerogenic dendritic cells induce functional regulatory T cells and ameliorate ongoing skin inflammation. J. Investig. Dermatol. 2012, 132, 1814–1824. [Google Scholar] [CrossRef] [Green Version]
  92. Ding, W.; Manni, M.; Stohl, L.L.; Zhou, X.K.; Wagner, J.A.; Granstein, R.D. Pituitary adenylate cyclase-activating peptide and vasoactive intestinal polypeptide bias Langerhans cell Ag presentation toward Th17 cells. Eur. J. Immunol. 2012, 42, 901–911. [Google Scholar] [CrossRef] [PubMed]
  93. Borici-Mazi, R.; Kouridakis, S.; Kontou-Fili, K. Cutaneous responses to substance P and calcitonin gene-related peptide in chronic urticaria: The effect of cetirizine and dimethindene. Allergy 1999, 54, 46–56. [Google Scholar] [CrossRef] [PubMed]
  94. Smith, C.H.; Atkinson, B.; Morris, R.W.; Hayes, N.; Foreman, J.C.; Lee, T.H. Cutaneous responses to vasoactive intestinal polypeptide in chronic idiopathic urticaria. Lancet 1992, 339, 91–93. [Google Scholar] [CrossRef] [PubMed]
  95. Fujisawa, D.; Kashiwakura, J.; Kita, H.; Kikukawa, Y.; Fujitani, Y.; Sasaki-Sakamoto, T.; Kuroda, K.; Nunomura, S.; Hayama, K.; Terui, T.; et al. Expression of Mas-related gene X2 on mast cells is upregulated in the skin of patients with severe chronic urticaria. J. Allergy Clin. Immunol. 2014, 134, 622. [Google Scholar] [CrossRef]
  96. Zheng, W.; Wang, J.; Zhu, W.; Xu, C.; He, S. Upregulated expression of substance P in basophils of the patients with chronic spontaneous urticaria: Induction of histamine release and basophil accumulation by substance P. Cell Biol. Toxicol. 2016, 32, 217–228. [Google Scholar] [CrossRef]
  97. Hashimoto, T.; Kursewicz, C.D.; Fayne, R.A.; Nanda, S.; Shah, S.M.; Nattkemper, L.; Yokozeki, H.; Yosipovitch, G. Pathophysiologic mechanisms of itch in bullous pemphigoid. J. Am. Acad. Dermatol. 2020, 83, 53–62. [Google Scholar] [CrossRef]
  98. Lonndahl, L.; Holst, M.; Bradley, M.; Killasli, H.; Heilborn, J.; Hall, M.A.; Theodorsson, E.; Holmberg, J.; Nordlind, K. Substance P Antagonist Aprepitant Shows no Additive Effect Compared with Standardized Topical Treatment Alone in Patients with Atopic Dermatitis. Acta Derm. Venereol. 2018, 98, 324–328. [Google Scholar] [CrossRef]
  99. Ohanyan, T.; Schoepke, N.; Eirefelt, S.; Hoey, G.; Koopman, W.; Hawro, T.; Maurer, M.; Metz, M. Role of Substance P and Its Receptor Neurokinin 1 in Chronic Prurigo: A Randomized, Proof-of-Concept, Controlled Trial with Topical Aprepitant. Acta Derm. Venereol. 2018, 98, 26–31. [Google Scholar] [CrossRef]
  100. Pojawa-Golab, M.; Jaworecka, K.; Reich, A. NK-1 Receptor Antagonists and Pruritus: Review of Current Literature. Dermatol. Ther. 2019, 9, 391–405. [Google Scholar] [CrossRef]
  101. Yosipovitch, G.; Stander, S.; Kerby, M.B.; Larrick, J.W.; Perlman, A.J.; Schnipper, E.F.; Zhang, X.; Tang, J.Y.; Luger, T.; Steinhoff, M. Serlopitant for psoriatic pruritus: A phase 2 randomized, double-blind, placebo-controlled clinical trial. J. Am. Acad. Dermatol. 2020, 82, 1314–1320. [Google Scholar]
  102. Heitman, A.; Xiao, C.F.; Cho, Y.; Polymeropoulos, C.; Birznieks, G.; Polymeropoulos, M. Serlopitant reduced pruritus in patients with prurigo nodularis in a phase 2, randomized, placebo-controlled trial. J. Am. Acad. Dermatol. 2019, 80, 1395–1402. [Google Scholar]
  103. Welsh, S.E.; Xiao, C.; Kaden, A.R.; Brzezynski, J.L.; Mohrman, M.A.; Wang, J.; Smieszek, S.P.; Przychodzen, B.; Stander, S.; Polymeropoulos, C.; et al. Serlopitant for the treatment of chronic pruritus: Results of a randomized, multicenter, placebo-controlled phase 2 clinical trial. J. Am. Acad. Dermatol. 2018, 78, 882. [Google Scholar]
  104. Roblin, D.; Yosipovitch, G.; Boyce, B.; Robinson, J.; Sandy, J.; Mainero, V.; Wickramasinghe, R.; Anand, U.; Anand, P. Tradipitant improves worst itch and disease severity in patients with chronic pruritus related to atopic dermatitis. J. Am. Acad. Dermatol. 2018, 79, Ab300. [Google Scholar]
  105. Lee, Y.W.; Won, C.H.; Jung, K.; Nam, H.J.; Choi, G.; Park, Y.H.; Park, M.; Kim, B. Neurokinin-1 receptor antagonist tradipitant has mixed effects on itch in atopic dermatitis: Results from EPIONE, a randomized clinical trial. J. Eur. Acad. Dermatol. Venereol. 2021, 35, E338–E340. [Google Scholar]
  106. Roblin, D.; Yosipovitch, G.; Boyce, B.; Robinson, J.; Sandy, J.; Mainero, V.; Wickramasinghe, R.; Anand, U.; Anand, P. Topical TrkA Kinase Inhibitor CT327 is an Effective, Novel Therapy for the Treatment of Pruritus due to Psoriasis: Results from Experimental Studies, and Efficacy and Safety of CT327 in a Phase 2b Clinical Trial in Patients with Psoriasis. Acta Derm. Venereol. 2015, 95, 542–548. [Google Scholar] [CrossRef] [Green Version]
  107. Lee, Y.W.; Won, C.H.; Jung, K.; Nam, H.J.; Choi, G.; Park, Y.H.; Park, M.; Kim, B. Efficacy and safety of PAC-14028 cream—A novel, topical, nonsteroidal, selective TRPV1 antagonist in patients with mild-to-moderate atopic dermatitis: A phase IIb randomized trial. Br. J. Dermatol. 2019, 180, 1030–1038. [Google Scholar] [CrossRef] [PubMed]
  108. Lim, K.M.; Park, Y.H. Development of PAC-14028, a Novel Transient Receptor Potential Vanilloid Type 1 (TRPV1) Channel Antagonist as a New Drug for Refractory Skin Diseases. Arch. Pharmacal Res. 2012, 35, 393–396. [Google Scholar] [CrossRef] [PubMed]
  109. Wollenberg, A.; Blauvelt, A.; Guttman-Yassky, E.; Worm, M.; Lynde, C.; Lacour, J.P.; Spelman, L.; Katoh, N.; Saeki, H.; Poulin, Y.; et al. Tralokinumab for moderate-to-severe atopic dermatitis: Results from two 52-week, randomized, double-blind, multicentre, placebo-controlled phase III trials (ECZTRA 1 and ECZTRA 2). Br. J. Dermatol. 2021, 184, 437–449. [Google Scholar] [CrossRef] [PubMed]
  110. Guttman-Yassky, E.; Blauvelt, A.; Eichenfield, L.F.; Paller, A.S.; Armstrong, A.W.; Drew, J.; Gopalan, R.; Simpson, E.L. Efficacy and Safety of Lebrikizumab, a High-Affinity Interleukin 13 Inhibitor, in Adults With Moderate to Severe Atopic Dermatitis: A Phase 2b Randomized Clinical Trial. JAMA Dermatol. 2020, 156, 411–420. [Google Scholar] [CrossRef]
  111. Chen, Y.L.; Gutowska-Owsiak, D.; Hardman, C.S.; Westmoreland, M.; MacKenzie, T.; Cifuentes, L.; Waithe, D.; Lloyd-Lavery, A.; Marquette, A.; Londei, M.; et al. Proof-of-concept clinical trial of etokimab shows a key role for IL-33 in atopic dermatitis pathogenesis. Sci. Transl. Med. 2019, 11, eaax2945. [Google Scholar] [CrossRef]
  112. Simpson, E.L.; Parnes, J.R.; She, D.; Crouch, S.; Rees, W.; Mo, M.; van der Merwe, R. Tezepelumab, an anti-thymic stromal lymphopoietin monoclonal antibody, in the treatment of moderate to severe atopic dermatitis: A randomized phase 2a clinical trial. J. Am. Acad. Dermatol. 2019, 80, 1013–1021. [Google Scholar] [CrossRef]
  113. Napolitano, M.; Fabbrocini, G.; Scalvenzi, M.; Nistico, S.P.; Dastoli, S.; Patruno, C. Effectiveness of Dupilumab for the Treatment of Generalized Prurigo Nodularis Phenotype of Adult Atopic Dermatitis. Dermatitis 2020, 31, 81–84. [Google Scholar] [CrossRef] [PubMed]
  114. Calugareanu, A.; Jachiet, M.; Tauber, M.; Nosbaum, A.; Aubin, F.; Misery, L.; Droitcourt, C.; Barbarot, S.; Debarbieux, S.; Saussine, A.; et al. Effectiveness and safety of dupilumab for the treatment of prurigo nodularis in a French multicenter adult cohort of 16 patients. J. Eur. Acad. Dermatol. Venereol. 2020, 34, e74–e76. [Google Scholar] [CrossRef] [PubMed]
  115. Giura, M.T.; Viola, R.; Fierro, M.T.; Ribero, S.; Ortoncelli, M. Efficacy of dupilumab in prurigo nodularis in elderly patient. Dermatol. Ther. 2020, 33, e13201. [Google Scholar] [CrossRef] [PubMed]
  116. Tanis, R.; Ferenczi, K.; Payette, M. Dupilumab Treatment for Prurigo Nodularis and Pruritis. J. Drugs Dermatol. 2019, 18, 940–942. [Google Scholar] [PubMed]
  117. Zhai, L.L.; Savage, K.T.; Qiu, C.C.; Jin, A.; Valdes-Rodriguez, R.; Mollanazar, N.K. Chronic Pruritus Responding to Dupilumab—A Case Series. Medicines 2019, 6, 72. [Google Scholar] [CrossRef]
  118. Kabashima, K.; Furue, M.; Hanifin, J.M.; Pulka, G.; Wollenberg, A.; Galus, R.; Etoh, T.; Mihara, R.; Nakano, M.; Ruzicka, T. Nemolizumab in patients with moderate-to-severe atopic dermatitis: Randomized, phase II, long-term extension study. J. Allergy Clin. Immunol. 2018, 142, 1121–1130.e7. [Google Scholar] [CrossRef]
  119. Nemoto, O.; Furue, M.; Nakagawa, H.; Shiramoto, M.; Hanada, R.; Matsuki, S.; Imayama, S.; Kato, M.; Hasebe, I.; Taira, K.; et al. The first trial of CIM331, a humanized antihuman interleukin-31 receptor A antibody, in healthy volunteers and patients with atopic dermatitis to evaluate safety, tolerability and pharmacokinetics of a single dose in a randomized, double-blind, placebo-controlled study. Br. J. Dermatol. 2016, 174, 296–304. [Google Scholar]
  120. Ruzicka, T.; Hanifin, J.M.; Furue, M.; Pulka, G.; Mlynarczyk, I.; Wollenberg, A.; Galus, R.; Etoh, T.; Mihara, R.; Yoshida, H.; et al. Anti-Interleukin-31 Receptor A Antibody for Atopic Dermatitis. N. Engl. J. Med. 2017, 376, 826–835. [Google Scholar] [CrossRef]
  121. Stander, S.; Yosipovitch, G.; Legat, F.J.; Lacour, J.P.; Paul, C.; Narbutt, J.; Bieber, T.; Misery, L.; Wollenberg, A.; Reich, A.; et al. Trial of Nemolizumab in Moderate-to-Severe Prurigo Nodularis. N. Engl. J. Med. 2020, 382, 706–716. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Involvement of immune cells and sensory nervous system in the skin. Three components are shown: the stratified epidermis and dermis; representative innate and adaptive immune cells; the sensory nerves that are derived from dorsal root ganglia (DRG), which have afferent endings anatomically close to functional immune cells. Figure created with Biorender.
Figure 1. Involvement of immune cells and sensory nervous system in the skin. Three components are shown: the stratified epidermis and dermis; representative innate and adaptive immune cells; the sensory nerves that are derived from dorsal root ganglia (DRG), which have afferent endings anatomically close to functional immune cells. Figure created with Biorender.
Pharmaceuticals 16 00246 g001
Figure 2. The skin–immune–nerve circuit in AD and type 2 inflammation. Environmental stimuli cause keratinocytes to release epithelial cytokines that trigger a type 2 inflammatory axis in AD. Either non-self elements or effector molecules (e.g., histamine, serotonin, proteases, cytokines) could directly activate sensory neurons to provoke itch sensation. Activated sensory neurons, in turn, release neuropeptides or transmitters to the skin, thereby aggravating inflammation through the modulation of immune responses. Baso, basophil; CGRP, calcitonin-gene-related peptide; DC, dendritic cell; HDM, house dust mites; IL, interleukin; ILC2, group 2 innate lymphoid cell; KC, keratinocyte; LC, Langerhans cell; LTC4, leukotriene C4; MC, mast cell; MRGPR, Mas-related G protein-coupled receptor; NMB, neuromedin B; NMBR, neuromedin B receptor; Nmur, neuromedin U receptor; NPY, neuropeptide Y; SP, substance P; TSLP, thymic stromal lymphopoietin; VIP, vasoactive intestinal peptide. Figure created with Biorender.
Figure 2. The skin–immune–nerve circuit in AD and type 2 inflammation. Environmental stimuli cause keratinocytes to release epithelial cytokines that trigger a type 2 inflammatory axis in AD. Either non-self elements or effector molecules (e.g., histamine, serotonin, proteases, cytokines) could directly activate sensory neurons to provoke itch sensation. Activated sensory neurons, in turn, release neuropeptides or transmitters to the skin, thereby aggravating inflammation through the modulation of immune responses. Baso, basophil; CGRP, calcitonin-gene-related peptide; DC, dendritic cell; HDM, house dust mites; IL, interleukin; ILC2, group 2 innate lymphoid cell; KC, keratinocyte; LC, Langerhans cell; LTC4, leukotriene C4; MC, mast cell; MRGPR, Mas-related G protein-coupled receptor; NMB, neuromedin B; NMBR, neuromedin B receptor; Nmur, neuromedin U receptor; NPY, neuropeptide Y; SP, substance P; TSLP, thymic stromal lymphopoietin; VIP, vasoactive intestinal peptide. Figure created with Biorender.
Pharmaceuticals 16 00246 g002
Figure 3. Connection of neural mediators and innate immune responses in psoriasis and type 1/17 inflammation. Genetics, environmental factors, and psychological stress shape keratinocytes in the context of psoriasis. Type 1/17 cytokines lead to keratinocyte proliferation and sustained skin inflammation. Neuropeptides, mainly from nerves, are sufficient to activate the innate immune responses either in the skin or gut, leading to immune cell recruitment, cytokine release, and persistent inflammation. α-MSH, α-melanocyte-stimulating hormone; CGRP, calcitonin-gene-related peptide; DC, dendritic cell; GAL, Galanin; IL, interleukin; ILC3, group 3 innate lymphoid cell; KC, keratinocyte; LC, Langerhans cell; MC, mast cell; MC1R, melanocortin 1 receptor; MRGPR, Mas-related G protein-coupled receptor; Neut, neutrophil; NK1R, neurokinin 1 receptor; SP, substance P; TSLP, thymic stromal lymphopoietin; VIP, vasoactive intestinal peptide; VIPR, VIP receptor. Figure created with Biorender.
Figure 3. Connection of neural mediators and innate immune responses in psoriasis and type 1/17 inflammation. Genetics, environmental factors, and psychological stress shape keratinocytes in the context of psoriasis. Type 1/17 cytokines lead to keratinocyte proliferation and sustained skin inflammation. Neuropeptides, mainly from nerves, are sufficient to activate the innate immune responses either in the skin or gut, leading to immune cell recruitment, cytokine release, and persistent inflammation. α-MSH, α-melanocyte-stimulating hormone; CGRP, calcitonin-gene-related peptide; DC, dendritic cell; GAL, Galanin; IL, interleukin; ILC3, group 3 innate lymphoid cell; KC, keratinocyte; LC, Langerhans cell; MC, mast cell; MC1R, melanocortin 1 receptor; MRGPR, Mas-related G protein-coupled receptor; Neut, neutrophil; NK1R, neurokinin 1 receptor; SP, substance P; TSLP, thymic stromal lymphopoietin; VIP, vasoactive intestinal peptide; VIPR, VIP receptor. Figure created with Biorender.
Pharmaceuticals 16 00246 g003
Table 1. Main neuroimmune mechanisms in atopic dermatitis and psoriasis.
Table 1. Main neuroimmune mechanisms in atopic dermatitis and psoriasis.
Atopic Dermatitis or Type 2 InflammationPsoriasis or Type 1/17 Inflammation
Predominant inflammatory factorsIL-4, IL-13, IL-31, IL-25, IL-33, TSLPIFN-γ, IL-17A, IL-22, IL-23, TNF-α
Neuropeptide mechanism
Substance PPromotes mast cell activation and degranulation; promotes migration of CD301b+ DCsPromotes mast cell degranulation
Calcitonin-gene-related peptidePromotes mast cell degranulation; limits ILC2 proliferation and IL-13 production; enhances LC functionPromotes mast cell degranulation; promotes infiltration of DCs and T cells
Vasoactive intestinal peptidePromotes mast cell degranulation; promotes IL-5 release from ILC2s; induces Th2 polarization Promotes mast cell degranulation; promotes ILC3 recruitment; influences IL-22 production from ILC3s; enhances LC function
Brain natriuretic
peptide
Stimulates inflammatory factors from DCsUnknown
Neuropeptide YPromotes mast cell degranulation; induces migration of human immature DCsUnknown
Neuromedin BLimits type 2 inflammationUnknown
Neuromedin UInduces ILC2 proliferation and activation Unknown
Nerve growth factorPromotes mast cell degranulationEnhances basophil function
GalaninUnknownInduces neo-vascularization, neutrophil infiltration, and cytokine release
α-Melanocyte-stimulating hormoneUnknownInduces tolerogenic DCs; leads to Treg proliferation; inhibits Th17 activities
Abbreviations: DC, dendritic cell; IFN-γ, interferon-γ; IL, interleukin; ILC, innate lymphoid cell; LC, Langerhans cell; TNF-α, tumor necrosis factor α; Treg, regulatory T cells; TSLP, thymic stromal lymphopoietin.
Table 2. Emerging therapeutic agents associated with neuroimmune interactions.
Table 2. Emerging therapeutic agents associated with neuroimmune interactions.
TargetTherapeutic AgentsModeIndicationsReferences/NCT Number
NK-1RAprepitantAntagonistAD; chronic prurigo[98,99]
SerlopitantAntagonistAD; psoriasis; PN; chronic refractory pruritus[100,101,102,103]; NCT02975206; NCT03343639; NCT03546816; NCT01951274
TradipitantAntagonistAD[104,105]; NCT02004041; NCT02651714
TrkAPegcantratinibInhibitorPsoriasis[106]; NCT03448081
KOR/MORNalbuphineAgonist of KOR/antagonist of MORChronic prurigoNCT02174419; NCT02174432
TRPV1AsivatrepAntagonistAD[107,108]
IL-13TralokinumabmAbAD[109]; NCT03363854
LebrikizumabmAbAD[110]; NCT04146363; NCT04178967; NCT04392154; NCT04250350; NCT04250337
IL-33EtokimabmAbAD[111]; NCT03533751
TSLPTezepelumabmAbAD[112]; NCT02525094; NCT03809663
IL-4RαDupilumabmAbAD; PN; chronic
pruritus
[113,114,115,116,117]
IL-31RANemolizumabmAbAD; PN[118,119,120,121]
Abbreviations: AD, atopic dermatitis; KOR, kappa opioid receptor; mAb, monoclonal antibody; MOR, Mu opioid receptor; NCT, National Clinical Trial; NK-1R, neurokinin 1 receptor; PN, prurigo nodularis; TRP, transient receptor potential; TSLP, thymic stromal lymphopoietin.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Huang, X.; Li, F.; Wang, F. Neural Regulation of Innate Immunity in Inflammatory Skin Diseases. Pharmaceuticals 2023, 16, 246. https://doi.org/10.3390/ph16020246

AMA Style

Huang X, Li F, Wang F. Neural Regulation of Innate Immunity in Inflammatory Skin Diseases. Pharmaceuticals. 2023; 16(2):246. https://doi.org/10.3390/ph16020246

Chicago/Turabian Style

Huang, Xiaobao, Fengxian Li, and Fang Wang. 2023. "Neural Regulation of Innate Immunity in Inflammatory Skin Diseases" Pharmaceuticals 16, no. 2: 246. https://doi.org/10.3390/ph16020246

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

Article Metrics

Back to TopTop