Open Access

Immunological signature of chronic spontaneous urticaria (Review)

  • Authors:
    • Carmen-Teodora Dobrican
    • Ioana Adriana Muntean
    • Irena Pintea
    • Carina Petricău
    • Diana-Mihaela Deleanu
    • Gabriela Adriana Filip
  • View Affiliations

  • Published online on: April 8, 2022     https://doi.org/10.3892/etm.2022.11309
  • Article Number: 381
  • Copyright: © Dobrican et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Chronic urticaria (CU) is a condition characterized by intensely pruritic, edematous, erythematous papules lasting for more than 6 weeks. Over half of the cases have concomitant swelling of deeper tissues, known as angioedema. The socio‑economic burden of the disease is significant. Unfortunately, patients with severe CU, refractory to conventional treatment, have limited and expensive therapeutic options. The pathogenesis of CU is not yet completely understood. Therefore, elucidating the pathophysiological mechanisms involved would potentially identify new therapeutic targets. It has been accepted in recent years that mast cells and their activation, followed by excessive degranulation represent the key pathophysiological events in chronic spontaneous urticaria (CSU). The triggering events and the complexity of the effector mechanisms, however, remain intensely debated topics with conflicting studies. One pathogenetic mechanism incriminated in chronic spontaneous urticaria is the response mediated by the high‑affinity receptor for IgE (FcεRI) expressed on mast cells. Increasing recognition of chronic spontaneous urticaria as an autoimmune disease linked to the cytokine‑chemokine network imbalance resulting from alteration of innate immune response is another pathogenetic explanation. It is likely that these different pathological mechanisms are more interconnected, both acting synergistically, rather than separately, to produce the clinical expression of CU. The discovery and understanding of pathogenic mechanisms represent the premise for the development of safe and effective immunomodulators and targeted biological treatment for severe, refractory CU.

1. Introduction

According to the 2018 European Academy of Allergology and Clinical Immunology (EAACI) guideline, urticaria is a disease characterized by the development of an urticarial rash (also called hives/wheals), angioedema, or wheals plus angioedema (1). By definition, chronic spontaneous urticaria (CSU) is a diagnosis of exclusion after ruling out other conditions defined by the appearance of the wheals, angioedema or both. Some of these conditions are: Autoinflammatory syndromes, anaphylaxis, vasculitis (bradykinin-mediated) or bradykinin-mediated angioedema, including hereditary angioedema (defined by angioedema unaccompanied by urticaria) (1). Urticaria is classified based on the duration of the symptoms into acute (≤6 weeks) or chronic (>6 weeks). Furthermore, in relation to precipitating factors, urticaria is subclassified into spontaneous (does not involve any identifiable specific trigger) or inducible (has a known trigger) (1). The present review examined chronic urticaria (CU), specifically CSU.

The effect of CSU on patients, their family and friends, their jobs, the health system and society is substantial. The use of reported patient measurements, such as urticaria activity score (UAS), Angioedema Activity Score (AAS), Quality of Life Questionnaire (CU-Q2oL), AE-QoL Questionnaire and Urticaria Control Test (UCT) in clinical trials has helped to better define the multi-faced effect of CSU (2-6). Numerous studies based on these and data obtained from these assessments have helped highlight the impact and reveal that CSU influences the lives of patients both functionally, objectively and subjectively (7-9). In 1997 O'Donnell et al revealed that the health scores of patients with CSU were comparable to those reported in patients with coronary artery disease (10). The costs for treating this disease are also extremely high, both for patients and society (11-13).

In recent years, a research team coordinated by Maurer revealed that the essential pathophysiological element in CSU is the mast cell (14). From what is known at present, a brief overview could be summarized by the release of histamine and other mediators, such as platelet activating factor (PAF) and cytokines by activated mast cells leading to sensory nerve stimulation, vasodilation and plasma extravasation, as well as the recruitment of other immune cells to the urticaria site. Moreover, the signals for activating mast cells leading to hives are incomplete and poorly understood and appear complicated, intricate and heterogeneous (14).

From a histological point of view, edema appears in the upper and middle dermis, due to dilation and increase of postcapillary venules and lymphatic vessel permeability in the upper dermis, leading to extravasation of fluids in the tissues. Regarding angioedema, the same changes occur, only that these changes are located initially in the deep dermis ultimately extending to the subcutaneous tissue. In the lesional skin affected by the hives, there is almost always a perivascular inflammatory infiltrate, composed of neutrophils with or without eosinophils, basophils, macrophages and T cells and an increase in the number of endothelial cells, adhesion molecules, neuropeptides and growth factors. In urticaria, there is no necrosis of the vascular wall, this being a pathognomonic sign of vasculitis (15-19). Even in the healthy skin of patients with CU, certain changes occur, such as eosinophilic infiltration, increased expression of adhesion molecules and of some cytokines (20,21). All the aforementioned highlight how complex the pathogenic mechanism of urticaria is and how much it exceeds what is known to date, proving that it is markedly more than the activation, degranulation of mast cells and basophils with the release of vasoactive mediators (22-24). Due to the fact that numerous of these molecular findings are also observed in other autoinflammatory diseases, they cannot be used as biomarkers or be identified as typical changes in CSU. Additional research could lead to an improved understanding and identification of more specific pathogenic pathways.

2. Methods

For this review the Google Scholar browser was used, with the following search terms: ‘Mast cells’ and/or ‘chronic spontaneous urticaria pathogenesis’. The selection period of articles was 2010-2018 and any type of article was selected, being sorted according to relevance. The results produced over 3,000 articles, and among them those referring mainly to the biological treatment known and already used in clinical practice were eliminated, while those focusing on pathogenesis were retained. Following an overview of each of the titles of these articles, the 200 most relevant to our topic were selected. Older publications presented in the selected articles were also reviewed as well and introduced in our study if deemed relevant. Finally, 101 articles helped us in compiling the data presented in this review.

3. Evidence for autoimmune etiology

The first indication that urticaria could have an autoimmune basis, with an intrinsic immune imbalance, excluding extrinsic factors as the cause, comes from the so-called ‘autologous serum skin test’, in which by intradermal injection of the serum of the CU patient, an erythematous papule is produced at the injection site (25-27).

Previous studies have revealed that IgG antibodies against the IgE or high affinity receptor (FcεRI) of the patient lead to the activation, degranulation of mast cells and the appearance of urticarial lesions, but this does not fully explain the complexity of pathogenesis (14,28-30). Strengthening the theory, it is known that urticarial lesions are induced by vasoactive mediators, the main representative remaining the histamine released by mast cells. Hide et al identified IgG autoantibodies against the α subunit of high-affinity IgE receptors in the plasma of patients with CSU (30). These autoantibodies cross-link with IgE receptors, being responsible in some cases for mast cell activation and induction of specific lesions in CSU and other mast cell-mediated autoimmune diseases (30-33). The same theory is supported by studies revealing that patients with CSU have high serum levels of IgE autoantibodies, such as IgE-anti-TPO and IgE-anti-dDNA (31,34,35). The concept of ‘overlapping autoimmune diseases’ is also presented, suggesting that the concomitant presence of autoimmune diseases is occasionally observed along with CSU (36).

In addition, more than 200 IgE-type autoantigens were found in patients with CSU that were not in the blood of controls. IgE can react with autoantigens identified in the plasma of patients with CSU and other autoimmune diseases (including systemic lupus erythematosus, rheumatoid arthritis and multiple sclerosis). This phenomenon stimulates the activation of mast cells in the CSU, which degranulates rapidly under the action of intrinsic factors (autoreactive IgG against IgE and FcεRI, autoreactive IgE against its own antigens) (37). Previous evidence has emphasized the importance of IgE-anti-IL-24, which was revealed in all patients with CSU, suggesting and supporting that IL-24 may be a dermal autoantigen specific to this condition (14,38,39). Therefore, a possible autoimmune mechanism of CSU is the development of autoantibodies for the high affinity IgE receptor, FcεRI, which is found on the surface of mast cells and to a lesser extent on basophils, and is responsible for the activation and degranulation of these cells, followed by the release of vasoactive mediators. Activation of the high affinity IgE receptor, FcεRI, apparently represents an important step in the occurrence of urticarial lesions (35,36,40,41). This receptor is composed of 4 subunits: α, β and two γ, the last two being susceptible to the pathological activity of mast cells when they have an inadequate regulation (42).

A population study of more than 12,000 subjects in Israel revealed an incidence of autoimmune diseases (Sjögren's syndrome, rheumatoid arthritis, celiac disease, type 1 diabetes and systemic lupus erythematosus) in women with CSU, significantly higher than in women without CSU (43). The prevalence of antinuclear antibodies, anti-thyroperoxidase (anti-TPO), anti-thyroglobulin, rheumatoid factor, anti-transglutaminase IgA and anti-cardiolipin have also been revealed to be increased among individuals with CSU and associated autoimmune diseases. Therefore, a common pathophysiological mechanism between CSU and other autoimmune diseases can be considered, due to the increased prevalence of autoantibodies and the existence of a chronic inflammatory process expressed by a persistently increased mean platelet volume (MPV) (43).

Another important aspect to mention is the association between CSU and autoimmune thyroid diseases. An increased prevalence of Hashimoto's thyroiditis and Basedow-Graves' disease has been revealed in patients with CSU, with increased values of anti-thyroid IgG antibodies (31). Numerous patients diagnosed with urticaria have increased values of anti-thyroglobulin (anti-TG) or antimicrosomal antibodies/anti-TPO (44). Significantly elevated anti-TPO Ac IgE values have also been revealed in patients with CSU, thus supporting the theory of autoallergic mast cell activation that contributes to the pathophysiology of CSU. IgE has been also credited with an important role in autoimmunity, thus complementing its well-known role in defending against helminth infections and exogenous allergens. Concurrently, it has been revealed that these anti-TPO antibodies have the ability to induce degranulation of basophils in vitro in the presence of TPO antigens and thus play an important role in the pathogenesis of CSU, which is another argument in favor of the autoimmune etiology of this disease (45-47). Autoallergy thus refers to a type I hypersensitivity reaction, mediated by IgE against its own antigens, which can stimulate the degranulation of mast cells, type I autoimmunity (14,25). It is important to understand the formation of IgE autoantibodies and their contribution to the pathogenesis of CSU compared with IgG autoantibodies. A summary of the autoantibodies identified in CSU are presented in Table I.

Table I

Autoantibodies identified in chronic spontaneous urticaria.

Table I

Autoantibodies identified in chronic spontaneous urticaria.

AutoantibodiesRefs.
IgG antibodies to αFcεRI receptor(36,37,40-42)
IgG antibodies to IgE(30)
IgG antibodies anti-TG(31,44)
IgE and IgG antibodies anti-TPO(21,22,31,44)
ANA, RF(30,35,42,43)
ACA-IgG, IgM or IgA(43)
Anti-transglutaminase IgA antibodies(43)

[i] Anti-TG, anti-thyroglobulin; anti-TPO, anti-thyroperoxidase; ANA, antinuclear antibodies; RF, rheumatoid factor; ACA, anti-cardiolipin antibodies.

In conclusion, it should be generally accepted that autoimmunity (type I, described by IgE antibodies against autoantigens or type 2b, characterized by IgG autoantibodies to IgE or their FcεRI receptor) is considered to be the most common cause of urticaria, with the mast cells rich in high affinity receptors, as the main cellular elements (14,39).

4. Mast cells

Mast cells are round, mononuclear cells up to 25 microns in diameter, with a round or oval-shaped unicellular nucleus, eccentrically positioned and containing numerous cytoplasmic granules. They can be identified by special stains (Giemsa and toluidine blue), which poorly identify granulated immature or degranulated mast cells or by more sensitive techniques, such as tryptase immunohistochemistry. Moreover, mast cells can be marked with monoclonal antibodies that recognize membrane receptor targets of these cells (kit receptor, the high-affinity receptor for IgE) and proteases from intracytoplasmic granules (tryptase, chymase and carboxypeptidase) (48-51).

Mast cells are defined according to the content of their intracytoplasmic granules. The majority of tryptase-rich mast cells are predominantly located in mucous membranes (T-mast cells), while those containing tryptase, carboxypeptidase and chymase predominate in the connective tissue of the skin (TC-mast cells) (52-55). With regard to the mast cells involved in the pathogenesis of CSU, particularly TC-mast cells are referred to, which contain both tryptase and chymase (connective tissue mast cells).

Mast cells in patients with urticaria, similar to those in healthy individuals, are located in the superficial and deep dermis, preferentially in the perivascular and periaxonal spaces (56,57). The largest number of mast cells is identified in the superficial dermis of skin lesions, and few in the deep reticular dermis (58,59). It has been revealed in several studies that the distribution of mast cells in the dermis with urticarial lesions is different from that in healthy dermis, but it has remained ambiguous and unknown whether the number of these cells changes, increasing in urticaria (14,56,57). A previous study regarding CSU revealed that there was an increase in the proportion of dermal mast cells from 11 to 14% (60). This study was not unique, others on the same topic also identified an increased number of mast cells in both injured and intact skin in patients with urticaria compared with the skin of healthy subjects (20,61). Studies supporting this increase indicate that the changes revealed, appear only after at least 10 weeks of illness, thus only in CU (20,60,61). It is surmised that there remains uncertainty as to whether or not the number of mast cells in the dermis of patients with CU changes, because contrary to the studies mentioned in favor of this theory, there are numerous others that demonstrate the opposite, that in fact there are no increases in the number of these cells in the dermis of the CU patients compared with the dermis of the control subjects (56,57,62).

Scanning and transmission electron microscopy has been used since the 1970s to demonstrate mast cell degranulation (63). It was later used to identify mast cell degranulation in the affected skin of individuals with hives as well. Numerous histological studies have revealed mononuclear cells and perivascular eosinophilic infiltration in areas with urticaria wheals (64-67). Thus, mast cell degranulation, an immediate process followed by the observation of persistent eosinophilic infiltrate, suggests that there is also a delayed allergic inflammation in the pathogenesis of CSU, reinforcing the theory that the mechanisms are combined and heterogeneous (20). Furthermore, several cytokines have been identified in CSU, supporting the existence of a chronic inflammatory response, most of them having mast cell origin.

5. Cytokines in CSU

Mast cells release proinflammatory factors including cytokines. IL-31 is involved in the onset of pruritus, the release of IL-4 and IL-13, chemotactism for basophils, proliferation of the epithelial basal layer and inhibition of filaggrin synthesis. IL-4 and IL-13 direct T-lymphocyte differentiation to T helper 2 and B lymphocytes toward IgE synthesis (68-74). The study of these cytokines has led to new therapeutic approaches, such as the use of nemolizumab (humanized monoclonal antibody against IL-31 receptor A). It has been observed that the itching cannot be completely prevented by antihistamines, which is why the role of other substances implicated in the mechanism of its occurrence have been considered. CSU patients have exhibited higher plasma concentration of IL-31, contributing significantly to the onset of itching. Serum levels of IL-31, a member of the IL-6 cytokine family, are higher in CSU than in control patients, but the values are lower than in patients with atopic dermatitis. Although secreted primarily by Th2 lymphocytes, IL-31 is also released by mast cells and basophils. They produce and release IL-31 through an IgE-dependent mechanism. IgE binding to FcεRI activates mast cells and basophils that release IL-31. Once released, IL-31 has an autocrine effect on basophils by binding to IL-31 receptors (IL-31RA) and oncostatin M (OSM) receptors (which have a role in increasing the binding affinity of IL-31 to IL-31RA) leading to release of Th2-type cytokines: IL-4 and IL-13 (73-76). Nemolizumab is an antibody targeted against the IL-31RA receptor, preventing the binding of IL-31 and consequently the development of the cascade of reactions implicated. The use of this monoclonal antibody in clinical trials has reduced pruritus, demonstrating the major role of IL-31 in this process (77).

IL-33 induces a strong Th2 response and acts as an alarm for the immune system when the tissue is injured (78). A previous study revealed increases of IL-33 in the plasma of patients with CSU (75). IL-33 is expressed locally in inflamed skin, both in keratinocytes and in mast cells, but not in basophils, a situation highlighted in animal models. IL-33 has two actions that may be essential in urticaria: First, it activates itch-mediating sensory neurons, and second, it can modulate aspects of mast cell function, including adhesion, maturation, degranulation, and the production of a variety of Th2 cytokines (17,20,79-81). With this last role, IL-33 contributes to the induction of Th2-type inflammation observed in CU.

IL-25 is a member of the IL-17 cytokine family, the so-called autoimmune cytokine family. It is a potent inflammatory promoter of the Th2 type, found in epithelial cells, mast cells and eosinophils involved in hives. The fact that mast cells could produce IL-25 after IgE-dependent activation suggests a possible pathway of innate immunity that can stimulate Th2 response in CU (82-84). The question of whether it can be considered as an important autoantigen involved in the pathological mechanism deserves to be addressed in future scientific research.

Thymus stromal lymphopoietin (TSLP) is another member of the IL-17 cytokine family, which has also been revealed to be increased in urticarial lesions (75). While the cellular origin of TSLP has not been described in this pathology, an increased number of TSLP-positive mast cells have been observed in the bronchial tissue of patients with asthma (85-87). The primary role of TSLP in the skin is related to the development of Th2 cells by recruitment and activation of dendritic cells. TSLP does not induce degranulation of the mast cells per se but stimulates them in the presence of IL-1 and TNF and induces the production and release of other proinflammatory cytokines/chemokines including IL-5, IL-13, IL-6, GM-CSF, CXCL8 and CCL1(88).

6. Treatment in CSU

At present, the therapeutic protocol provided by The EAACI/GA²LEN/EDF/WAO guideline of urticaria can be summarized as revealed in Table II (1).

Table II

Treatment in chronic spontaneous urticariaa.

Table II

Treatment in chronic spontaneous urticariaa.

Step 1: Second-generation H1-antihistamines
     If symptom control is not achieved after 2-4 weeks of use or earlier if symptoms are very severe, proceed to step 2
Step 2: Increase second-generation H1-antihistamine dose (up to 4x)
     If symptom control is not achieved after 2-4 weeks of use or earlier if symptoms are very severe, proceed to step 3
Step 3: Omalizumab (add-on to second-generation H1-antihistamines)
     If symptom control is not achieved after 6 months of use or earlier if symptoms are very severe, proceed to step 4
Step 4: Cyclosporin A (add-on to second-generation H1-antihistamines)

[i] aAdapted from ref 1.

As it can be observed, although numerous therapies have been tried for refractory CSU treated with four times the dose of second-generation antihistamines, H1, the only ones approved by scientific consensus are omalizumab and cyclosporine A.

Omalizumab is a humanized G1K immunoglobulin monoclonal antibody produced by recombinant DNA technology which binds selectively to human IgE. It was originally approved in the US in 2003 for the treatment of moderate to severe persistent allergic bronchial asthma, and then in the European Union (EU) in 2005. In 2014, omalizumab was also approved in patients with CU, both in the US and in the EU, being the first drug indicated in patients who remain symptomatic despite maximal anti-H1 antihistamine treatment. Accumulated evidence has revealed that IgE, by binding to FcεRI on mast cells, can upregulate the receptors by promoting their proliferation and survival, thereby maintaining an increased number available (89,90). IgE coupling with FcεRI may also decrease the release threshold of mast cells and increase their sensitivity to different stimuli, either by FcεRI or by other receptors, resulting in an overactive degranulation process. Furthermore, the IgE-FcεRI binding potentiates the ability of mast cells to store and synthesize inflammatory mediators and cytokines de novo. Omalizumab administration, by depleting IgEs, attenuates their multiple effects, such as managing mast cell activities, thereby reducing the ability of these cells to manifest inflammatory mechanisms in patients with CSU (91). There has even been a decrease in serum IL-31 levels observed after treatment with omalizumab (91), which leads us to consider this interleukin as a potential biomarker in the diagnosis and monitoring of urticaria treatment, providing hope for the future.

The effectiveness of cyclosporine A in combination with a second-generation H1 antihistamine has been demonstrated in clinical trials on CSU, but this drug cannot be recommended as a standard treatment due to its numerous side effects (92-97). Cyclosporine A is only recommended for patients with severe refractory disease at any dose of antihistamine and omalizumab combination. Compared with long-term use of glucocorticosteroids, cyclosporine A has a more favorable risk/benefit ratio.

7. Conclusions

As it could be perceived in the present review, the immunological signature of urticaria is complicated, with numerous potential players and possible research points. Certainly, many aspects have remained unidentified and require future studies, but exploring each known part of the immunopathological mechanism may pave the way for possible therapeutic targets.

Every question can lead to an answer, thus it is definitely worth asking! Some of our queries after this introduction to the mysteries of CSU would be: i) How important is the role of the mast cell in the pathogenesis of this disease? Does this cell change its location, number and predisposition to degranulation depending on the clinical severity of urticaria? ii) What are the optimal biomarkers for both the diagnosis and monitoring of different treatments in CSU? Can IL-31 be considered a serological response biomarker to omalizumab? iii) The study of the IL-31 target in CSU may represent a new therapeutic approach, which merits further research. Can nemolizumab be used in the treatment of CSU? iv) Evaluation of various cytokine involvement (IL-31, IL-33, IL-25 and TSLP) in the pathophysiological mechanism of urticaria. Does their serum value change in the blood of patients in accordance with the evolution of the disease? Which of these cytokines deserves to be the most studied in this entity? Can IL-25 be considered as an autoantigen? v) Evaluation of IgE, FcεRI, and IgG-anti-IgE and IgG-anti-FcεRI antibodies in CU to provide more evidence of autoimmune etiology. vi) Omalizumab resistance mechanism; when does it become inefficient? vii) Identifying a correlation between the genetic polymorphism of an interleukin with mast cell origin and the predisposition for the development of CSU is also an area which requires study and exploration.

Collectively, this review shed light on new therapeutic targets and identified potential new defense mechanisms against CSU, which is a great burden to sufferers.

Acknowledgements

This review was published under the frame of the European Social Found, Human Capital Operational Programme 2014-2020, project no. POCU/380/6/13/125171.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

CTD and GAF conceived the present review. CTD developed the theory and performed the data collection. IAM and DMD supervised the findings of this work. CTD, GAF, IP and CP contributed to writing the manuscript and revising it critically for important intellectual content. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Zuberbier T, Aberer W, Asero R, Abdul Latiff AH, Baker D, Ballmer-Weber B, Bernstein JA, Bindslev-Jensen C, Brzoza Z, Buense Bedrikow R, et al: The EAACI/GA²LEN/EDF/WAO guideline for the definition, classification, diagnosis and management of urticaria. Allergy. 73:1393–1414. 2018.PubMed/NCBI View Article : Google Scholar

2 

Mathias SD, Crosby RD, Zazzali JL, Maurer M and Saini SS: Evaluating the minimally important difference of the urticaria activity score and other measures of disease activity in patients with chronic idiopathic urticaria. Ann Allergy Asthma Immunol. 108:20–24. 2012.PubMed/NCBI View Article : Google Scholar

3 

Weller K, Groffik A, Magerl M, Tohme N, Martus P, Krause K, Metz M, Staubach P and Maurer M: Development and construct validation of the angioedema quality of life questionnaire. Allergy. 67:1289–1298. 2012.PubMed/NCBI View Article : Google Scholar

4 

Hawro T, Ohanyan T, Schoepke N, Metz M, Peveling-Oberhag A, Staubach P, Maurer M and Weller K: The urticaria activity score-validity, reliability, and responsiveness. J Allergy Clin Immunol Pract. 6:1185–1190.e1. 2018.PubMed/NCBI View Article : Google Scholar

5 

Hawro T, Ohanyan T, Schoepke N, Metz M, Peveling-Oberhag A, Staubach P, Maurer M and Weller K: Comparison and interpretability of the available urticaria activity scores. Allergy. 73:251–255. 2018.PubMed/NCBI View Article : Google Scholar

6 

Baiardini I, Braido F, Bindslev-Jensen C, Bousquet PJ, Brzoza Z, Canonica GW, Compalati E, Fiocchi A, Fokkens W, Gerth van Wijk R, et al: Recommendations for assessing patient-reported outcomes and health-related quality of life in patients with urticaria: A GA(2) LEN taskforce position paper. Allergy. 66:840–844. 2011.PubMed/NCBI View Article : Google Scholar

7 

Maurer M, Staubach P, Raap U, Richter-Huhn G, Bauer A, Ruëff F, Jakob T, Yazdi AS, Mahler V, Wagner N, et al: H1-antihistamine-refractory chronic spontaneous urticaria: It's worse than we thought-first results of the multicenter real-life AWARE study. Clin Exp Allergy. 47:684–692. 2017.PubMed/NCBI View Article : Google Scholar

8 

Maurer M, Staubach P, Raap U, Richter-Huhn G, Baier-Ebert M and Chapman-Rothe N: ATTENTUS, a German online survey of patients with chronic urticaria highlighting the burden of disease, unmet needs and real-life clinical practice. Br J Dermatol. 174:892–894. 2016.PubMed/NCBI View Article : Google Scholar

9 

Maurer M, Abuzakouk M, Bérard F, Canonica W, Oude Elberink H, Giménez-Arnau A, Grattan C, Hollis K, Knulst A, Lacour JP, et al: The burden of chronic spontaneous urticaria is substantial: Real-world evidence from ASSURE-CSU. Allergy. 72:2005–2016. 2017.PubMed/NCBI View Article : Google Scholar

10 

O'Donnell BF, Lawlor F, Simpson J, Morgan M and Greaves MW: The impact of chronic urticaria on the quality of life. Br J Dermatol. 136:197–201. 1997.PubMed/NCBI

11 

Parisi CA, Ritchie C, Petriz N and Morelo Torres C: Direct medical costs of chronic urticaria in a private health organization of Buenos Aires, Argentina. Value Health Reg Issues. 11:57–59. 2016.PubMed/NCBI View Article : Google Scholar

12 

Broder MS, Raimundo K, Antonova E and Chang E: Resource use and costs in an insured population of patients with chronic idiopathic/spontaneous urticaria. Am J Clin Dermatol. 16:313–321. 2015.PubMed/NCBI View Article : Google Scholar

13 

Graham J, McBride D, Stull D, Halliday A, Alexopoulos ST, Balp MM, Griffiths M, Agirrezabal I, Zuberbier T and Brennan A: Cost utility of omalizumab compared with standard of care for the treatment of chronic spontaneous urticaria. Pharmacoeconomics. 34:815–827. 2016.PubMed/NCBI View Article : Google Scholar

14 

Church MK, Kolkhir P, Metz M and Maurer M: The role and relevance of mast cells in urticaria. Immunol Rev. 282:232–247. 2018.PubMed/NCBI View Article : Google Scholar

15 

Peteiro C and Toribio J: Incidence of leukocytoclastic vasculitis in chronic idiopathic urticaria. Study of 100 cases. Am J Dermatopathol. 11:528–533. 1989.PubMed/NCBI View Article : Google Scholar

16 

Ito Y, Satoh T, Takayama K, Miyagishi C, Walls AF and Yokozeki H: Basophil recruitment and activation in inflammatory skin diseases. Allergy. 66:1107–1113. 2011.PubMed/NCBI View Article : Google Scholar

17 

Kay AB, Clark P, Maurer M and Ying S: Elevations in T-helper-2-initiating cytokines (interleukin-33, interleukin-25 and thymic stromal lymphopoietin) in lesional skin from chronic spontaneous (‘idiopathic’) urticaria. Br J Dermatol. 172:1294–1302. 2015.PubMed/NCBI View Article : Google Scholar

18 

Kay AB, Ying S, Ardelean E, Mlynek A, Kita H, Clark P and Maurer M: Calcitonin gene-related peptide and vascular endothelial growth factor are expressed in lesional but not uninvolved skin in chronic spontaneous urticaria. Clin Exp Allergy. 44:1053–1060. 2014.PubMed/NCBI View Article : Google Scholar

19 

Zuberbier T, Schadendorf D, Haas N, Hartmann K and Henz BM: Enhanced P-selectin expression in chronic and dermographic urticaria. Int Arch Allergy Immunol. 114:86–89. 1997.PubMed/NCBI View Article : Google Scholar

20 

Kay AB, Ying S, Ardelean E, Mlynek A, Kita H, Clark P and Maurer M: Elevations in vascular markers and eosinophils in chronic spontaneous urticarial weals with low-level persistence in uninvolved skin. Br J Dermatol. 171:505–511. 2014.PubMed/NCBI View Article : Google Scholar

21 

Greaves MW: Chronic urticaria. N Engl J Med. 332:1767–1772. 1995.PubMed/NCBI View Article : Google Scholar

22 

Kaplan AP: Clinical practice. Chronic urticaria and angioedema. N Engl J Med. 346:175–179. 2002.PubMed/NCBI View Article : Google Scholar

23 

Hermes B, Prochazka AK, Haas N, Jurgovsky K, Sticherling M and Henz BM: Upregulation of TNF-alpha and IL-3 expression in lesional and uninvolved skin in different types of urticaria. J Allergy Clin Immunol. 103:307–314. 1999.PubMed/NCBI View Article : Google Scholar

24 

Haas N, Schadendorf D and Henz BM: Differential endothelial adhesion molecule expression in early and late whealing reactions. Int Arch Allergy Immunol. 115:210–214. 1998.PubMed/NCBI View Article : Google Scholar

25 

Rorsman H: Basophilic leucopenia in different forms of urticaria. Acta Allergol. 17:168–184. 1962.PubMed/NCBI View Article : Google Scholar

26 

Gruber BL, Baeza ML, Marchese MJ, Agnello V and Kaplan AP: Prevalence and functional role of anti-IgE autoantibodies in urticarial syndromes. J Invest Dermatol. 90:213–217. 1988.PubMed/NCBI View Article : Google Scholar

27 

Grattan CE, Wallington TB, Warin RP, Kennedy CT and Bradfield JW: A serological mediator in chronic idiopathic urticaria-a clinical, immunological and histological evaluation. Br J Dermatol. 114:583–590. 1986.PubMed/NCBI View Article : Google Scholar

28 

Grattan CE, Boon AP, Eady RA and Winkelmann RK: The pathology of the autologous serum skin test response in chronic urticaria resembles IgE-mediated late-phase reactions. Int Arch Allergy Appl Immunol. 93:198–204. 1990.PubMed/NCBI View Article : Google Scholar

29 

Grattan CE, Francis DM, Hide M and Greaves MW: Detection of circulating histamine releasing autoantibodies with functional properties of anti-IgE in chronic urticaria. Clin Exp Allergy. 21:695–704. 1991.PubMed/NCBI View Article : Google Scholar

30 

Hide M, Francis DM, Grattan CE, Hakimi J, Kochan JP and Greaves MW: Autoantibodies against the high-affinity IgE receptor as a cause of histamine release in chronic urticaria. N Engl J Med. 328:1599–1604. 1993.PubMed/NCBI View Article : Google Scholar

31 

Kolkhir P, Metz M, Altrichter S and Maurer M: Comorbidity of chronic spontaneous urticaria and autoimmune thyroid diseases: A systematic review. Allergy. 72:1440–1460. 2017.PubMed/NCBI View Article : Google Scholar

32 

Puccetti A, Bason C, Simeoni S, Millo E, Tinazzi E, Beri R, Peterlana D, Zanoni G, Senna G, Corrocher R and Lunardi C: In chronic idiopathic urticaria autoantibodies against Fc epsilonRII/CD23 induce histamine release via eosinophil activation. Clin Exp Allergy. 35:1599–1607. 2005.PubMed/NCBI View Article : Google Scholar

33 

Hatada Y, Kashiwakura J, Hayama K, Fujisawa D, Sasaki-Sakamoto T, Terui T, Ra C and Okayama Y: Significantly high levels of anti-dsDNA immunoglobulin E in sera and the ability of dsDNA to induce the degranulation of basophils from chronic urticaria patients. Int Arch Allergy Immunol. 161 (Suppl 2):S154–S158. 2013.PubMed/NCBI View Article : Google Scholar

34 

Bracken SJ, Abraham S and MacLeod AS: Autoimmune theories of chronic spontaneous urticaria. Front Immunol. 10(627)2019.PubMed/NCBI View Article : Google Scholar

35 

Bansal CJ and Bansal AS: Stress, pseudoallergens, autoimmunity, infection and inflammation in chronic spontaneous urticaria. Allergy Asthma Clin Immunol. 15(56)2019.PubMed/NCBI View Article : Google Scholar

36 

Zhang M, Murphy RF and Agrawal DK: Decoding IgE Fc receptors. Immunol Res. 37:1–16. 2007.PubMed/NCBI View Article : Google Scholar

37 

Zuberbier T, Henz BM, Fiebiger E, Maurer D and Stingl G: Anti-FcepsilonRIalpha serum autoantibodies in different subtypes of urticaria. Allergy. 55:951–954. 2000.PubMed/NCBI View Article : Google Scholar

38 

Schmetzer O, Lakin E, Topal FA, Preusse P, Freier D, Church MK and Maurer M: IL-24 is a common and specific autoantigen of IgE in patients with chronic spontaneous urticaria. J Allergy Clin Immunol. 142:876–882. 2018.PubMed/NCBI View Article : Google Scholar

39 

Kolkhir P, Church MK, Weller K, Metz M, Schmetzer O and Maurer M: Autoimmune chronic spontaneous urticaria: What we know and what we do not know. J Allergy Clin Immunol. 139:1772–1781.e1. 2017.PubMed/NCBI View Article : Google Scholar

40 

Blank U, Ra C, Miller L, White K, Metzger H and Kinet JP: Complete structure and expression in transfected cells of high affinity IgE receptor. Nature. 337:187–189. 1989.PubMed/NCBI View Article : Google Scholar

41 

Turner H and Kinet JP: . Signalling through the high-affinity IgE receptor Fc epsilonRI. Nature. 402 (Suppl 6760):B24–B30. 1999.PubMed/NCBI View Article : Google Scholar

42 

Panaszek B, Pawłowicz R, Grzegrzółka J and Obojski A: Autoreactive IgE in chronic spontaneous/idiopathic urticaria and basophil/mastocyte priming phenomenon, as a feature of autoimmune nature of the syndrome. Arch Immunol Ther Exp (Warsz). 65:137–143. 2017.PubMed/NCBI View Article : Google Scholar

43 

Confino-Cohen R, Chodick G, Shalev V, Leshno M, Kimhi O and Goldberg A: Chronic urticaria and autoimmunity: Associations found in a large population study. J Allergy Clin Immunol. 129:1307–1313. 2012.PubMed/NCBI View Article : Google Scholar

44 

Rottem M: Chronic urticaria and autoimmune thyroid disease: Is there a link? Autoimmun Rev. 2:69–72. 2003.PubMed/NCBI View Article : Google Scholar

45 

Shin YS, Suh DH, Yang EM, Ye YM and Park HS: Serum specific IgE to thyroid peroxidase activates basophils in aspirin intolerant urticaria. J Korean Med Sci. 30:705–709. 2015.PubMed/NCBI View Article : Google Scholar

46 

Altrichter S, Peter HJ, Pisarevskaja D, Metz M, Martus P and Maurer M: IgE mediated autoallergy against thyroid peroxidase-a novel pathomechanism of chronic spontaneous urticaria? PLoS One. 6(e14794)2011.PubMed/NCBI View Article : Google Scholar

47 

Altman K and Chang C: Pathogenic intracellular and autoimmune mechanisms in urticaria and angioedema. Clinic Rev Allerg Immunol. 45:47–62. 2013.PubMed/NCBI View Article : Google Scholar

48 

Krystel-Whittemore M, Dileepan KN and Wood JG: Mast cell: A multi-functional master cell. Front Immunol. 6(620)2016.PubMed/NCBI View Article : Google Scholar

49 

da Silva EZ, Jamur MC and Oliver C: Mast cell function: A new vision of an old cell. J Histochem Cytochem. 62:698–738. 2014.PubMed/NCBI View Article : Google Scholar

50 

Galli SJ and Tsai M: Mast cells in allergy and infection: Versatile effector and regulatory cells in innate and adaptive immunity. Eur J Immunol. 40:1843–1851. 2010.PubMed/NCBI View Article : Google Scholar

51 

Jamur MC, Grodzki AC, Berenstein EH, Hamawy MM, Siraganian RP and Oliver C: Identification and characterization of undifferentiated mast cells in mouse bone marrow. Blood. 105:4282–4289. 2005.PubMed/NCBI View Article : Google Scholar

52 

Metcalfe DD and Boyce JA: Mast cell biology in evolution. J Allergy Clin Immunol. 117:1227–1229. 2006.PubMed/NCBI View Article : Google Scholar

53 

Collington SJ, Williams TJ and Weller CL: Mechanisms underlying the localisation of mast cells in tissues. Trends Immunol. 32:478–485. 2001.PubMed/NCBI View Article : Google Scholar

54 

Irani AA, Schechter NM, Craig SS, DeBlois G and Schwartz LB: Two types of human mast cells that have distinct neutral protease compositions. Proc Natl Acad Sci USA. 83:4464–4468. 1986.PubMed/NCBI View Article : Google Scholar

55 

Schwartz LB: Analysis of MC(T) and MC(TC) mast cells in tissue. Methods Mol Biol. 315:53–62. 2006.PubMed/NCBI

56 

Caproni M, Giomi B, Volpi W, Melani L, Schincaglia E, Macchia D, Manfredi M, D'Agata A and Fabbri P: Chronic idiopathic urticaria: Infiltrating cells and related cytokines in autologous serum-induced wheals. Clin Immunol. 114:284–292. 2005.PubMed/NCBI View Article : Google Scholar

57 

Caproni M, Giomi B, Melani L, Volpi W, Antiga E, Torchia D and Fabbri P: Cellular infiltrate and related cytokines, chemokines, chemokine receptors and adhesion molecules in chronic autoimmune urticaria: Comparison between spontaneous and autologous serum skin test induced wheal. Int J Immunopathol Pharmacol. 19:507–515. 2006.PubMed/NCBI View Article : Google Scholar

58 

Terhorst D, Koti I, Krause K, Metz M and Maurer M: In chronic spontaneous urticaria, high numbers of dermal endothelial cells, but not mast cells, are linked to recurrent angio-oedema. Clin Exp Dermatol. 43:131–136. 2018.PubMed/NCBI View Article : Google Scholar

59 

Haas N, Toppe E and Henz BM: Microscopic morphology of different types of urticaria. Arch Dermatol. 134:41–46. 1998.PubMed/NCBI View Article : Google Scholar

60 

Elias J, Boss E and Kaplan AP: Studies of the cellular infiltrate of chronic idiopathic urticaria: Prominence of T-lymphocytes, monocytes, and mast cells. J Allergy Clin Immunol. 78:914–918. 1986.PubMed/NCBI View Article : Google Scholar

61 

Czarnetzki BM, Zwadlo-Klarwasser GZ, Bröcker EB and Sorg C: Macrophage subsets in different types of urticaria. Arch Dermatol Res. 282:93–97. 1990.PubMed/NCBI View Article : Google Scholar

62 

English JS, Murphy GM, Winkelmann RK and Bhogal B: A sequential histopathological study of dermographism. Clin Exp Dermatol. 13:314–317. 1988.PubMed/NCBI View Article : Google Scholar

63 

Kessler S and Kuhn C: Scanning electron microscopy of mast cell degranulation. Lab Invest. 32:71–77. 1975.PubMed/NCBI

64 

Toyoda M, Maruyama T, Morohashi M and Bhawan J: Free eosinophil granules in urticaria: A correlation with the duration of wheals. Am J Dermatopathol. 18:49–57. 1996.PubMed/NCBI View Article : Google Scholar

65 

Irwin RB, Lieberman P, Friedman MM, Kaliner M, Kaplan R, Bale G, Treadwell G and Yoo TJ: Mediator release in local heat urticaria: Protection with combined H1 and H2 antagonists. J Allergy Clin Immunol. 76:35–39. 1985.PubMed/NCBI View Article : Google Scholar

66 

Mayou SC, Kobza Black A, Eady RA and Greaves MW: Cholinergic dermographism. Br J Dermatol. 115:371–377. 1986.PubMed/NCBI View Article : Google Scholar

67 

Mekori YA, Dobozin BS, Schocket AL, Kohler PF and Clark RA: Delayed pressure urticaria histologically resembles cutaneous late-phase reactions. Arch Dermatol. 124:230–235. 1988.PubMed/NCBI

68 

Arai I, Tsuji M, Takeda H, Akiyama N and Saito S: A single dose of interleukin-31 (IL-31) causes continuous itch-associated scratching behaviour in mice. Exp Dermatol. 22:669–671. 2013.PubMed/NCBI View Article : Google Scholar

69 

Bodoor K, Al-Qarqaz F, Heis LA, Alfaqih MA, Oweis AO, Almomani R and Obeidat MA: IL-33/13 axis and IL-4/31 axis play distinct roles in inflammatory process and itch in psoriasis and atopic dermatitis. Clin Cosmet Investig Dermatol. 13:419–424. 2020.PubMed/NCBI View Article : Google Scholar

70 

Dillon SR, Sprecher C, Hammond A, Bilsborough J, Rosenfeld-Franklin M, Presnell SR, Haugen HS, Maurer M, Harder B, Johnston J, et al: Interleukin 31, a cytokine produced by activated T cells, induces dermatitis in mice. Nat Immunol. 5:752–760. 2004.PubMed/NCBI View Article : Google Scholar

71 

Ezzat MH, Hasan ZE and Shaheen KY: Serum measurement of interleukin-31 (IL-31) in paediatric atopic dermatitis: Elevated levels correlate with severity scoring. J Eur Acad Dermatol Venereol. 25:334–339. 2011.PubMed/NCBI View Article : Google Scholar

72 

Furue M, Yamamura K, Kido-Nakahara M, Nakahara T and Fukui Y: Emerging role of interleukin-31 and interleukin-31 receptor in pruritus in atopic dermatitis. Allergy. 73:29–36. 2018.PubMed/NCBI View Article : Google Scholar

73 

Gonzales AJ, Humphrey WR, Messamore JE, Fleck TJ, Fici GJ, Shelly JA, Teel JF, Bammert GF, Dunham SA, Fuller TE and McCall RB: Interleukin-31: Its role in canine pruritus and naturally occurring canine atopic dermatitis. Vet Dermatol. 24:48–53.e11-e22. 2013.PubMed/NCBI View Article : Google Scholar

74 

Kim S, Kim HJ, Yang HS, Kim E, Huh IS and Yang JM: IL-31 serum protein and tissue mRNA levels in patients with atopic dermatitis. Ann Dermatol. 23:468–473. 2011.PubMed/NCBI View Article : Google Scholar

75 

Lin W, Zhou Q, Liu C, Ying M and Xu S: Increased plasma IL-17, IL-31, and IL-33 levels in chronic spontaneous urticaria. Sci Rep. 7(17797)2017.PubMed/NCBI View Article : Google Scholar

76 

Zhang Q, Putheti P, Zhou Q, Liu Q and Gao W: Structures and biological functions of IL-31 and IL-31 receptors. Cytokine Growth Factor Rev. 19:347–356. 2008.PubMed/NCBI View Article : Google Scholar

77 

Kasutani K, Fujii E, Ohyama S, Adachi H, Hasegawa M, Kitamura H and Yamashita N: Anti-IL-31 receptor antibody is shown to be a potential therapeutic option for treating itch and dermatitis in mice. Br J Pharmacol. 171:5049–5058. 2014.PubMed/NCBI View Article : Google Scholar

78 

Moussion C, Ortega N and Girard JP: The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: A novel ‘alarmin’? PLoS One. 3(e3331)2008.PubMed/NCBI View Article : Google Scholar

79 

Hsu CL, Neilsen CV and Bryce PJ: IL-33 is produced by mast cells and regulates IgE-dependent inflammation. PLoS One. 5(e11944)2010.PubMed/NCBI View Article : Google Scholar

80 

Saluja R, Khan M, Church MK and Maurer M: The role of IL-33 and mast cells in allergy and inflammation. Clin Transl Allergy. 5(33)2015.PubMed/NCBI View Article : Google Scholar

81 

Morita H, Arae K, Unno H, Miyauchi K, Toyama S, Nambu A, Oboki K, Ohno T, Motomura K, Matsuda A, et al: An interleukin-33-mast cell-interleukin-2 axis suppresses papain-induced allergic inflammation by promoting regulatory T cell numbers. Immunity. 43:175–186. 2015.PubMed/NCBI View Article : Google Scholar

82 

Hongjia L, Caiqing Z, Degan L, Fen L, Chao W, Jinxiang W and Liang D: IL-25 promotes Th2 immunity responses in airway inflammation of asthmatic mice via activation of dendritic cells. Inflammation. 37:1070–1077. 2014.PubMed/NCBI View Article : Google Scholar

83 

Ikeda K, Nakajima H, Suzuki K, Kagami S, Hirose K, Suto A, Saito Y and Iwamoto I: Mast cells produce interleukin-25 upon Fc epsilon RI-mediated activation. Blood. 101:3594–3596. 2003.PubMed/NCBI View Article : Google Scholar

84 

Hepworth MR, Maurer M and Hartmann S: Regulation of type 2 immunity to helminths by mast cells. Gut Microbes. 3:476–481. 2012.PubMed/NCBI View Article : Google Scholar

85 

West EE, Kashyap M and Leonard WJ: TSLP: A key regulator of asthma pathogenesis. Drug Discov Today Dis Mech. 9(10)2012.PubMed/NCBI View Article : Google Scholar

86 

Ying S, O'Connor B, Ratoff J, Meng Q, Fang C, Cousins D, Zhang G, Gu S, Gao Z, Shamji B, et al: Expression and cellular provenance of thymic stromal lymphopoietin and chemokines in patients with severe asthma and chronic obstructive pulmonary disease. J Immunol. 181:2790–2798. 2008.PubMed/NCBI View Article : Google Scholar

87 

Ying S, O'Connor B, Ratoff J, Meng Q, Mallett K, Cousins D, Robinson D, Zhang G, Zhao J, Lee TH and Corrigan C: Thymic stromal lymphopoietin expression is increased in asthmatic airways and correlates with expression of Th2-attracting chemokines and disease severity. J Immunol. 174:8183–8190. 2005.PubMed/NCBI View Article : Google Scholar

88 

Hong GU, Ro JY, Bae Y, Kwon IH, Park GH, Choi YH and Choi JH: Association of TG2 from mast cells and chronic spontaneous urticaria pathogenesis. Ann Allergy Asthma Immunol. 117:290–297. 2016.PubMed/NCBI View Article : Google Scholar

89 

Maurer M, Altrichter S, Metz M, Zuberbier T, Church MK and Bergmann KC: Benefit from reslizumab treatment in a patient with chronic spontaneous urticaria and cold urticaria. J Eur Acad Dermatol Venereol. 32:e112–e113. 2018.PubMed/NCBI View Article : Google Scholar

90 

Chang TW, Chen C, Lin CJ, Metz M, Church MK and Maurer M: The potential pharmacologic mechanisms of omalizumab in patients with chronic spontaneous urticaria. J Allergy Clin Immunol. 135:337–342. 2015.PubMed/NCBI View Article : Google Scholar

91 

Altrichter S, Hawro T, Hänel K, Czaja K, Lüscher B, Maurer M, Church MK and Baron JM: Successful omalizumab treatment in chronic spontaneous urticaria is associated with lowering of serum IL-31 levels. J Eur Acad Dermatol Venereol. 30:454–455. 2016.PubMed/NCBI View Article : Google Scholar

92 

Stellato C, de Paulis A, Ciccarelli A, Cirillo R, Patella V, Casolaro V and Marone G: Anti-inflammatory effect of cyclosporin A on human skin mast cells. J Invest Dermatol. 98:800–804. 1992.PubMed/NCBI View Article : Google Scholar

93 

Harrison CA, Bastan R, Peirce MJ, Munday MR and Peachell PT: Role of calcineurin in the regulation of human lung mast cell and basophil function by cyclosporine and FK506. Br J Pharmacol. 150:509–518. 2007.PubMed/NCBI View Article : Google Scholar

94 

Grattan CE, O'Donnell BF, Francis DM, Niimi N, Barlow RJ, Seed PT, Kobza Black A and Greaves MW: Randomized double-blind study of cyclosporin in chronic ‘idiopathic’ urticaria. Br J Dermatol. 143:365–372. 2000.PubMed/NCBI View Article : Google Scholar

95 

Vena GA, Cassano N, Colombo D, Peruzzi E and Pigatto P: Neo-I-30 Study Group. Cyclosporine in chronic idiopathic urticaria: A double-blind, randomized, placebo-controlled trial. J Am Acad Dermatol. 55:705–709. 2006.PubMed/NCBI View Article : Google Scholar

96 

Kulthanan K, Chaweekulrat P, Komoltri C, Hunnangkul S, Tuchinda P, Chularojanamontri L and Maurer M: Cyclosporine for chronic spontaneous urticaria: A meta-analysis and systematic review. J Allergy Clin Immunol Pract. 6:586–599. 2018.PubMed/NCBI View Article : Google Scholar

97 

Doshi DR and Weinberger MM: Experience with cyclosporine in children with chronic idiopathic urticaria. Pediatr Dermatol. 26:409–413. 2009.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2022
Volume 23 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dobrican C, Muntean IA, Pintea I, Petricău C, Deleanu D and Filip GA: Immunological signature of chronic spontaneous urticaria (Review). Exp Ther Med 23: 381, 2022
APA
Dobrican, C., Muntean, I.A., Pintea, I., Petricău, C., Deleanu, D., & Filip, G.A. (2022). Immunological signature of chronic spontaneous urticaria (Review). Experimental and Therapeutic Medicine, 23, 381. https://doi.org/10.3892/etm.2022.11309
MLA
Dobrican, C., Muntean, I. A., Pintea, I., Petricău, C., Deleanu, D., Filip, G. A."Immunological signature of chronic spontaneous urticaria (Review)". Experimental and Therapeutic Medicine 23.6 (2022): 381.
Chicago
Dobrican, C., Muntean, I. A., Pintea, I., Petricău, C., Deleanu, D., Filip, G. A."Immunological signature of chronic spontaneous urticaria (Review)". Experimental and Therapeutic Medicine 23, no. 6 (2022): 381. https://doi.org/10.3892/etm.2022.11309