Open access

Introductory Chapter: The Multispectrum Faces of Atopic Dermatitis

Written By

Celso Pereira

Submitted: 17 September 2020 Published: 05 May 2021

DOI: 10.5772/intechopen.95394

From the Edited Volume

Atopic Dermatitis - Essential Issues

Edited by Celso Pereira

Chapter metrics overview

469 Chapter Downloads

View Full Metrics

1. Introduction

Atopic dermatitis is a chronic or recurrent inflammatory skin disease usually related to the atopic march and atopic morbidity. Although the onset of symptoms occurs predominantly at pediatric ages, the disease can start at any age and even the elderly.

A constellation of conditioning factors has been identified, most of them sustained by genetic and epigenetic aspects favoring a Th2 cell profile, intrinsically associated with many other factors of the skin itself, namely the localized immunoinflammatory responses, the skin barrier dysfunction, dysbiosis, neuroimmune dysregulation and obviously environmental determinants and many others conditions [1]. As a consequence, different phenotypes and endotypes result from multiple heterogeneous and complex pathogenic mechanisms that are clinically expressed with different levels of severity and in the specific location of the lesions [2].

At pediatric ages, the thickness of the skin is markedly reduced compared to adults, as well as the population and diversity of resident cells [3]. In case of skin inflammatory disease the homeostatic disruption on cells as well on the matrix structures determines intrinsic changes in the skin’s adaptive immunity facilitating an inducible skin-associated lymphoid tissue (iSALT) with critical consequences [4].

The microbiome has a particular interest in all medical areas and aim of profuse scientific interest in the last decade. However, it was the results of the Sanford study that surprisingly came to prove the magnitude and diversity of cell-free DNA in our body [5]. In fact, the placental microbiome is decisive in the fetal period. So, gut and skin newborn microbial flora is markedly dependent on the maternal clinical condition and the type of delivery, eutocic or instrumented [6, 7]. However, the postnatal period, depending on intrinsic and exogenous factors, will allow the acquisition of new microorganisms. It therefore occurs a process of specialization in niches, and the subsequently interaction with the host comes to allow functional differentiations in this own flora having obvious consequences on mucosal structures [7].

The skin microbiota in the healthy individuals is highly variable between designated areas of moist, such as the surfaces of the antecubital and popliteal fossae, and areas of naturally drier skin or sebaceous skin areas, particularly regarding the proportions of the Phyla Actinobacteria, Firmicutes, Proteocbacteria e Bacteriodetes [8].

Dysbiosis in atopic dermatitis resulting from increased expression of Staphylococcus genera, particularly S aureus species, has been consensual for a long time and the acute outbreaks had been frequently described corroborate this increase with a parallel decreases in Streptococcus or Propionibacterium species [9]. The overexposure in the skin of S aureus and, less frequently to Malassezia furfur, results on environment specific conditions that favor a local immunoinflammatory hyperactivation in opposition to the protective effect of Acinetobacter species [10].

Atopic dermatitis also has an apparent correlation between the intestinal axis and the cutaneous mucosa, with over-expression of Clostridium difficile and Faecalibacterium prausnitzii species, reduction of short-chain fatty acids metabolites, which may enable future and potential extra-cutaneous therapeutic implications [2, 9, 11, 12].

This recent data amplify the complexity of the pathophysiology and immunological mechanisms in atopic dermatitis already accepted, namely: the skin barrier dysfunction, the type 2 inflammatory immune response and other important cell axes such as the dependents of cell ways Th22, Th1, Th17 or JAK–STAT signaling pathway [13].

The enormous complexity of pathways and mechanisms involved in atopic dermatitis is based on a huge heterogeneity of polygenic determinisms, so the genome-wide association studies (GWAS) only allow confirming the heritability identification in a very limited number of patients. The best documented gene mutation is related to filaggrin, but is only present in around 30% of the European population [14]. Concerning skin barrier dysfunction other genes has been also described (loricrin, keratin-16 periplakin e SPINK5/LEKTI), but it requires more research to assess the magnitude and prevalence in a large patients samples studies [2]. Likewise for a typical type T2 inflammation the dysfunctions of IL-4 and IL-13 loci are also far from being present in all patients [2].

Pruritus is one of the hallmarks of atopic dermatitis and can have a dramatic and severe impact on the quality of life of patients at all ages. From a clinical and pathophysiological point of view, the pruritus induced by histaminergic pathway is not a standard feature in opposition to other atopic diseases. In addition to the classic inflammatory type T2 cytokines (IL-4 and IL-13), the IL31 and, to a lesser extent, TSLP, are strictly related to pruritus signaling, particularly in severe forms. Despite the presence of IL-4Rα in afferent neural fibers, these neural structures also have IL-31Rα receptors, activated by the release of this cytokine by T2 cells, but also by mast cells, dendritic cells and activated keratinocytes [15].

Thus, pruritus is, rather than a clinical sign dependent on the inflammatory process, but itself an active intervening part on the pathogenic mechanism, allowing increased keratinocyte IL-31 release by damage and scratching, and also an increased activation of dermal dendritic cells via TSLP. Symptomatic control is, therefore, decisive, in terms of the immense discomfort experienced by the patient and also because allow a negative feedback to the underlying skin inflammation [13].

In the light of the above, inhibition of IL-4Rα receptors by dupilumab allows for a significant control of pruritus thresholds in a very expressive number of patients. However, in others, this control is not achieved. Trials with nemolizumab, an anti-IL-31Rα monoclonal, look like it is highly effective in controlling itching, but with significant clinically adverse effects [15].

In an interesting experimental model with human mast cell lines, an alcoholic extract of Commiphora myrrha reduced not only the release of histamine, but also the production and release of IL-31 by kinase suppressor regulated by an extracellular signal and activation of NF κB [16]. If these results are confirmed in vivo, this essential oil could have an enormous impact in the routine clinic associated with a reduced burden economic impact.

The treatments currently recommended by different guidelines and consensus from different international scientific societies are strictly guiding in pharmacological and non-pharmacological care plans, namely: general measures, hydration to minimize barrier defects, topical treatments, systemic anti-inflammatory drugs for specific conditions and biological treatment for severe forms. Naturally, many of the new therapies are very expensive, which limits their access to many patients around the world [2, 13, 17].

However, several articles have pointed out other strategies that appear to be promising and highly elective in atopic dermatitis.

Emollients are essential to care, and it has been suggested an additional inclusion of ceramides or plant extracts with anti-inflammatory activity, despite the need for robust evidence [18, 19, 20].

Regarding anti-inflammatory therapy, the different classes of corticosteroids, calcineurin inhibitors and an inhibitor of the intracellular enzyme phosphodiesterase 4 represent the drugs currently available for topical treatment. Inhibitors of the JAK–STAT and tyrosine kinase pathways are currently in clinical trials. Delgocitinib (a pan-JAK inhibitor) has recently been approved in Japan for the treatment of adults with atopic dermatitis [2, 21].

Regarding systemic therapy it is recommended to severe presentations of atopic dermatitis, and in recent years it has been seen a profusion of new approaches with new biological drugs, some of them already available and many others in clinical trials [22, 23]. This topic is developed in a specific chapter in this book.

The spectrum of action of these new drugs, targeting glycoproteins (IgE), cytokines, chemokines (or their receptors), cell signaling pathways and cell surface receptors, should be submitted to a extensive drug-safety monitoring and rigourous pharmacovigilance programs, because most of them are generally ubiquitous in multiple cells, tissues and organs and could have potential uncontrolled consequences, even in short-term treatments. In addition, this profusion of new drugs is not accompanied by biomarkers that allow a specific selection for different phenotypes and endotypes, in a pathology as heterogeneous as atopic dermatitis.

The new data that comes from the knowledge of dysbiosis in atopic dermatitis must be consistent with new research lines. These findings will allow new therapeutic approaches in earlier stages and more primary pathophysiological conditions that support the immune-inflammatory process and support the chronicity and clinical severity.

The presence of cutaneous biofilms in severe clinical forms and with skin recurrent infections and impetiginization has extremely relevance on the clinical point of view [24]. Current treatment strategies do not establish nor allow the elimination of these structures on the skin surface. However, a growing number of in vivo and in vitro investigations have demonstrated that topical extracts of some medicinal plants had the ability of biofilms dysruption from bacteria (Staphilococcus spp e Streptococcus spp) or fungi (Malassesia furfur and Candida spp) [24, 25].

Like many other chronic conditions in which dysbiosis is subject to extensive research, also in allergic diseases and atopic dermatitis in particular, several studies sustain that in the treatment plan new approaches will be necessary, some of which with very promising in vitro and in vivo results [26]. There are countless potential manipulation and modulation possibilities of dysbiosis, but additional studies are still necessary to prove the efficiency and safety criteria [27].

Topical application of bacterial lysates or bacteriophage-derived enzymes seems to be one of the most feasible strategies. This strategy tends to, selective, replace strains considered commensal identified on the skin of healthy individuals or others comproved strains with competing effect for S aureus [27]. Another researches address the topical application of non-replicating probiotics that may allow a negative modulation in the viability of dysbiotic bacterial flora [28].

Bacterial lysates administrated sublingually have been in clinical use since the 1970s, for the prevention of infectious risk in respiratory pathology. Resulting from lysis of heat-inactivated bacterial strains, the composition can be selected and adjusted to a very diverse number of strains [29]. Without an obvious reason, its use in the clinic was markedly decrease in the late 1990s, but it has recently been a subject of interest and rediscovery given the potential of the mechanisms of action. In this context, the designation of “trained immunity-based vaccines” has been proposed since these vaccines allow effects on both innate and adaptive immunity [30, 31]. Among the most relevant mechanisms are the antimicrobial cytokine response, overexpression of TLRs; production of specific IgG and induction of regulatory T responses and regulation of dendritic cells [31]. In patients with strongly impetiginized forms, there is the possibility of personalized prescription with one extract containing strains adjusted to dysbiosis. This formulation administred at sublingual region, adjacent to the MALT structure, is highly promising from an immune point of view, compared to the oral formulas, which is usually restricted to formulations and fixed compositions of lysates [32].

Other potential treatment strategies involve the autologous bacterial transplant, allogeneic bacterial transplant or even fecal microbial transplantation. On these grounds, further and roboust research is necessary [33, 34, 35].

Likewise, well-designed studies are needed to define the position and the role of probiotics, prebiotics or symbiotics in atopic dermatitis. This subject is developed in a chapter of this book, as well the specific criteria for phototherapy in select patients.

In view of the enormous clinical heterogeneity, of the physiopathological mechanisms, the treatment of atopic dermatitis is a real concern over the opportunities and therapeutic options already available and all other strategies under development and trials [2, 16, 17, 18, 19, 20, 21, 22, 23, 36, 37].

So, nowadays the atopic dermatitis treatment is a real challenge in view of the enormous clinical heterogeneity, the physiopathological mechanisms, the immunogenic aspects and the tremendous variability regarding age, race and ethnicity [37].

In this context, the opportunities with the available therapeutics and all the other strategies under development will certainly allow us to increasingly personalize the management plan and in a future that is believed to be close the identification of robust biomarkers for phenotypes and endotyps, as also on monitoring approach.

The scientific review of some of these aspects in the chapters of this book will certainly be tools of great interest and importance in the knowledge of a pathology with colossal prevalence and a tremendous impact on the quality of life of these patients and their families.

References

  1. 1. Paller AS, Spergel JM, Mina-Osorio P, Irvine AD. The atopic march and atopic multimorbidity: Many trajectories, many pathways. The Journal of Allergy and Clinical Immunology. 2019;143(1):46-55
  2. 2. Langan SM, Irvine AD, Weidinger S. Atopic dermatitis. Lancet. 2020;396(10247):345-360
  3. 3. Saitoh A. Aizawa Y2 Sato I, Hirano H, Sakai T, Mori M. skin thickness in young infants and adolescents: Applications for intradermal vaccination. Vaccine. 2015;33(29):3384-3391
  4. 4. Egawa G, Kabashima K. Role of lymphoid structure in skin immunity. Current Topics in Microbiology and Immunology. 2020;426:65-82
  5. 5. Kowarsky M, Camunas-Soler J, Kertesz M, et al. Numerous uncharacterized and highly divergent microbes which colonize humans are revealed by circulating cell-free DNA. Proceedings of the National Academy of Sciences of the United States of America. 2017;114(36):9623-9628
  6. 6. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta harbors a unique microbiome. Sci Transl Med. 2014;6(237):237ra65.
  7. 7. Davenport ER, Sanders JG, Song SJ, Amato KR, Clark AG, Knight R. The human microbiome in evolution. BMC Biology. 2017;15(1):127
  8. 8. Chen YE, Tsao H. The skin microbiome: Current perspectives and future challenges. Journal of the American Academy of Dermatology. 2013;69(1):143-155
  9. 9. Pascal M, Perez-Gordo M, Caballero T, et al. Microbiome and allergic diseases. Frontiers in Immunology. 2018;9:1584
  10. 10. Fyhrquist N, Ruokolainen L, Suomalainen A, Lehtimaki S, Veckman V, Vendelin J, et al. Acinetobacter species in the skin microbiota protect against allergic sensitization and inflammation. J Allergy Clin Immunol. 2014; 134(6):1301-1309.e11.
  11. 11. Melli LCFL, Carmo-Rodrigues MS, Araújo-Filho HB, et al. Gut microbiota of children with atopic dermatitis: Controlled study in the metropolitan region of São Paulo, Brazil. Allergol Immunopathol (Madr). 2020;48(2):107-115
  12. 12. Pothmann A, Illing T, Wiegand C, Hartmann AA, Elsner P. The microbiome and atopic dermatitis: A review. American Journal of Clinical Dermatology. 2019;20(6):749-761
  13. 13. Munera-Campos M, Carrascosa JM. Innovation in atopic dermatitis: From pathogenesis to treatment. Actas Dermo-Sifiliográficas. 2020;111(3):205-221
  14. 14. Drislane C, Irvine AD. The role of filaggrin in atopic dermatitis and allergic disease. Annals of Allergy, Asthma & Immunology. 2020;124(1):36-43
  15. 15. Bağci IS, Ruzicka T. IL-31: A new key player in dermatology and beyond. The Journal of Allergy and Clinical Immunology 2018;141(3):858-866.
  16. 16. Shin JY, Che DN, Byoung BO, Kang HJ, Kim J, Jang SI. Commiphora myrrha inhibits itch-associated histamine and IL-31 production in stimulated mast cells. Experimental and Therapeutic Medicine. 2019;18(3):1914-1920
  17. 17. Newsom M, Bashyam AM, Balogh EA, Feldman SR, Strowd LC. New and emerging systemic treatments for atopic dermatitis. Drugs. 2020;80(11):1041-1052
  18. 18. Lowe A, Su J, Tang M, et al. PEBBLES study protocol: A randomised controlled trial to prevent atopic dermatitis, food allergy and sensitisation in infants with a family history of allergic disease using a skin barrier improvement strategy. BMJ Open. 2019;9:e024594
  19. 19. Lin T-K, Zhong L, Santiago JL. Anti-inflammatory and skin barrier repair effects of topical application of some plant oils. International Journal of Molecular Sciences. 2018;19(1):70
  20. 20. Ornelas J, Routt E, Kallis P, Lev-Tov H. Use of the hCONSORT criteria as a reporting standard for herbal interventions for common dermatoses: A systematic review. The British Journal of Dermatology. 2018;178(4):889-896
  21. 21. Dhillon S. Delgocitinib: First approval. Drugs. 2020 Apr;80(6):609-615
  22. 22. Newsom M, Bashyam AM, Balogh EA, Feldman SR, Strowd LC. New and emerging systemic treatments for atopic dermatitis. Drugs. 2020;80(11):1041-1052
  23. 23. Renert-Yuval Y, Guttman-Yassky E. New treatments for atopic dermatitis targeting beyond IL-4/IL-13 cytokines. Annals of Allergy, Asthma & Immunology. 2020;124(1):28-35
  24. 24. Gonzalez T, Biagini-Myers JM, Herr AB, Khurana-Hershey GK. Staphylococcal biofilms in atopic dermatitis. Current Allergy and Asthma Reports. 2017;17(12):81
  25. 25. Melanda H. Biofilmes e plantas medicinais: evidência científica. In Master Thesis. Medicinal Plants in Clinical Practice Unity, Celso Pereira. Medicine Faculty. Coimbra University. 2019 (April):1-72.
  26. 26. Myles IA. Allergy as a disease of dysbiosis: Is it time to shift the treatment paradigm? Frontiers in Cellular and Infection Microbiology. 2019;9:50
  27. 27. Hendricks AJ, Mills BW, Shi VY. Skin bacterial transplant in atopic dermatitis: Knowns, unknowns and emerging trends. Journal of Dermatological Science. 2019;95(2):56-61
  28. 28. Rosignoli C, Thibaut de Menonville S, Orfila D, et al. A topical treatment containing heat-treated lactobacillus johnsonii NCC 533 reduces Staphylococcus aureus adhesion and induces antimicrobial peptide expression in an in vitro reconstructed human epidermis model. Experimental Dermatology. 2018;27(4):358-365
  29. 29. Alecsandru D, Valor L, Sánchez-Ramón S, et al. Sublingual therapeutic immunization with a polyvalent bacterial preparation in patients with recurrent respiratory infections: Immunomodulatory effect on antigen-specific memory CD4+ T cells and impact on clinical outcome. Clinical and Experimental Immunology. 2011;164(1):100-107
  30. 30. Sánchez-Ramón S, Conejero L, Netea MG, Sancho D, Palomares O, Subiza JL. Trained immunity-based vaccines: A new paradigm for the development of broad-Spectrum anti-infectious formulations. Frontiers in Immunology. 2018;9:2936
  31. 31. Kearney SC, Dziekiewicz M, Feleszko W. Immunoregulatory and immunostimulatory responses of bacterial lysates in respiratory infections and asthma. Annals of Allergy, Asthma & Immunology. 2015;114(5):364-369
  32. 32. Lau S. Oral application of bacterial lysate in infancy diminishes the prevalence of atopic dermatitis in children at risk for atopy. Benef Microbes. 2014;5(2):147-149
  33. 33. Nakatsuji T, Chen TH, Narala S, et al. Antimicrobials from human skin commensal bacteria protect against Staphylococcus aureus and are deficient in atopic dermatitis. Science Translational Medicine. 2017;9:378
  34. 34. Perin B, Addetia A, Qin X. Transfer of skin microbiota between two dissimilar autologous microenvironments: A pilot study. PLoS One. 2019;14(12):e0226857
  35. 35. Mashiah J. Efficacy of Fecal Microbial Transplantation Treatment in Adults With Atopic Dermatitis. Tel-Aviv Sourasky Medical Center. 2020. ClinicalTrials.gov Identifier: NCT04283968.
  36. 36. Li Q, Fang H, Dang E, Wang G. The role of ceramides in skin homeostasis and inflammatory skin diseases. Journal of Dermatological Science. 2020;97(1):2-8
  37. 37. Nomura T, Wu J, Kabashima K, Guttman-Yassky E. Endophenotypic variations of atopic dermatitis by age, race. and Ethnicity J Allergy Clin Immunol Pract. 2020;8(6):1840-1852

Written By

Celso Pereira

Submitted: 17 September 2020 Published: 05 May 2021