eISSN: 2299-0046
ISSN: 1642-395X
Advances in Dermatology and Allergology/Postępy Dermatologii i Alergologii
Current issue Archive Manuscripts accepted About the journal Editorial board Reviewers Abstracting and indexing Subscription Contact Instructions for authors Publication charge Ethical standards and procedures
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
4/2022
vol. 39
 
Share:
Share:
Review paper

Th1 regulatory events by infectious pathogens, herpes zoster and herpes simplex viruses: prospects for therapeutic options for atopic eczema

Yasuhiro Horiuchi
1

1.
Division of Dermatology, Tsuruse Orthopaedic Clinic, Saitama, Japan
Adv Dermatol Allergol 2022; XXXIX (4): 662-667
Online publish date: 2022/09/01
Article file
- Th1 regulatory.pdf  [0.22 MB]
Get citation
 
 

Introduction

Infections by pathogens, such as viruses and bacteria, are typically perceived as harmful, such as in cases of herpes (varicella) zoster [1] and herpes simplex virus infections [2]. The herpes (HZV) or varicella zoster virus (VZV) [3], which is a member of the α-herpesviridae subfamily, comprising a polyhedral capsid surrounded by a membranous envelope structure (Figure 1) [3], and is one of eight herpes viruses known to infect humans. It causes varicella (chickenpox) (Figure 1) [1], a disease most commonly affecting children, teenagers, and young adults, and herpes zoster (HZ) in adults, particularly the elderly. Herpes simplex viruses (HSV) [4] are categorized into two types: herpes type 1 (HSV-1, or oral herpes), is also a member of the α-herpesviridae subfamily, and its structure is composed of linear dsDNA, an icosahedral capsid with a spikey envelope (Figure 1) [4], and herpes type 2 (HSV-2, or genital herpes). Kaposi’s varicelliform eruption (VE), eczema herpeticum (Figure 1) [5], is a rare but severe disseminated infection, predominantly found accompanying HSV infections. It generally occurs at sites of skin damage [6], such as eczema, particularly, in severe atopic dermatitis (AD), occurring in about 3% of AD patients [5], long-term external use of steroids, or occasionally burns.

Figure 1

Schematic representation of infectious diseases caused by HZV and HSV

/f/fulltexts/PDIA/47673/PDIA-39-47673-g001_min.jpg

AD [7] is a complex pathology mainly characterized by immune response dysfunction [8], T helper type (Th)2 dominant conditions [9], overexpression of Th2 cytokines such as interleukin (IL)-4 and IL-13, which play a key role in type 2 inflammation [10], and lower interferon-γ (IFN-γ) protein production is also significant [11], as downregulation of Th1 [12] but lesional skin also shows a mixed type-1 and type-2 immune responsiveness [13]. AD patients also exhibit unbalanced dysbiosis of skin microbiota characterized by Staphylococcus aureus colonization [14, 15], an increased risk of developing bacterial and viral infections with a suspected low production of the antimicrobial peptide cathelicidin [16], and impairment of the skin barrier such as a deficit in filaggrin [5, 16]. These are deemed to be defects in functions of immune cells [17] such as dendritic cells (DC), natural killer (NK) cells, and regulatory T cells (Tregs) [18, 19]; in particular, the function of Tregs, which are responsible for initial response to infection [20], is important in these virus infections which play a pivotal role in immune regulation and are integral to the control of allergic responses [21]. Potential treatments can be designed to amplify these cells to suppress the allergic inflammatory cascade in AD [21]. Atopic conditions are aggravated as a result of insufficient IL-12 production by DCs [22]. Acquired functional impairment of Tregs in AD patients and the correlation between the increased frequency of Tregs and disease severity support their important role in AD pathogenesis [23].

Alternatively, clinical observation of an improvement in severe atopic skin lesions after the onset of varicella has been noted [24], the improved state continued for at least 2 to 3 months and up to 5 to 6 months. In my daily dermatological practice, the author has occasionally observed improvements associated with HZV or HSV infections, especially in those with Kaposi’s VE (eczema herpeticum) in AD patients. I have earlier reported remarkable improvement in the eczematous lesions of both axillar regions that caused Kaposi’s VE in an elderly patient [25] with senile atopy and a secondary erythroderma due to a predisposition to atopy. It took considerably longer for skin lesions at other sites to heal. So far, the author has noted that protective responses to these infectious agents are expected to improve atopic eczema. Based on these clinical observations, this study focuses on Th1 immunoregulatory events caused by infectious pathogens, particularly herpes viruses.

Immunoregulatory events in HZV infection

Varicella (Figure 1) is a primary infection of VZV, which becomes latent in the peripheral ganglia [1]. However, declining T cell immunity [26] in ageing individuals or those undergoing immune restrictive treatments can lead to VZV reactivation and the development of herpes zoster (Figure 1) [1]. Varicella immunization can provide the opportunity to analyse the kinetics of IL-10, IL-12, and IFN-γ production [27] elicited during primary in vivo sensitization with VZV proteins [28]. VZV antigens on infected cells may be processed by monocytes for presentation to T cells [29]. In our previous reports, we found that monocytosis [30, 31], suggested as part of the defence against infection, was observed in the leukocyte fraction of the peripheral blood, which is typically composed of 2–10% of all leukocytes in the human body. Monocytes play a role with their diverse functional properties to protect infection in mobilizing dendritic cells (DCs) [32]. Monocytes derived from DCs, which are recruited during infection defence, produce large amounts of IL-12 [33]. Monocytes serve multiple roles in immune function and are the largest type of leukocyte and can differentiate into macrophages and myeloid lineage DCs, characterized by a high level expression of the CD14 cell surface receptor (CD14+ CD16++ monocyte) [34]. CD14 [35] is a human monocyte differentiation antigen as the Toll-like co-receptor for the detection of pathogen-associated molecular patterns. After stimulation with microbial products, the CD14+CD16++ monocytes [34] produce high amounts of pro-inflammatory cytokines like tumour necrosis factor (TNF) and IL-12. IFN-γ is also released upon activation of Th1 NK cells [36], which take the role of the innate immune defence against infection [37], during protection against viral infections. It is assumed that Th1 induction is involved in the suppression of Th2 atopy conditions through this process. Although studies of immunological processes have advanced, monocyte immune function and the positive aspects of the human immune response system caused by viruses have not been widely examined. Moreover, IL-12 was discovered as a key immunoregulatory cytokine in various infections and is effective in fighting a wide range of viral infections [38, 39] and may promote a Th1 response and regulate Th1 stability [40]. As shown in our atopic mouse experiments with bacterial components of Streptococcus pyogenes [41], IL-12 was amplified. It has been confirmed that depending on these bacterial infections, an IL-12 inducer of the defence process exerts effects on Th1 conditions. In human varicella infection, Fujimura et al. [24] showed that a switch from Th2 to Th1 regulation occurred, supporting observations of atopic dermatitis improvement after varicella infection. Thus far, HZV may have contributed to the switch to Th1 dominance [24]. In such lesions, expression of Th1 type cytokines predominated [24], suggesting downregulation of Th2 dominant AD conditions. They also pointed out that IL-12 may regulate the switch of the recall response of allergen-specific T cells of atopic donors from a Th2- to a Th1-like phenotype in vitro [24]. Although the detailed mechanisms remain unclear, immunity is known to decline with age [42] and in those with cancer or various infectious diseases, and the side effects of drugs [43] are often observed in the elderly. These accumulated findings suggest that the deficiency in the immune response due to ageing and diseases such as atopy can be repaired and normalized despite a temporary immune response to protect against infection caused by HZV.

Immunoregulatory events in HSV infection

In HSV infections, the type I interferon (INF-α and β) signalling pathway [44] plays an important role in the innate immunity [45, 46] along with activated neutrophils, monocytes, macrophages, and dendritic cells (DCs). DCs [47] are antigen presenting cells that are important for pathogen recognition at sites of infection and for priming of protective HSV-specific T cells [48]. NK cells [49] play an important role in the host response against viral infections being able to kill virus-infected cells. The control of Tregs may also maintain the delicate balance between inflammation and healing in controlling HSV infections [18]. It is likely that children with AD may be susceptible to HSV due to reduced numbers of NK cells and a decrease of IL-2 receptors, a marker for lymphocyte activation, during early eczema herpeticum [50]. HSV infection in vitro also has been found to up-regulate the expression of IL-12 (p40) mRNA as a triggering event that biases HSV-specific immunity to a type 1 T cell response [51]. Patients with Kaposi’s VE have impaired the type II interferon (IFN-γ) production in the protective response to HSV [52]. The impaired INF-γ production may account for the abnormal immunopathogenesis of severe, intractable AD [11]. Patients with Kaposi’s VE (Figure 1) exhibit reduced IFN-γ production, which may contribute to an impaired immune response to HSV [11, 52]. Of note, some reports suggest that HSV increases the levels of Th2 cytokines, such as IL-4 [53] and IL-25 [54], which, in turn, promote HSV replication; in fact, IL-25 [54] was shown to promote HSV replication by inhibiting the expression of filaggrin, suggesting this protein as an aggravating factor in Kaposi’s VE. It has also been pointed out that CD14(dim)CD16(+) monocytes [20] have a compromised ability to produce pro-inflammatory cytokines. Therefore, Kaposi’s VE is attributed to a decrease in the immune function of the skin of AD patients [8]. Nevertheless, improvement in AD skin lesions of Kaposi’s VE cases suggests that immune response to HSV infection involves a specific immunomodulatory activating factor. On the whole, this field has not always been actively investigated. The authors have experimented with animal models to scientifically support an observed clinical event. We have previously reported an improvement in AD-like lesions by UV-inactivated HSV 1 [55] in a murine atopy model (Nc/Nga mice), supporting a remarkable improvement in eczema lesions at the site of Kaposi’s VE. On the other hand, Kawakami et al. [56] described a relationship between defective NK cell activity and development of HSV 1-induced severe skin lesions (eczema herpeticum) in eczematous Nc/Nga mice. Herpes virus disease symptoms in patients with deficiencies in NK cell activity may result in life-threatening conditions [49]. Human plasmacytoid dendritic cells (pDCs) in the activation of NK cells control such herpes virus infections [47]. So far, NK cell activation [17, 47] could be induced by HSV in some individuals with improved atopic skin lesions. The difference between our previously reported results [55] and those of Kawakami et al. [56] is likely due to the use of different methodologies. In our study [55], 2 × 105 pfu UV-inactivated HSV 1 per mouse was injected intra- and/or subcutaneously at six sites in the eczema skin lesions. Contrarily, in Kawakami’s experiments [56], 4.5 × 103 pfu (in a volume of 3 μl per site) of live HSV 1 per site was intradermally injected at four sites in the skin lesions. Although the dose of virus administered in the studies differed by a factor of 10, the live and inactivated viruses induced completely different immune responses. Furthermore, there may be some difficulty in deciding on the appropriate dosage of live virus for small laboratory animals. Dose-dependent differences in immune response must also be taken into account when comparing different studies [57]. We hope to further analyse the mechanism of eczema lesion improvement in Kaposi’s VE using the response against HSV infection.

Prospects of Th1 regulation by infectious pathogens

In our previous studies [41, 55], we have shown through animal experiments that allergic dermatitis can be alleviated not only by these viruses [55] but also by S. pyogenes components, such as OK-432 [41]. Findings [58] in support of this result have already been shown. These pathogens may have components that induce immunological events contributing to Th2 to Th1 shifts [41]. While the severity of the immune response in infectious diseases is important, humanity has succumbed to numerous infectious agents despite the development of vaccines [28]. Although the results of animal experiments cannot be immediately reproduced in humans, they have been evaluated continuously to increase their applicability in humans later on. Therefore, we will continue to study the benefits of the immune response in some infectious pathogens. There is also a worldwide need for immunological studies to ascertain the components of these pathogens that can suppress or improve the symptoms of allergies. If these beneficial components of pathogens are found, it may be possible to replenish and improve atopic and age-related immune dysfunction and maintain patients in a stable condition. Further advances in this field will require identification of the molecular components of pathogens [59] that determine the direction of immune response and act as suppressors of allergic dermatitis. Lundberg et al. [59] reported that HSV DNA or HSV-derived oligodeoxyribonucleotides (ODNs) can induce the production of inflammatory cytokines, such as IFN-γ, which regulate Th1 response. Moreover, the augmentation of Th1 responses by bacterial cellular components such as lipopolysaccharides (LPS) in Gram-negative bacteria and peptidoglycans and lipopeptides in gram-positive bacteria, has also been highlighted [59]. As our study using components of S. pyogenes showed, lipoteichoic acid-related molecules [60] induced improvement of AD-like skin lesions in Nc/Nga mice. AD cases effectively treated with S. pyogenes bacterial extracts, such as OK-432 [61], have been previously reported. T lymphocytes recognize VZV glycoproteins (gpI-V), the immediate early/tegument protein, and the product of gene 62 (IE62) [62]. Thus far, identification of herpes virus components regulating such Th1 shifts are yet to be elucidated. Alternatively, the attenuated VZV vaccine (Oka/Merck) and inactivated zoster vaccine, prepared by heat or irradiation, recruit amplification of VZV-specific T-cell mediated responses with the enhancement of the vaccine-induced IFN-γ [63]. It may be worthwhile not only to use vaccines as protection against infection, but also for immunostimulation with Th1 induction. This may be effective in elderly people [42, 63], whose immune response has declined due to age. In atopy patients [64], as reported by Foerster and Molêda [65], vaccine targeting IL-13, a Th2 cytokine, may prove beneficial.

Conclusions

From the accumulated findings discussed here, both HZV and HSV may induce Th1 regulatory events along with the protective immune response to infection, although HZV appears to induce more systemic immunostimulation. HSV appears to be a local response in affected skin lesions. To date, great progress in immunopathological analysis in allergic diseases and AD has been made, but much work remains to elucidate the mechanism underlying this phenomenon and establish a reliable and safe treatment strategy. Induction of Th1 response by these viruses should be evaluated to improve the complex immune responses induced by the mobilization of Th1-regulating cytokines such as INF-γ and IL-12, to improve the symptoms of Th2-dominated AD. Although it is also true that these viral infections are nearly always harmful, there are rare cases in which atopic eczematous lesions improve upon exposure to these viral infections. However, it is possible that immune responses to the infectious agent may vary among individuals. Analysis of the immune events induced by these viral infections would provide novel insights for atopic therapy. It seems reasonable to suppose that infectious pathogens are not necessarily all bad, and further analysis in this field is required to uncover the potential therapeutic benefits of some pathogens.

Acknowledgments

The author would like to thank Editage for English language editing.

Conflict of interest

The author declares no conflict of interest.

References

1 

Kennedy PGE, Gershon AA. Clinical features of varicella-zoster virus infection. Viruses 2018; 10: E609.

2 

Kriesel JD, Hull CM. Herpes simplex virus infection. In: Netter’s Infectious Diseases. Jong EC, Stevens DL, (eds). Elsevier, Amsterdam 2012.

3 

Zerboni L, Sen N, Stefan L, Arvin AM. Molecular mechanisms of varicella zoster virus pathogenesis. Nat Rev Microbiol 2014; 12: 197-210.

4 

Saleh D, Sharma S. Herpes simplex type 1. In: Sharma S (ed). StatPearls [Internet]. StatPearls Publishing, Florida. Cited 2019 Dec 27. https://www.statpearls.com/as/infectious/22839/

5 

Damour A, Garcia M, Seneschal J, et al. Eczema herpeticum: clinical and pathophysiological aspects. Clin Rev Allergy Immunol 2020; 59: 1-18.

6 

Wollenberg A, Zoch C, Wetzel S, et al. Predisposing factors and clinical features of eczema herpeticum: a retrospective analysis of 100 cases. J Am Acad Dermatol 2003; 49: 198-205.

7 

Fiset PO, Leung DY, Hamid Q. Immunopathology of atopic dermatitis. J Allergy Clin Immunol 2006; 118: 287-90.

8 

De Benedetto A, Agnihothri R, McGirt LY, et al. Atopic dermatitis: a disease caused by innate immune defects. J Invest Dermatol 2009; 129: 14-30.

9 

Brandt EB, Sivaprasad U. Th2 cytokines and atopic dermatitis. J Clin Cell Immunol 2011; 2: pii:110.

10 

Bieber T. Interleukin-13: targeting an underestimated cytokine in atopic dermatitis. Allergy 2020; 75: 54-62.

11 

Katsunuma T, Kawahara H, Yuki K, et al. Impaired interferon-gamma production in a subset population of severe atopic dermatitis. Int Arch Allergy Immunol 2004; 134: 240-7.

12 

Brunner PM. Early immunologic changes during the onset of atopic dermatitis. Ann Allergy Asthma Immunol 2019; 123: 152-7.

13 

Pastar Z, Lipozencić J, Ljubojević S. Etiopathogenesis of atopic dermatitis: an overview. Acta Dermatovenerol Croat 2005; 13: 54-62.

14 

Di Domenico EG, Cavallo I, Capitanio B, et al. Staphylococcus aureus and the cutaneous microbiota biofilms in the pathogenesis of atopic dermatitis. Microorganisms 2019; 7: pii: E301.

15 

Ong PY, Leung DY. The infectious aspects of atopic dermatitis. Immunol Allergy Clin North Am 2010; 30: 309-21.

16 

Kopfnagel V, Harder J, Werfel T. Expression of antimicrobial peptides in atopic dermatitis and possible immunoregulatory functions. Curr Opin Allergy Clin Immunol 2013; 13: 531-6.

17 

Chattopadhyay D, Mukhopadhyay A, Ojha D, et al. Immuno-metabolic changes in herpes virus infection. Cytokine 2018; 112: 52-62.

18 

Levin MJ, Weinberg A. Regulatory T cells in infections: getting it just right. J Infect Dis 2016; 214: 4-5.

19 

Shiohara T, Sato Y, Takahashi R, et al. Increased susceptibility to cutaneous viral infections in atopic dermatitis: the roles of regulatory T cells and innate immune defects. Curr Probl Dermatol 2011; 41: 125-35.

20 

Takahashi R , Sato Y, Kurata M, et al. Pathological role of regulatory T cells in the initiation and maintenance of eczema herpeticum lesions. J Immunol 2014; 192: 969-78.

21 

Agrawal R, Wisniewski JA, Woodfolk JA. The role of regulatory T cells in atopic dermatitis. Curr Probl Dermatol 2011; 41: 112-24.

22 

Reider N, Reider D, Ebner S, et al. Dendritic cells contribute to the development of atopy by an insufficiency in IL-12 production. J Allergy Clin Immunol 2002; 109: 89-95.

23 

Gáspár K, Baráth S, Nagy G, et al. Regulatory T-cell subsets with acquired functional impairment: important indicators of disease severity in atopic dermatitis. Acta Derm Venereol 2015; 95: 151-5.

24 

Fujimura T, Yamanashi R, Masuzawa M, et al. Conversion of the CD4+ T cell profile from T(h2)-dominant type to T(h1)-dominant type after varicella-zoster virus infection in atopic dermatitis. J Allergy Clin Immunol 1997; 100: 274-82.

25 

Horiuchi Y. Kaposi’s varicelliform eruptions during the course of steroid withdrawal in a senile erythroderma patient: cure of regional erythrodermic lesions following infection. J Dermatol 1999; 26: 375-8.

26 

Gagliardi AM, Andriolo BN, Torloni MR, et al. Vaccines for preventing herpes zoster in older adults. Cochrane Database Syst Rev 2016; 3: CD008858.

27 

Jenkins DE, Redman RL, Lam EM, et al. Interleukin(IL)-10, IL-12, and interferon-gamma production in primary and memory immune responses to varicella-zoster virus. J Infect Dis 1998; 178: 940-8.

28 

Weinberg A, Canniff J, Rouphael N, et al. Varicella-zoster virus-specific cellular immune responses to the live attenuated zoster vaccine in young and older adults. J Immunol 2017; 199: 604-12.

29 

Pontesilli O, Carotenuto P, Levin MJ, et al. Processing and presentation of cell-associated varicella-zoster virus antigens by human monocytes. Clin Exp Immunol 1987; 70: 127-35.

30 

Tsukahara T, Yaguchi A, Horiuchi Y. Significance of monocytosis in varicella and herpes zoster. J Dermatol 1992; 19: 94-8.

31 

Horiuchi Y. Mo2+ HLA-DR- monocytosis in varicella zoster virus infection. J Dermatol 1997; 24: 205-7.

32 

Kennedy JJ, Steain M, Slobedman B, Abendroth A. Infection and functional modulation of human monocytes and macrophages by varicella-zoster virus. J Virol 2019; 93: e01887-18.

33 

Muller-Berghaus J, Olson WC, Moulton RA, et al. IL-12 production by human monocyte-derived dendritic cells: looking at the single cell. J Immunother 2005; 28: 306-13.

34 

Ancuta P, Weiss L, Haeffner-Cavaillon N. CD14+CD16++ cells derived in vitro from peripheral blood monocytes exhibit phenotypic and functional dendritic cell-like characteristics. Eur J Immunol 2000; 30: 1872-83.

35 

Wu Z, Zhang Z, Lei Z, Lei P. CD14: biology and role in thepathogenesis of disease. Cytokine Growth Factor Rev 2019; 48: 24-31.

36 

Campbell TM, McSharry BP, Steain M, et al. Functional paralysis of human natural killer cells by alphaherpesviruses. PLoS Pathog 2019; 15: e1007784.

37 

Laing KJ, Ouwendijk WJD, Koelle DM, Verjans GMGM. Immunobiology of varicella-zoster virus infection. J Infect Dis 2018; 218 (Suppl 2): S68-74.

38 

Guo Y, Cao W, Zhu Y. Immunoregulatory functions of the IL-12 family of cytokines in antiviral systems. Viruses 2019; 11: pii: E772.

39 

Horiuchi Y, Nishioka K. IL-12 in varicella zoster and herpes simplex virus infection. J Dermatol 2002; 29: 391-3.

40 

Brombacher F, Kastelein RA, Alber G. Novel IL-12 family members shed light on the orchestration of Th1 responses. Trends Immunol 2003; 24: 207-12.

41 

Horiuchi Y, Bae SJ, Katayama I, Nishioka K. Therapeutic effects of streptococcal preparation OK-432 on atopic dermatitis-like lesions in Nc/Nga mice: possible shift from a Th2 to Th1-predominance. J Dermatol Sci 2004; 35: 187-97.

42 

Fuentes E, Fuentes M, Alarcón M, Palomo I. Immune system dysfunction in the elderly. An Acad Bras Cienc 2017; 89: 285-99.

43 

Davies EA, O’Mahony MS. Adverse drug reactions in special populations – the elderly. Br J Clin Pharmacol 2015; 80: 796-807.

44 

Amin I, Younas S, Afzal S, et al. Herpes simplex virus type 1 and host antiviral immune responses: an update. Viral Immunol 2019; 32: 424-9.

45 

Alandijany T. Host intrinsic and innate intracellular immunity during herpes simplex virus type 1 (HSV-1) infection. Front Microbiol 2019; 10: 2611.

46 

Su C, Zhang G, Zheng C. Evasion of host innate antiviral immunity by HSV-1, an update. Virol J 2016; 13: 38.

47 

Vogel K, Thomann S, Vogel B, et al. Both plasmacytoid dendritic cells and monocytes stimulate natural killer cells early during human herpes simplex virus type 1 infections. Immunology 2014; 143: 588-600.

48 

Bedoui S, Greyer M. The role of dendritic cells in immunity against primary herpes simplex virus infections. Front Microbiol 2014; 5: 533.

49 

De Pelsmaeker S, Romero N, Vitale M, Favoreel HW. Herpes virus evasion of natural killer cells. J Virol 2018; 92: pii: e02105-17.

50 

Goodyear HM, McLeish P, Randall S, et al. Immunological studies of herpes simplex virus infection in children with atopic eczema. Br J Dermatol 1996; 134: 85-93.

51 

Kanangat S, Thomas J, Gangappa S, et al. Herpes simplex virus type 1 mediated up-regulation of IL-12 (p40) mRNA expression. Implications in immunopathogenesis and protection. J Immunol 1996; 156: 1110-6.

52 

Leung DY, Gao PS, Grigoryev DN, et al. Human atopic dermatitis complicated by eczema herpeticum is associated with abnormalities in IFN-γ response. J Allergy Clin Immunol 2011; 127: 965-73.e1-e5.

53 

Raychaudhuri SP, Raychaudhuri SK. Revisit to Kaposi’s varicelliform eruption: role of IL-4. Int J Dermatol 1995; 34: 854-6.

54 

Kim BE, Bin L, Ye YM, et al. IL-25 enhances HSV-1 replication by inhibiting filaggrin expression, and acts synergistically with Th2 cytokines to enhance HSV-1 replication. J Invest Dermatol 2013; 133: 2678-85.

55 

Horiuchi Y, Bae SJ, Katayama I, et al. Effects of ultraviolet inactivated herpes simplex virus type I on atopic dermatitis-like lesions in Nc/Nga mice: role of the suppressor of cytokine signaling in the skin. Allergol Int 2004; 53: 331-40.

56 

Kawakami Y, Ando T, Lee JR, et al. Defective natural killer cell activity in a mouse model of eczema herpeticum. J Allergy Clin Immunol 2017; 139: 997-1006.

57 

Yang K, Liang Y, Sun Z, et al. T cell-derived lymphotoxin limits Th1 response during HSV-1 infection. Sci Rep 2018; 8: 17727.

58 

Hovden AO, Karlsen M, Jonsson R, et al. Maturation of monocyte derived dendritic cells with OK432 boosts IL-12p70 secretion and conveys strong T-cell responses. BMC Immunol 2011; 12: 2.

59 

Lundberg P, Welander P, Han X, Cantin E. Herpes simplex virus type 1 DNA is immunostimulatory in vitro and in vivo. J Virol 2003; 77: 11158-69.

60 

Horiuchi Y, Bae SJ, Katayama I, et al. Lipoteichoic acid-related molecule derived from the streptococcal preparation, OK-432, which suppresses atopic dermatitis-like lesions in Nc/Nga mice. Arch Dermatol Res 2006; 298: 163-73.

61 

Horiuchi Y. Topical streptococcal preparation, OK-432, for atopic dermatitis. J Dermatolog Treat 2005; 16: 117-20.

62 

Arvin AM. Cell-mediated immunity to varicella-zoster virus. J Infect Dis 1992; 166 (Suppl 1): S35-41.

63 

Levin MJ, Smith JG, Kaufhold RM, et al. Decline in varicella-zoster virus (VZV)-specific cell-mediated immunity with increasing age and boosting with a high-dose VZV vaccine. J Infect Dis 2003; 188: 1336-44.

64 

Schneider L, Weinberg A, Boguniewicz M, et al. Immune response to varicella vaccine in children with atopic dermatitis compared to non-atopic controls. J Allergy Clin Immunol 2010; 126: 1306-7.e2.

65 

Foerster J, Molêda A. Feasibility analysis of interleukin-13 as a target for a therapeutic vaccine. Vaccines 2019; 7: pii: E20.

Copyright: © 2022 Termedia Sp. z o. o. This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.