ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Pathophysiology of plaque psoriasis

Pathophysiology of plaque psoriasis
Literature review current through: Jan 2024.
This topic last updated: Oct 18, 2021.

INTRODUCTION — Psoriasis is a complex immune-mediated inflammatory disease that occurs in genetically susceptible individuals and presents with the development of inflammatory plaques on the skin (picture 1A-B). Although early concepts of the pathogenesis of psoriasis focused primarily on keratinocyte hyperproliferation, dysregulation of the immune system is now recognized as a critical event in this disease. The evolving knowledge of the role of the immune system in psoriasis has had a significant impact on treatment development. Many new and emerging therapeutic agents target specific immunologic aspects of psoriatic disease. (See "Treatment of psoriasis in adults".)

The pathophysiology of plaque psoriasis will be discussed here. The epidemiology, genetics, clinical features, diagnosis, and management of psoriasis, as well as the pathogenesis of the less common subtype of pustular psoriasis, are reviewed separately. (See "Psoriasis: Epidemiology, clinical manifestations, and diagnosis" and "Treatment selection for moderate to severe plaque psoriasis in special populations" and "Pustular psoriasis: Pathogenesis, clinical manifestations, and diagnosis".)

OVERVIEW — Involvement of the immune system in psoriasis was first indicated in early studies that identified complex infiltrates of leukocytes involved in both innate and adaptive immunity in psoriatic skin [1,2]. Subsequent studies have supported the concept that interactions between dendritic cells, T cells, keratinocytes, neutrophils, and the cytokines released from immune cells likely contribute to the initiation and perpetuation of the cutaneous inflammation that is characteristic of psoriasis [3,4]. A basic sequence of the immunologic events that are theorized to occur in psoriasis is described below [3,4]:

Antigenic stimuli contribute to the activation of plasmacytoid dendritic cells and other innate immune cells in the skin.

Proinflammatory cytokines produced by innate immune cells, including interferon (IFN)-alpha, stimulate the activation and migration of various myeloid dendritic cells in the skin.

Activated macrophages and myeloid dendritic cells produce cytokines, in particular interleukin (IL) 23, which stimulate the attraction, differentiation, and activation of effector T cells.

Recruited effector T cells, in particular T helper type 17 (Th17) and Tc17 cells, produce cytokines, importantly IL-17A, which synergizes with other cytokines to stimulate keratinocytes to proliferate and produce antimicrobial peptides and proinflammatory cytokines.

Cytokines produced by immune cells and keratinocytes perpetuate the inflammatory process via participation in positive feedback loops.

The specific components of this pathway are reviewed below.

THE INNATE IMMUNE RESPONSE — The cellular components of the innate immune system that have been linked to the pathophysiology of psoriasis include the professional antigen-presenting cells (APCs), dendritic cells and macrophages, as well as neutrophils. The cytokines produced by these cells that appear to play major roles in the development of psoriasis include interferon (IFN)-alpha, tumor necrosis factor (TNF)-alpha, and interleukin (IL) 23. Modulating the production of these inflammatory cytokines and the innate immune cells responsible for their production with the small molecule phosphodiesterase 4 (PDE4) inhibitor apremilast and the small molecule Janus kinase (JAK) and tyrosine kinase 2 (Tyk2) inhibitors tofacitinib and deucravacitinib, respectively, has demonstrated benefit in the treatment of psoriasis [5-8]. (See "Treatment of psoriasis in adults", section on 'Emerging therapies' and "Treatment of psoriasis in adults", section on 'Apremilast'.)

Cellular components

Dendritic cells — Two types of dendritic cells (plasmacytoid dendritic cells and various myeloid dendritic cells) have been implicated as contributors to psoriasis:

Plasmacytoid dendritic cells – Plasmacytoid dendritic cells are a unique type of dendritic cell largely absent from normal human skin but are greatly increased in number in early lesions of psoriasis [9]. These cells are the primary producers of IFN-alpha, a key cytokine involved in the initiation phase of autoimmune responses and antiviral immunity. (See 'Interferon-alpha' below.)

Upregulation of IFN-alpha has been detected in early psoriatic lesions [10]. In addition, in a mouse xenograft model of psoriasis, the development of psoriatic lesions was dependent on IFN-alpha production by plasmacytoid dendritic cells [11].

Myeloid dendritic cells – The numbers of myeloid dendritic cells are markedly elevated in psoriatic skin. In particular, an inflammatory subset of myeloid dendritic cells (including TNF-alpha and iNOS-expressing dendritic cells [TIP-DCs], 6-sulfo LacNAc dendritic cells [slanDCs], and epidermal dendritic cells [eDCs]), recruited in response to the release of IFN-alpha and other proinflammatory cytokines and chemokines, has been detected in lesional, psoriatic skin [12-18]. Treatment with biologics, like secukinumab and guselkumab, decreases inflammatory dendritic cells within psoriatic skin [18].

Myeloid dendritic cells are potent APCs that produce an array of inflammatory cytokines that influence T cell activity, such as TNF-alpha (see 'Tumor necrosis factor-alpha' below). Myeloid dendritic cells also produce IL-23, a cytokine that causes the differentiation of precursor CD4+ cells into Th17 and Tc17 cells, and IL-12, a cytokine that stimulates the development of Th1 cells [16,17,19] (see 'Interleukin-23' below and 'Interleukin-12' below). Myeloid dendritic cells also affect keratinocytes and the skin vasculature through the production of IL-20 (a modulator of keratinocyte function) and nitric oxide (a vasodilating agent) [20]. (See 'Keratinocytes' below and 'Vascular changes' below.)

Macrophages and neutrophils — The importance of macrophages in psoriasis in humans has been evolving. Macrophages accumulate in psoriatic dermis near the basement membrane [21]. Inflammatory macrophages, and not myeloid dendritic cells, have been identified as the most abundant source of IL-23 in psoriatic skin [18]. Thus, these cells contribute to psoriasis through antigen presentation to T cells as well as cytokine production. The depletion of macrophages can reverse psoriasis-like skin changes in mouse models [22]. These cells are also reduced with biologic treatment of psoriasis [18].

Neutrophils are prominent in lesions of psoriasis, and are found in collections throughout the epidermis that are referred to as Munro microabscesses. The neutrophil chemoattractant IL-8 is highly elevated in psoriatic skin [23,24]. Like macrophages, neutrophils are believed to participate in psoriatic inflammation. Studies highlight their potential role as producers of IL-17A, an important cytokine with multiple effects in psoriatic plaques [25]. (See 'Interleukin-17A' below.)

Cytokines

Interferon-alpha — Type I IFN pathways are upregulated in early lesions of psoriasis [10]. IFN-alpha present in psoriatic skin is largely derived from plasmacytoid dendritic cells [10]. (See 'Dendritic cells' above.)

Evidence in support of a key role for IFN-alpha in psoriasis includes the observation that systemic treatment with IFN-alpha can exacerbate psoriasis [26,27]. In addition, topical treatment with imiquimod, which induces local production of IFN-alpha in the skin, has stimulated the development of psoriasis in humans and psoriasis-like disease in mice [28,29]. Furthermore, mice that lack a transcription factor that represses type I IFN signaling develop psoriasis-like disease [30].

Tumor necrosis factor-alpha — TNF-alpha is a critical proinflammatory cytokine common to many inflammatory disease states, including psoriasis [31]. Activated dendritic cells, Th17/Tc17 and Th1 cells, and keratinocytes in psoriatic skin produce TNF-alpha and respond to its effects. TNF-alpha also functions synergistically with other cytokines to promote disease pathogenesis.

The following observations support an important role for TNF-alpha in psoriasis:

Elevated levels of TNF-alpha are found in lesional skin of psoriatic patients [32].

Dramatic clinical improvement of psoriasis is seen with pharmacologic inhibitors of TNF-alpha (infliximab, adalimumab, etanercept, and certolizumab pegol) [33-36].

Removal of circulating TNF-alpha via the administration of etanercept (a TNF-alpha inhibitor) results in decreased numbers of dendritic cells and T cells along with a reduction of epidermal hyperplasia in psoriatic skin [14].

The central role of TNF-alpha in both innate and adaptive immune responses makes this cytokine a key target for therapeutic blockade. (See "Treatment of psoriasis in adults", section on 'Biologic agents'.)

Interleukin-23 — IL-23 is the regulatory cytokine responsible for proliferation and survival of Th17 and Tc17 cells, which are important T cell subsets in many autoimmune diseases, including psoriasis and Crohn disease [37-39]. IL-23 is produced by inflammatory macrophages, inflammatory myeloid dendritic cells, and (at low levels) by keratinocytes [40,41]. Production of this cytokine by dendritic cells can occur through toll-like receptor signaling pathways [42,43]. (See "Toll-like receptors: Roles in disease and therapy".)

The importance of IL-23 to the pathogenesis of psoriasis has been demonstrated by the following:

In psoriatics, IL-23 is elevated in lesions of psoriasis compared with unaffected skin and localizes to dermal macrophages, dermal dendritic cells, and (to a lesser extent) keratinocytes; levels of IL-23 fall with effective treatment of psoriasis [44-48].

Injection of IL-23 into normal mouse skin produces changes that are clinically and histologically similar to psoriasis; this process depends on downstream production of IL-22 and IL-17A [44,49,50]. Blockade of IL-23 in a mouse model prevented development of psoriasis in human xenografted skin from patients with psoriasis [51].

Polymorphisms in the genes encoding a component of the IL-23 receptor, IL23R, and the p40 and p19 subunits of IL-23 have been linked to psoriasis [52].

Several monoclonal antibodies bind selectively to the p19 subunit of IL-23 (guselkumab, tildrakizumab, risankizumab), thereby blocking IL-23-mediated effects on Th17 and Tc17 cells. These drugs are highly effective for psoriasis [53-55]. Additionally, the biologic agent ustekinumab, which inhibits both IL-23 and IL-12 signaling by binding the shared p40 subunit of these cytokines, is effective for psoriasis [56]. (See "Treatment of psoriasis in adults", section on 'Ustekinumab'.)

Deucravacitinib inhibits IL-23 receptor-mediated signaling by selective blockade of Tyk2, a tyrosine kinase acting downstream of the IL-23 receptor. Treatment with this drug has resulted in improvement of psoriasis in clinical trials [7,8].

Interleukin-12 — IL-12, like IL-23, is produced by activated myeloid dendritic cells, and this cytokine promotes the differentiation of Th1 cells. Indirect evidence for a contributory role for IL-12 in psoriasis comes from findings of increased Th1 cells and IFN-gamma (a product of Th1 cells) in psoriatic skin [57,58]. However, the degree to which IL-12 contributes to psoriasis is brought into question by the failure of a study to detect upregulation of the p35 subunit of IL-12 in psoriatic skin [46]. Additionally, preclinical mouse models suggest a regulatory or protective role for IL-12 in counteracting the IL-23/Th17 immunologic pathway [59]. It is possible that ustekinumab, an effective biologic agent for psoriasis that targets both IL-12 and IL-23 [51,56], may function primarily through the drug's effects on the IL-23 pathway. (See 'T helper type 1 cells' below and 'Interleukin-23' above and "Treatment of psoriasis in adults", section on 'Ustekinumab'.)

THE ADAPTIVE IMMUNE RESPONSE — The importance of T cells and their effector cytokines in psoriasis was established early on with the success of medications that inhibit global T cell responses, such as cyclosporine [60]. Therapies that block T cell activation or induce T cell death, such as alefacept (an inhibitor of the T cell-activating interaction between LFA3 on antigen-presenting cells (APCs) and CD2 on T cells that is no longer commercially available), have been effective for psoriasis [61]. In addition, injection of lymphocytes from psoriatics can induce psoriatic features in nonlesional human skin transplanted onto severe combined immunodeficient (SCID) mice [62]. Inhibition of inflammatory cytokine production via phosphodiesterase 4 (PDE4) blockade or Janus kinase (JAK)/tyrosine kinase 2 (Tyk2) inhibition in lymphocytes has improved psoriasis as well [6-8,63,64]. (See "Treatment of psoriasis in adults", section on 'Emerging therapies' and "Treatment of psoriasis in adults", section on 'Apremilast'.)

Cellular components

CD4+ T cells — CD4+ helper T cells are found throughout dermal inflammatory infiltrates in psoriatic skin [65]. Data in support of a role for these cells include the finding that the injection of CD4+ (but not CD8+) T cells from patients with psoriasis into graft sites on SCID mice transplanted with human skin induces psoriatic changes in the engrafted skin [66]. In addition, clinical improvement has occurred in patients with psoriasis treated with monoclonal antibodies against the CD4+ molecule on T cells [67].

The Th17 subset of CD4+ T cells, and to a lesser extent Th1 and Th22 cells, has been implicated in psoriasis. Early investigational studies presumed a dominating role for Th1 cells; however, Th17 cells are now known to play the more critical role. The effects of these cells and other components of the adaptive immune system in psoriasis are reviewed below.

T helper type 17 cells — The discovery of T helper type 17 (Th17) cells has led to important insights on the pathophysiology of psoriasis and new target-specific approaches to treatment. Th17 cells develop in psoriatic skin under the polarizing effects of interleukin (IL) 1, IL-6, transforming growth factor (TGF)-beta, and IL-23 produced by inflammatory dendritic cells and macrophages [68] (see 'Dendritic cells' above and 'Macrophages and neutrophils' above). The activation of Th17 cells by IL-23 stimulates these cells to produce IL-17A and IL-22, cytokines that promote keratinocyte activation and growth [49,69,70]. (See 'Interleukin-17A' below and 'Interleukin-22' below.)

The following study results highlight the role of these cells in psoriasis:

Th17 cells produce an array of proinflammatory cytokines, including IL-17A, IL-17F, IL-21, IL-22, IL-6, and TNF-alpha [39], all of which have been linked to psoriasis.

Th17 cells are found in lesional skin and at elevated levels in the circulation in patients with psoriasis, and IL-17A is abundantly expressed by these cells [57,71,72].

Transgenic mice overexpressing the p19 subunit of IL-23, a key factor for Th17 cell functioning, have severe widespread inflammatory disease that includes inflammation of the skin [73].

Effective therapies for psoriasis, such as phototherapy, cyclosporine, etanercept, and infliximab, have all been shown to modulate the IL-23/T17 pathway in psoriasis [14,72,74-76].

Selective targeting of IL-17A (the main effector cytokine produced by Th17 cells) by secukinumab and ixekizumab, as well as duel blockade of IL-17A and IL-17F by bimekizumab, leads to dramatic improvement in psoriasis [77-79].

Similar success in clearing psoriatic skin is seen with blockade of the IL-17RA, a subunit of the IL-17A receptor, by brodalumab [80]. (See "Treatment of psoriasis in adults", section on 'Secukinumab' and "Treatment of psoriasis in adults", section on 'Emerging therapies'.)

Affecting Th17 cells via inhibition of IL-23 is also a proven therapeutic approach for psoriasis [53,81,82]. Ustekinumab, a highly effective drug that is in use for psoriasis, is a monoclonal antibody that binds to p40, a subunit of both IL-23 and IL-12, which leads to death of Th17 cells. Similarly, guselkumab, tildrakizumab, and risankizumab are selective blockers of IL-23 that show high efficacy in clinical practice and clinical trials. Agents that interrupt JAK/STAT signaling pathways (key intracellular events in Th17 cytokine production) are also under investigation for the treatment of psoriasis [6,7,64].

T helper type 1 cells — T helper type 1 (Th1) cells produce an array of proinflammatory cytokines, including interferon (IFN)-gamma, IL-2, and TNF-alpha [83]. IFN-gamma, the prototypic cytokine produced by these cells, can promote psoriasis-like changes in nonlesional, psoriatic skin [84]. Of note, infliximab, an effective anti-TNF therapy for psoriasis, is also capable of inhibiting IFN-gamma production by Th1 cells [85].

IL-12, a cytokine produced by activated myeloid dendritic cells, promotes the differentiation of Th1 cells. The degree to which IL-12-mediated effects contribute to psoriasis is uncertain [46]. (See 'Dendritic cells' above and 'Interleukin-12' above.)

CD8+ T cells — Cytotoxic CD8+ T cells, especially Tc17 cells, are primarily found in the epidermis of psoriatic skin and are also considered to play a prominent role in psoriasis [86]. Although they produce cytolytic enzymes, their role in psoriasis is speculated to involve the elaboration of inflammatory cytokines, including IL-17A [87-90]. Interestingly, clonal restriction of resident memory epidermal T cells seems limited to the CD8+ compartment [91]. This finding might suggest a link between viral or self-antigens in the epidermis and triggering of psoriasis, since these types of MHC class I-restricted antigens are typically presented to CD8+ T cells, and not CD4+ T cells [92,93].  

In addition, resident memory T cells that are long-lived CD69+, CD103+, IL-17A-producing epidermal CD8+ T cells have been linked to the recurrence of psoriasis in previously healed skin [94]. Further research on these cells may result in therapeutic strategies to target them, perhaps leading to greater long-term control of psoriasis in the future.

Regulatory T cells — CD18-knockout mice that are deficient in regulatory T cells develop skin with features of psoriasis [95]. T regulatory cells have also been found to limit psoriasiform inflammation in imiquimod-induced mouse skin via modulation of IFN-alpha [96]. Additionally, defects in the suppressive function of regulatory T cells have been found in psoriasis lesions [97]. In the absence of properly functioning regulatory cells, downregulation of immune responses is inadequate. This may contribute to unchecked inflammation in psoriasis.

Cytokines

Interleukin-17A — IL-17A, an effector cytokine produced by Th17 and Tc17 cells, is elevated in lesions of psoriasis and serum of patients with psoriasis [44,98]. IL-17A has many functions that are relevant to psoriasis, including the activation, recruitment, and inhibition of apoptosis in neutrophils; the enhancement of angiogenesis; the promotion of the release of other inflammatory cytokines (TNF-alpha, IL-1, and IL-6); and the direct activation of keratinocytes leading to increased production of chemokines [49,70,99-105]. Therapeutic agents for psoriasis that block IL-17A production or the downstream effects of IL-17 are commercially available. The anti-IL-17A monoclonal antibodies secukinumab and ixekizumab have shown efficacy results equivalent to or better than other available biologic agents for moderate to severe plaque psoriasis [77,78]. Bimekizumab, a monoclonal antibody that blocks IL-17A and its close homolog IL-17F, shows promising results in clinical trials [106]. Similarly, high efficacy results in psoriasis are achieved with brodalumab, an antibody that blocks the binding of IL-17A to its receptor [80].

Interleukin-22 — IL-22 levels are increased in the blood of patients with psoriasis and in psoriatic plaques. Treatment of psoriasis decreases these levels [72,98,107].

IL-22 is produced by Th17 cells, Tc17 cells, and Th22 cells. This cytokine stimulates the growth and activation of keratinocytes and has little effect on immune cells. In keratinocytes, IL-22-mediated signaling via STAT3 stimulates cell hyperproliferation, secretion of antimicrobial peptides, and production of matrix metalloproteinases that support increased cell mobility [50,107-110]. Of note, trials utilizing fezakinumab, an anti-IL-22 monoclonal antibody, failed in psoriasis, indicating this cytokine is not critical for maintenance of psoriatic lesions [111].

KERATINOCYTES — A role for epidermal keratinocytes as triggers for the initiation of psoriasis is a subject of much debate. Keratinocyte-derived antimicrobial peptides (AMPs), including beta-defensins, cathelicidins, and psoriasin (S100A7), may be induced by trauma, and are upregulated in psoriatic epidermis early in the course of lesion development [112]. AMPs have both chemoattractant and immunomodulatory effects on dendritic cells and T cells and may contribute to cutaneous inflammation. Similarly, the melanocyte-derived ADAMTS-like protein 5 may also function as an antigenic trigger of the interleukin (IL) 17 pathway in psoriatics [113]. One theory on the initiation of psoriasis involves the upregulation of the AMP cathelicidin LL-37 in skin. LL-37 may bind self-DNA and stimulate the production of interferon (IFN)-alpha by plasmacytoid dendritic cells through Toll-like receptor (TLR)-9 [114]. LL-37 has also been shown to be antigenic in psoriasis; LL-37 stimulates both CD4+ and CD8+ T cells in an HLA-restricted manner [115].

Keratinocyte hyperplasia characteristic of psoriasis may develop because of the effects of cytokines produced by immune cells. IL-22 is produced by Th17 and Tc17 cells and promotes many of the epidermal changes that occur in psoriatic lesions (see 'Interleukin-22' above). Additionally, IL-20 production by keratinocytes plays an important synergistic role downstream of IL-22.

Both IL-22 and IL-20 are highly expressed in psoriatic skin and promote alterations in epidermal thickness, maturation defects, and upregulation of AMPs [116,117]. A role for IL-20 in psoriasis is supported by the observation that blocking IL-20 in a SCID xenotransplant model of psoriasis induces resolution of psoriasis and blocks initiation of disease [118]. Therapeutics directed against both IL-22 and IL-20 have been tested in early clinical trials [111].

The cytokines and chemokines produced by activated keratinocytes contribute to the sustainment of the inflammatory response in psoriasis via their effects on innate and adaptive immune cells. Topical vitamin D analogs as well as topical and oral retinoids may affect this pathway in psoriasis. Methotrexate was originally thought to modulate the epidermal component of psoriasis as well, although current thinking suggests that its direct effects on the immune system are more important. (See "Treatment of psoriasis in adults", section on 'Topical vitamin D analogs' and "Treatment of psoriasis in adults", section on 'Tazarotene' and "Treatment of psoriasis in adults", section on 'Methotrexate'.)

VASCULAR CHANGES — Endothelial cells within psoriatic plaques express elevated levels of vascular endothelial growth factor (VEGF), prostaglandins, and nitric oxide, all contributing to the characteristic leaky and tortuous vessels that are abundant in psoriatic skin [119,120]. Transgenic mice overexpressing VEGF in the epidermis have been found to develop psoriasiform skin changes [121].

The activated vasculature promotes attraction and transmigration of the leukocytes discussed above. Antiangiogenic agents have demonstrated some success in psoriasis patients, but more investigation is needed [122].

ENVIRONMENTAL FACTORS — Medications, trauma, alcohol, cigarette smoking, stress, and infections have all been linked to the onset of autoimmune inflammatory conditions and are also known to trigger psoriasis. The current understanding of this process is rudimentary.

A proposed pathogenic model highlights the possibility of innate recognition of conserved sequences in microbes through TLR signaling as one possible explanation for infectious triggers. The TLR-7/8 antagonist imiquimod can induce a psoriasis-like skin disease in mice through production of interferon (IFN)-alpha by plasmacytoid dendritic cells. This process is dependent on the interleukin (IL) 23/T17 pathway [29].

SUMMARY

Psoriasis is a complex immune-mediated disorder that presents as inflammatory plaques in the skin (picture 1A-B). Dysregulation or alteration of components of the innate and adaptive immune systems, keratinocyte function, and vascular structure contribute to the manifestations of this disease. (See 'Overview' above.)

Plasmacytoid and myeloid dendritic cells are key contributors to development of inflammation in psoriasis. Interferon (IFN)-alpha produced by plasmacytoid dendritic cells stimulates the activation of myeloid dendritic cells, which contribute to the adaptive immune response through the activation of T cells. (See 'The innate immune response' above.)

Interleukin (IL) 23 and IL-12 produced by activated macrophages and myeloid dendritic cells promote the development of T helper type 17 (Th17)/Tc17 and T helper type 1 (Th1) cells, respectively. Th17 and Tc17 cells play a major role in the pathogenesis of psoriasis. (See 'The adaptive immune response' above.)

Keratinocytes may contribute to the initiation of psoriasis via the production of antimicrobial peptides. Epidermal hyperplasia is driven by cytokines that stimulate keratinocyte activation and proliferation. (See 'Keratinocytes' above.)

Knowledge of the pathophysiology of psoriasis is evolving. Advances in the understanding of the mechanisms of disease will likely contribute to the development of new therapeutic agents and improved patient outcomes. (See 'Introduction' above and "Treatment of psoriasis in adults".)

  1. Bos JD, Hulsebosch HJ, Krieg SR, et al. Immunocompetent cells in psoriasis. In situ immunophenotyping by monoclonal antibodies. Arch Dermatol Res 1983; 275:181.
  2. Braun-Falco O, Burg G. [Inflammatory infiltrate in psoriasis vulgaris. A cytochemical study]. Arch Klin Exp Dermatol 1970; 236:297.
  3. Hawkes JE, Yan BY, Chan TC, Krueger JG. Discovery of the IL-23/IL-17 Signaling Pathway and the Treatment of Psoriasis. J Immunol 2018; 201:1605.
  4. Yan D, Gudjonsson JE, Le S, et al. New Frontiers in Psoriatic Disease Research, Part I: Genetics, Environmental Triggers, Immunology, Pathophysiology, and Precision Medicine. J Invest Dermatol 2021; 141:2112.
  5. Schafer PH, Parton A, Capone L, et al. Apremilast is a selective PDE4 inhibitor with regulatory effects on innate immunity. Cell Signal 2014; 26:2016.
  6. Papp KA, Menter A, Strober B, et al. Efficacy and safety of tofacitinib, an oral Janus kinase inhibitor, in the treatment of psoriasis: a Phase 2b randomized placebo-controlled dose-ranging study. Br J Dermatol 2012; 167:668.
  7. Papp K, Gordon K, Thaçi D, et al. Phase 2 Trial of Selective Tyrosine Kinase 2 Inhibition in Psoriasis. N Engl J Med 2018; 379:1313.
  8. Krueger JG, McInnes IB, Blauvelt A. Tyrosine kinase 2 and Janus kinase‒signal transducer and activator of transcription signaling and inhibition in plaque psoriasis. J Am Acad Dermatol 2022; 86:148.
  9. Wollenberg A, Wagner M, Günther S, et al. Plasmacytoid dendritic cells: a new cutaneous dendritic cell subset with distinct role in inflammatory skin diseases. J Invest Dermatol 2002; 119:1096.
  10. van der Fits L, van der Wel LI, Laman JD, et al. In psoriasis lesional skin the type I interferon signaling pathway is activated, whereas interferon-alpha sensitivity is unaltered. J Invest Dermatol 2004; 122:51.
  11. Nestle FO, Conrad C, Tun-Kyi A, et al. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J Exp Med 2005; 202:135.
  12. Farkas A, Kemény L. Interferon-α in the generation of monocyte-derived dendritic cells: recent advances and implications for dermatology. Br J Dermatol 2011; 165:247.
  13. Lowes MA, Chamian F, Abello MV, et al. Increase in TNF-alpha and inducible nitric oxide synthase-expressing dendritic cells in psoriasis and reduction with efalizumab (anti-CD11a). Proc Natl Acad Sci U S A 2005; 102:19057.
  14. Zaba LC, Cardinale I, Gilleaudeau P, et al. Amelioration of epidermal hyperplasia by TNF inhibition is associated with reduced Th17 responses. J Exp Med 2007; 204:3183.
  15. Zaba LC, Krueger JG, Lowes MA. Resident and "inflammatory" dendritic cells in human skin. J Invest Dermatol 2009; 129:302.
  16. Hänsel A, Günther C, Ingwersen J, et al. Human slan (6-sulfo LacNAc) dendritic cells are inflammatory dermal dendritic cells in psoriasis and drive strong TH17/TH1 T-cell responses. J Allergy Clin Immunol 2011; 127:787.
  17. Martini E, Wikén M, Cheuk S, et al. Dynamic Changes in Resident and Infiltrating Epidermal Dendritic Cells in Active and Resolved Psoriasis. J Invest Dermatol 2017; 137:865.
  18. Mehta H, Mashiko S, Angsana J, et al. Differential Changes in Inflammatory Mononuclear Phagocyte and T-Cell Profiles within Psoriatic Skin during Treatment with Guselkumab vs. Secukinumab. J Invest Dermatol 2021; 141:1707.
  19. McKenzie BS, Kastelein RA, Cua DJ. Understanding the IL-23-IL-17 immune pathway. Trends Immunol 2006; 27:17.
  20. Wang F, Lee E, Lowes MA, et al. Prominent production of IL-20 by CD68+/CD11c+ myeloid-derived cells in psoriasis: Gene regulation and cellular effects. J Invest Dermatol 2006; 126:1590.
  21. Boehncke WH, Wortmann S, Kaufmann R, et al. A subset of macrophages located along the basement membrane ("lining cells") is a characteristic histopathological feature of psoriasis. Am J Dermatopathol 1995; 17:139.
  22. Wang H, Peters T, Kess D, et al. Activated macrophages are essential in a murine model for T cell-mediated chronic psoriasiform skin inflammation. J Clin Invest 2006; 116:2105.
  23. Glowacka E, Lewkowicz P, Rotsztejn H, Zalewska A. IL-8, IL-12 and IL-10 cytokines generation by neutrophils, fibroblasts and neutrophils- fibroblasts interaction in psoriasis. Adv Med Sci 2010; 55:254.
  24. van de Kerkhof PC, Lammers AM. Intraepidermal accumulation of polymorphonuclear leukocytes in chronic stable plaque psoriasis. Dermatologica 1987; 174:224.
  25. Reich K, Papp KA, Matheson RT, et al. Evidence that a neutrophil-keratinocyte crosstalk is an early target of IL-17A inhibition in psoriasis. Exp Dermatol 2015; 24:529.
  26. Funk J, Langeland T, Schrumpf E, Hanssen LE. Psoriasis induced by interferon-alpha. Br J Dermatol 1991; 125:463.
  27. Ketikoglou I, Karatapanis S, Elefsiniotis I, et al. Extensive psoriasis induced by pegylated interferon alpha-2b treatment for chronic hepatitis B. Eur J Dermatol 2005; 15:107.
  28. Patel U, Mark NM, Machler BC, Levine VJ. Imiquimod 5% cream induced psoriasis: a case report, summary of the literature and mechanism. Br J Dermatol 2011; 164:670.
  29. van der Fits L, Mourits S, Voerman JS, et al. Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J Immunol 2009; 182:5836.
  30. Hida S, Ogasawara K, Sato K, et al. CD8(+) T cell-mediated skin disease in mice lacking IRF-2, the transcriptional attenuator of interferon-alpha/beta signaling. Immunity 2000; 13:643.
  31. Tracey D, Klareskog L, Sasso EH, et al. Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol Ther 2008; 117:244.
  32. Uyemura K, Yamamura M, Fivenson DF, et al. The cytokine network in lesional and lesion-free psoriatic skin is characterized by a T-helper type 1 cell-mediated response. J Invest Dermatol 1993; 101:701.
  33. Menter A, Tyring SK, Gordon K, et al. Adalimumab therapy for moderate to severe psoriasis: A randomized, controlled phase III trial. J Am Acad Dermatol 2008; 58:106.
  34. Papp KA, Tyring S, Lahfa M, et al. A global phase III randomized controlled trial of etanercept in psoriasis: safety, efficacy, and effect of dose reduction. Br J Dermatol 2005; 152:1304.
  35. Reich K, Nestle FO, Papp K, et al. Infliximab induction and maintenance therapy for moderate-to-severe psoriasis: a phase III, multicentre, double-blind trial. Lancet 2005; 366:1367.
  36. Blauvelt A, Reich K, Lebwohl M, et al. Certolizumab pegol for the treatment of patients with moderate-to-severe chronic plaque psoriasis: pooled analysis of week 16 data from three randomized controlled trials. J Eur Acad Dermatol Venereol 2019; 33:546.
  37. Stockinger B, Veldhoen M. Differentiation and function of Th17 T cells. Curr Opin Immunol 2007; 19:281.
  38. Blauvelt A. T-helper 17 cells in psoriatic plaques and additional genetic links between IL-23 and psoriasis. J Invest Dermatol 2008; 128:1064.
  39. Tesmer LA, Lundy SK, Sarkar S, Fox DA. Th17 cells in human disease. Immunol Rev 2008; 223:87.
  40. Gerosa F, Baldani-Guerra B, Lyakh LA, et al. Differential regulation of interleukin 12 and interleukin 23 production in human dendritic cells. J Exp Med 2008; 205:1447.
  41. Piskin G, Sylva-Steenland RM, Bos JD, Teunissen MB. In vitro and in situ expression of IL-23 by keratinocytes in healthy skin and psoriasis lesions: enhanced expression in psoriatic skin. J Immunol 2006; 176:1908.
  42. Higgins SC, Jarnicki AG, Lavelle EC, Mills KH. TLR4 mediates vaccine-induced protective cellular immunity to Bordetella pertussis: role of IL-17-producing T cells. J Immunol 2006; 177:7980.
  43. Re F, Strominger JL. Toll-like receptor 2 (TLR2) and TLR4 differentially activate human dendritic cells. J Biol Chem 2001; 276:37692.
  44. Chan JR, Blumenschein W, Murphy E, et al. IL-23 stimulates epidermal hyperplasia via TNF and IL-20R2-dependent mechanisms with implications for psoriasis pathogenesis. J Exp Med 2006; 203:2577.
  45. Chamian F, Lowes MA, Lin SL, et al. Alefacept reduces infiltrating T cells, activated dendritic cells, and inflammatory genes in psoriasis vulgaris. Proc Natl Acad Sci U S A 2005; 102:2075.
  46. Lee E, Trepicchio WL, Oestreicher JL, et al. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. J Exp Med 2004; 199:125.
  47. Piskin G, Tursen U, Sylva-Steenland RM, et al. Clinical improvement in chronic plaque-type psoriasis lesions after narrow-band UVB therapy is accompanied by a decrease in the expression of IFN-gamma inducers -- IL-12, IL-18 and IL-23. Exp Dermatol 2004; 13:764.
  48. Toichi E, Torres G, McCormick TS, et al. An anti-IL-12p40 antibody down-regulates type 1 cytokines, chemokines, and IL-12/IL-23 in psoriasis. J Immunol 2006; 177:4917.
  49. Rizzo HL, Kagami S, Phillips KG, et al. IL-23-mediated psoriasis-like epidermal hyperplasia is dependent on IL-17A. J Immunol 2011; 186:1495.
  50. Zheng Y, Danilenko DM, Valdez P, et al. Interleukin-22, a T(H)17 cytokine, mediates IL-23-induced dermal inflammation and acanthosis. Nature 2007; 445:648.
  51. Tonel G, Conrad C, Laggner U, et al. Cutting edge: A critical functional role for IL-23 in psoriasis. J Immunol 2010; 185:5688.
  52. Liu Y, Helms C, Liao W, et al. A genome-wide association study of psoriasis and psoriatic arthritis identifies new disease loci. PLoS Genet 2008; 4:e1000041.
  53. Papp KA, Blauvelt A, Bukhalo M, et al. Risankizumab versus Ustekinumab for Moderate-to-Severe Plaque Psoriasis. N Engl J Med 2017; 376:1551.
  54. Blauvelt A, Papp KA, Griffiths CE, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the continuous treatment of patients with moderate to severe psoriasis: Results from the phase III, double-blinded, placebo- and active comparator-controlled VOYAGE 1 trial. J Am Acad Dermatol 2017; 76:405.
  55. Papp K, Thaçi D, Reich K, et al. Tildrakizumab (MK-3222), an anti-interleukin-23p19 monoclonal antibody, improves psoriasis in a phase IIb randomized placebo-controlled trial. Br J Dermatol 2015; 173:930.
  56. Krueger GG, Langley RG, Leonardi C, et al. A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N Engl J Med 2007; 356:580.
  57. Lowes MA, Kikuchi T, Fuentes-Duculan J, et al. Psoriasis vulgaris lesions contain discrete populations of Th1 and Th17 T cells. J Invest Dermatol 2008; 128:1207.
  58. Szabo SK, Hammerberg C, Yoshida Y, et al. Identification and quantitation of interferon-gamma producing T cells in psoriatic lesions: localization to both CD4+ and CD8+ subsets. J Invest Dermatol 1998; 111:1072.
  59. Kulig P, Musiol S, Freiberger SN, et al. IL-12 protects from psoriasiform skin inflammation. Nat Commun 2016; 7:13466.
  60. Ellis CN, Gorsulowsky DC, Hamilton TA, et al. Cyclosporine improves psoriasis in a double-blind study. JAMA 1986; 256:3110.
  61. Krueger GG, Papp KA, Stough DB, et al. A randomized, double-blind, placebo-controlled phase III study evaluating efficacy and tolerability of 2 courses of alefacept in patients with chronic plaque psoriasis. J Am Acad Dermatol 2002; 47:821.
  62. Wrone-Smith T, Nickoloff BJ. Dermal injection of immunocytes induces psoriasis. J Clin Invest 1996; 98:1878.
  63. Papp KA, Kaufmann R, Thaçi D, et al. Efficacy and safety of apremilast in subjects with moderate to severe plaque psoriasis: results from a phase II, multicenter, randomized, double-blind, placebo-controlled, parallel-group, dose-comparison study. J Eur Acad Dermatol Venereol 2013; 27:e376.
  64. Papp KA, Menter MA, Raman M, et al. A randomized phase 2b trial of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor, in patients with moderate-to-severe psoriasis. Br J Dermatol 2016; 174:1266.
  65. Nikaein A, Phillips C, Gilbert SC, et al. Characterization of skin-infiltrating lymphocytes in patients with psoriasis. J Invest Dermatol 1991; 96:3.
  66. Nickoloff BJ, Wrone-Smith T. Injection of pre-psoriatic skin with CD4+ T cells induces psoriasis. Am J Pathol 1999; 155:145.
  67. Gottlieb AB, Lebwohl M, Shirin S, et al. Anti-CD4 monoclonal antibody treatment of moderate to severe psoriasis vulgaris: results of a pilot, multicenter, multiple-dose, placebo-controlled study. J Am Acad Dermatol 2000; 43:595.
  68. Di Cesare A, Di Meglio P, Nestle FO. The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol 2009; 129:1339.
  69. Chiricozzi A, Guttman-Yassky E, Suárez-Fariñas M, et al. Integrative responses to IL-17 and TNF-α in human keratinocytes account for key inflammatory pathogenic circuits in psoriasis. J Invest Dermatol 2011; 131:677.
  70. Harper EG, Guo C, Rizzo H, et al. Th17 cytokines stimulate CCL20 expression in keratinocytes in vitro and in vivo: implications for psoriasis pathogenesis. J Invest Dermatol 2009; 129:2175.
  71. Teunissen MB, Koomen CW, de Waal Malefyt R, et al. Interleukin-17 and interferon-gamma synergize in the enhancement of proinflammatory cytokine production by human keratinocytes. J Invest Dermatol 1998; 111:645.
  72. Kagami S, Rizzo HL, Lee JJ, et al. Circulating Th17, Th22, and Th1 cells are increased in psoriasis. J Invest Dermatol 2010; 130:1373.
  73. Wiekowski MT, Leach MW, Evans EW, et al. Ubiquitous transgenic expression of the IL-23 subunit p19 induces multiorgan inflammation, runting, infertility, and premature death. J Immunol 2001; 166:7563.
  74. Haider AS, Lowes MA, Suárez-Fariñas M, et al. Identification of cellular pathways of "type 1," Th17 T cells, and TNF- and inducible nitric oxide synthase-producing dendritic cells in autoimmune inflammation through pharmacogenomic study of cyclosporine A in psoriasis. J Immunol 2008; 180:1913.
  75. Rácz E, Prens EP, Kurek D, et al. Effective treatment of psoriasis with narrow-band UVB phototherapy is linked to suppression of the IFN and Th17 pathways. J Invest Dermatol 2011; 131:1547.
  76. Zaba LC, Suárez-Fariñas M, Fuentes-Duculan J, et al. Effective treatment of psoriasis with etanercept is linked to suppression of IL-17 signaling, not immediate response TNF genes. J Allergy Clin Immunol 2009; 124:1022.
  77. Langley RG, Elewski BE, Lebwohl M, et al. Secukinumab in plaque psoriasis--results of two phase 3 trials. N Engl J Med 2014; 371:326.
  78. Gordon KB, Blauvelt A, Papp KA, et al. Phase 3 Trials of Ixekizumab in Moderate-to-Severe Plaque Psoriasis. N Engl J Med 2016; 375:345.
  79. Reich K, Papp KA, Blauvelt A, et al. Bimekizumab versus ustekinumab for the treatment of moderate to severe plaque psoriasis (BE VIVID): efficacy and safety from a 52-week, multicentre, double-blind, active comparator and placebo controlled phase 3 trial. Lancet 2021; 397:487.
  80. Lebwohl M, Strober B, Menter A, et al. Phase 3 Studies Comparing Brodalumab with Ustekinumab in Psoriasis. N Engl J Med 2015; 373:1318.
  81. Kurzeja M, Rudnicka L, Olszewska M. New interleukin-23 pathway inhibitors in dermatology: ustekinumab, briakinumab, and secukinumab. Am J Clin Dermatol 2011; 12:113.
  82. Alunno A, Carubbi F, Cafaro G, et al. Targeting the IL-23/IL-17 axis for the treatment of psoriasis and psoriatic arthritis. Expert Opin Biol Ther 2015; 15:1727.
  83. Murphy KM, Reiner SL. The lineage decisions of helper T cells. Nat Rev Immunol 2002; 2:933.
  84. Wei L, Debets R, Hegmans JJ, et al. IL-1 beta and IFN-gamma induce the regenerative epidermal phenotype of psoriasis in the transwell skin organ culture system. IFN-gamma up-regulates the expression of keratin 17 and keratinocyte transglutaminase via endogenous IL-1 production. J Pathol 1999; 187:358.
  85. Haider AS, Cohen J, Fei J, et al. Insights into gene modulation by therapeutic TNF and IFNgamma antibodies: TNF regulates IFNgamma production by T cells and TNF-regulated genes linked to psoriasis transcriptome. J Invest Dermatol 2008; 128:655.
  86. Lowes MA, Bowcock AM, Krueger JG. Pathogenesis and therapy of psoriasis. Nature 2007; 445:866.
  87. Conrad C, Boyman O, Tonel G, et al. Alpha1beta1 integrin is crucial for accumulation of epidermal T cells and the development of psoriasis. Nat Med 2007; 13:836.
  88. Ortega C, Fernández-A S, Carrillo JM, et al. IL-17-producing CD8+ T lymphocytes from psoriasis skin plaques are cytotoxic effector cells that secrete Th17-related cytokines. J Leukoc Biol 2009; 86:435.
  89. Gaspari AA. Innate and adaptive immunity and the pathophysiology of psoriasis. J Am Acad Dermatol 2006; 54:S67.
  90. Liang Y, Pan HF, Ye DQ. IL-17A-producing CD8(+)T cells as therapeutic targets in autoimmunity. Expert Opin Ther Targets 2015; 19:651.
  91. Cheuk S, Wikén M, Blomqvist L, et al. Epidermal Th22 and Tc17 cells form a localized disease memory in clinically healed psoriasis. J Immunol 2014; 192:3111.
  92. Lin WJ, Norris DA, Achziger M, et al. Oligoclonal expansion of intraepidermal T cells in psoriasis skin lesions. J Invest Dermatol 2001; 117:1546.
  93. Prinz JC, Vollmer S, Boehncke WH, et al. Selection of conserved TCR VDJ rearrangements in chronic psoriatic plaques indicates a common antigen in psoriasis vulgaris. Eur J Immunol 1999; 29:3360.
  94. Gallais Sérézal I, Classon C, Cheuk S, et al. Resident T Cells in Resolved Psoriasis Steer Tissue Responses that Stratify Clinical Outcome. J Invest Dermatol 2018; 138:1754.
  95. Wang H, Peters T, Sindrilaru A, et al. TGF-beta-dependent suppressive function of Tregs requires wild-type levels of CD18 in a mouse model of psoriasis. J Clin Invest 2008; 118:2629.
  96. Stockenhuber K, Hegazy AN, West NR, et al. Foxp3+ T reg cells control psoriasiform inflammation by restraining an IFN-I-driven CD8+ T cell response. J Exp Med 2018; 215:1987.
  97. Sugiyama H, Gyulai R, Toichi E, et al. Dysfunctional blood and target tissue CD4+CD25high regulatory T cells in psoriasis: mechanism underlying unrestrained pathogenic effector T cell proliferation. J Immunol 2005; 174:164.
  98. Wilson NJ, Boniface K, Chan JR, et al. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol 2007; 8:950.
  99. Albanesi C, Scarponi C, Cavani A, et al. Interleukin-17 is produced by both Th1 and Th2 lymphocytes, and modulates interferon-gamma- and interleukin-4-induced activation of human keratinocytes. J Invest Dermatol 2000; 115:81.
  100. Dragon S, Saffar AS, Shan L, Gounni AS. IL-17 attenuates the anti-apoptotic effects of GM-CSF in human neutrophils. Mol Immunol 2008; 45:160.
  101. Homey B, Dieu-Nosjean MC, Wiesenborn A, et al. Up-regulation of macrophage inflammatory protein-3 alpha/CCL20 and CC chemokine receptor 6 in psoriasis. J Immunol 2000; 164:6621.
  102. Laan M, Cui ZH, Hoshino H, et al. Neutrophil recruitment by human IL-17 via C-X-C chemokine release in the airways. J Immunol 1999; 162:2347.
  103. Numasaki M, Fukushi J, Ono M, et al. Interleukin-17 promotes angiogenesis and tumor growth. Blood 2003; 101:2620.
  104. Ruddy MJ, Wong GC, Liu XK, et al. Functional cooperation between interleukin-17 and tumor necrosis factor-alpha is mediated by CCAAT/enhancer-binding protein family members. J Biol Chem 2004; 279:2559.
  105. Starnes T, Broxmeyer HE, Robertson MJ, Hromas R. Cutting edge: IL-17D, a novel member of the IL-17 family, stimulates cytokine production and inhibits hemopoiesis. J Immunol 2002; 169:642.
  106. Papp KA, Merola JF, Gottlieb AB, et al. Dual neutralization of both interleukin 17A and interleukin 17F with bimekizumab in patients with psoriasis: Results from BE ABLE 1, a 12-week randomized, double-blinded, placebo-controlled phase 2b trial. J Am Acad Dermatol 2018; 79:277.
  107. Wolk K, Witte E, Wallace E, et al. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential role in psoriasis. Eur J Immunol 2006; 36:1309.
  108. Boniface K, Bernard FX, Garcia M, et al. IL-22 inhibits epidermal differentiation and induces proinflammatory gene expression and migration of human keratinocytes. J Immunol 2005; 174:3695.
  109. Liang SC, Tan XY, Luxenberg DP, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med 2006; 203:2271.
  110. Wolk K, Kunz S, Witte E, et al. IL-22 increases the innate immunity of tissues. Immunity 2004; 21:241.
  111. Nograles KE, Krueger JG. Anti-cytokine therapies for psoriasis. Exp Cell Res 2011; 317:1293.
  112. Hollox EJ, Huffmeier U, Zeeuwen PL, et al. Psoriasis is associated with increased beta-defensin genomic copy number. Nat Genet 2008; 40:23.
  113. Arakawa A, Siewert K, Stöhr J, et al. Melanocyte antigen triggers autoimmunity in human psoriasis. J Exp Med 2015; 212:2203.
  114. Lande R, Gregorio J, Facchinetti V, et al. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature 2007; 449:564.
  115. Lande R, Botti E, Jandus C, et al. The antimicrobial peptide LL37 is a T-cell autoantigen in psoriasis. Nat Commun 2014; 5:5621.
  116. Blumberg H, Conklin D, Xu WF, et al. Interleukin 20: discovery, receptor identification, and role in epidermal function. Cell 2001; 104:9.
  117. Wolk K, Witte E, Warszawska K, et al. The Th17 cytokine IL-22 induces IL-20 production in keratinocytes: a novel immunological cascade with potential relevance in psoriasis. Eur J Immunol 2009; 39:3570.
  118. Stenderup K, Rosada C, Worsaae A, et al. Interleukin-20 plays a critical role in maintenance and development of psoriasis in the human xenograft transplantation model. Br J Dermatol 2009; 160:284.
  119. Costa C, Incio J, Soares R. Angiogenesis and chronic inflammation: cause or consequence? Angiogenesis 2007; 10:149.
  120. Detmar M, Brown LF, Claffey KP, et al. Overexpression of vascular permeability factor/vascular endothelial growth factor and its receptors in psoriasis. J Exp Med 1994; 180:1141.
  121. Xia YP, Li B, Hylton D, et al. Transgenic delivery of VEGF to mouse skin leads to an inflammatory condition resembling human psoriasis. Blood 2003; 102:161.
  122. Ehrlich A, Booher S, Becerra Y, et al. Micellar paclitaxel improves severe psoriasis in a prospective phase II pilot study. J Am Acad Dermatol 2004; 50:533.
Topic 15277 Version 9.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟