• Open Access
    Systematic Review

    Signaling pathways and targets of natural products in psoriasis treatment

    Ly Thi Huong Nguyen *

    Explor Med. 2022;3:345–367 DOI: https://doi.org/10.37349/emed.2022.00098

    Received: January 28, 2022 Accepted: June 06, 2022 Published: August 29, 2022

    Academic Editor: Xiaohui Fan, Zhejiang University, China

    Abstract

    Aim:

    Psoriasis is a common chronic inflammatory skin disorder, which has adverse effects on patients’ quality of life. Natural products exhibit significant therapeutic capacities with small side effects and might be preferable alternative treatments for patients with psoriasis. This study summarizes the signaling pathways with the potential targets of natural products and their efficacy for psoriasis treatment.

    Methods:

    The literature for this article was acquired from PubMed and Web of Science, from January 2010 to December 2020. The keywords for searching included “psoriasis” and “natural product”, “herbal medicine”, “herbal therapy”, “medicinal plant”, “medicinal herb” or “pharmaceutical plant”.

    Results:

    Herbal extracts, natural compounds, and herbal prescriptions could regulate the signaling pathways to alleviate psoriasis symptoms, such as T helper 17 (Th17) differentiation, Janus kinase (JAK)/signal transducer and activator of transcription (STAT), nuclear factor-kappa B (NF-κB), mitogen-activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR), and other signaling pathways, which are involved in the inflammatory response and keratinocyte hyperproliferation. The anti-psoriatic effect of natural products in clinical trials was summarized.

    Conclusions:

    Natural products exerted the anti-psoriatic effect by targeting multiple signaling pathways, providing evidence for the investigation of novel drugs. Further experimental research should be performed to screen and characterize the therapeutic targets of natural products for application in psoriasis treatment.

    Keywords

    Natural products, herbal medicine, psoriasis, mechanism, signaling pathway, target

    Introduction

    Psoriasis is a chronic, immune-mediated inflammatory skin disorder characterized by hyperproliferation and disrupted differentiation of keratinocytes and skin infiltration of inflammatory cells, leading to the formation of erythematous, scaly, and thickened plaques on skin lesions [1]. The psoriatic plaques symmetrically distribute with major occurrence on the extensor areas of elbows and knees, on the scalp, but also can appear on any skin surface of the body [2]. Psoriasis is one of the most common human skin diseases that affects 2–3% of the global population and the prevalence varies among different regions with the highest rate of approximately 11% in some European countries [35]. A variety of comorbidities associated with psoriasis have been reported, including psychological disorders, arthritis, and cardiovascular diseases that significantly reduced the quality of life of the patients [6].

    Psoriasis has been considered a multifactorial disease with the pathogenesis remains unclear. However, accumulating evidence has suggested that the complex interaction of genetic, immunological, and environmental factors plays a crucial role in the initiation as well as the progression of psoriasis [1]. In the early stage, peripheral dendritic cells (DCs) are recruited into skin lesions. In response to environmental stimuli, keratinocytes secrete pro-inflammatory cytokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α, which are involved in the activation of DCs in the dermis [7]. Subsequently, activated DCs produce various inflammatory mediators such as IL-12 and IL-23, to trigger the differentiation of naive T cells into T helper 1 (Th1) and Th17 cells. In turn, Th1 and Th17 also secrete TNF-α, interferon (IFN)-γ, IL-17, and IL-22, which have feedback to DCs. These pro-inflammatory molecules promote keratinocyte hyperproliferation and maintain chronic skin inflammation, which are hallmarks of psoriasis [8]. Apart from lymphocytes, monocytes and macrophages also play a role in the pathogenesis of psoriasis [9, 10]. The population of monocytes/macrophages was increased in the psoriatic skin lesions, compared to normal skin. These cells can produce IL-17 and TNF-α in response to cytokine stimulation, demonstrating their roles in skin inflammation in psoriasis [9].

    Psoriasis is an incurable disease; therefore, all available therapeutic approaches target alleviating skin manifestation of this disease. Treatments of psoriasis include topical application, systemic administration, and phototherapy. Corticosteroids (such as dexamethasone, clobetasol) and vitamin D analogs (tacalcitol, calcitriol) are the most common topical agents used to treat psoriasis by reducing inflammation, itching, and improving psoriatic scales [11, 12]. Oral administration of immunosuppressants (cyclosporine, methotrexate) or retinoids (acitretin) has shown beneficial effects on psoriasis symptoms by inhibiting inflammation and excessive proliferation of keratinocytes [13]. Recently, various biological drugs have been used in the management of psoriasis, including IL-17 inhibitors (secukinumab, brodalumab), IL-23 inhibitor (ustekinumab, tildrakizumab), or TNF-α inhibitors (infliximab, certolizumab), which directly target key molecules in the pathogenesis of psoriasis to inhibit the progression of the disease [14, 15]. Phototherapy is often suggested as an additional therapy for higher treatment outcomes [16]. However, all these therapies are associated with various adverse effects, leading to low satisfaction from the patients. For example, long-term application of steroids results in skin atrophy, susceptibility to infection, and risk of psychiatric disorders [17]. Patients who experienced low-dose long-term treatment of methotrexate might be suffered from liver and gastric abnormalities, bone marrow suppression, and hair loss [18]. Biologics such as secukinumab also have several side effects, including nasopharyngitis and upper respiratory tract infection [19]. This evidence raises concerns about alternative therapeutic approaches with fewer side effects for psoriasis management.

    Natural products or herbal medicines have been traditionally used to treat various chronic diseases for centuries, including psoriasis. In comparison with synthetic drugs, natural products exhibit fewer side effects, therefore they are preferable alternative treatments for patients with psoriasis. Approximately 50% of psoriasis patients in Southern Europe have used natural medicine during their treatment, this prevalence is up to 60% of patients in Asian countries [20, 21]. Herbal products possess a variety of bioactive components with a diversity of structures, pharmacological activities, and multiple mechanisms of action, leading to their potentials for an effective treatment, which cannot be observed in synthetic drugs [22]. Moreover, natural products are considered cost-effective and safe for patients. Therefore, studies employing herbal medicines for anti-psoriatic activity are still conducted to investigate new alternative treatments for psoriasis. This review aims to summarize the potential signaling pathways and targets, as well as the efficacy of natural products in the treatment of psoriasis based on the results from both preclinical and clinical studies.

    Materials and methods

    PubMed and Web of Science databases were used for searching the literature published from January 2010 to December 2020 for this review article. The keywords included “psoriasis” and “natural product”, “herbal medicine”, “herbal therapy”, “medicinal plant”, “medicinal herb” or “pharmaceutical plant”.

    Inclusion criteria include clinical studies using natural products (herbs, natural compounds, herbal formula) with placebo or drug control treatment and preclinical studies demonstrated effects and target signaling pathways of natural products in psoriasis treatment. The flow chart of this study is shown in Figure 1.

    Flow chart of study selection

    Results

    Signaling pathways and targets of natural products related to psoriasis

    Various signaling pathways have been demonstrated to play a role in the development of psoriasis. The effects of natural products, including herbs, natural compounds, and herbal formulas on psoriasis-related signaling pathways are shown in Tables 13.

    The effects of extracts on psoriasis

    PlantUsed partExtract methodIn vitroIn vivoSignaling pathway(s)Target(s)Reference
    Actinidia argutaFruitWaterHaCaTMiceNF-κB; STATIL-17[87]
    Alpinia galangaRhizomeEtOHHaCaT-NF-κBIL-8, CD40, CSF1[45]
    Annona squamosaLeaf
    Curcuma longaRhizome
    Antrodia cinnamomeaFruitEtOHT cellsMiceTh17 cell differentiationIL-17A, IL-21, IL-22, TNF-α[30]
    Artemisia capillarisWhole partEtOHHaCaTMiceApoptosisKi67, ICAM-1[103]
    Astragalus sinicus L.RootMeOH/CH2Cl2HaCaT; T cellsMiceNF-κB; JAK/STAT; PI3K/AktIL-17, IL-22, IL-10[86]
    Baphicacanthus cusia (Ness) BremekAerial part-aHK; HMEC-1; Jurkat T; U937MiceTh17 cell differentiationS100A9, IL-6, IL-8, CCL20[31]
    Copaifera langsdorffii Desf.Oleoresin-THP-1-NF-κBIL-1β, IL-6, TNF-α[46]
    Datura metel L.FlowerEtOH-MiceTLR7/8–MyD88– NF-κB–NLRP3 inflammasomeIL-1β, IL-2, IL-6, IL-10, IL-12, IL-17, IL-22, IL-23, TNF-α, MCP-1[89]
    Dictamnus dasycarpus Turcz.Root barkMeOH-MiceSTAT3; Th17 cell differentiationIL-17, IFN-γ[72]
    Euphorbia kansuiRootMeOHT cellsMiceTh17 cell differentiationIL-17, IL-22, IL-23, IL-12[32]
    Gynura pseudochina (L.)LeafEtOHHaCaT-NF-κBIL-8[44]
    Illicium verum Hook. f.FruitEtOHHaCaT-JAK/STATICAM-1[60]
    Lavandula angustifoliaEssential oil--MiceTh17 cell differentiationIL-17, IL-22, TNF-α, IL-1β[28]
    Picea marianaCortexWaterKeratinocytes-NF-κBICAM-1, IL-6, IL-8, NO[47]
    Pinus massonianaRosinWaterSplenocytesMiceTh17 cell differentiationIL-17, IL-22, IL-23, TNF-α, K17, PCNA[27]
    Rehmannia glutinosaRootEtOH-MiceJAK/STATTNF-α, IL-6, IL-23, IL-17[61]
    Salvia miltiorrhizaRoot-HaCaTMiceHippoCaspase-3, Bcl-2, BAX, p53, p21[105]
    Sinapis Alba LinnSeed--MiceNLRP3 inflammasomeIL-1β, IL-18, caspase-1, ASC[88]
    SplenocytesNF-κBIL-17, IL-22, IFN-α, iNOS[90]
    Solanum xanthocarpumStemEtOH-MiceTh17 cell differentiationTNF-α, IL-1β, IL-6, IL-17[29]
    Tripterygium wilfordii Hook. f.--T cellsMiceSTAT3IL-17, IL-22[62]
    Vitis vinifera L.LeafWaterTHP-1; HEKaMiceAIM2 inflammasomeIL-17, IL-1β, IL-18, caspase-1, ASC[104]
    Withania somniferaSeed-A431; THP-1; RAW264.7MiceNF-κBTNF-α, IL-6[48]
    Display full size

    aHK: adult human keratinocyte; AIM2: absent in melanoma 2; ASC: apoptosis-associated speck-like protein containing a caspase recruitment domain; BAX: B cell lymphoma-2 associated X; Bcl-2: B cell lymphoma-2; CCL20: C-C motif chemokine ligand 20; CH2Cl2: dichloromethane; CSF1: colony-stimulating factor 1; EtOH: ethanol; HaCaT: human immortalized keratinocytes; HEKa: human epidermal keratinocytes, adult; HMEC-1: human dermal microvascular endothelial cell line; ICAM-1: intercellular adhesion molecule-1; iNOS: inducible nitric oxide synthase; JAK: Janus kinase; MCP-1: monocyte chemotactic protein 1; MeOH: methanol; MyD88: myeloid differentiation primary response 88; NF-κB: nuclear factor-kappa B; NLRP3: nucleotide-binding oligomerization domain-like receptor protein 3; NO: nitric oxide; PCNA: proliferating cell nuclear antigen; PI3K: phosphatidylinositol 3-kinase; STAT: signal transducer and activator of transcription; THP-1: Tohoku hospital pediatrics-1; TLR: toll-like receptor; -: not applicable

    The effects of natural compounds on psoriasis

    CompoundPlantIn vitroIn vivoSignaling pathway(s)Target(s)Reference
    9,19-cycloartenol glycosides G3Cimicifuga simplexPBMCsMiceJAK/STAT; Th17 cell differentiationIL-17, IL-10, IFN-γ, IL-4[73]
    Aloe polysaccharideAloe veraHaCaT-NF-κBTNF-α, IL-12, IL-8[51]
    AndrographolideAndrographis paniculataBMDCsMiceTLR/MyD88IL-23, IL-1β, IL-6[106]
    Ar-turmeroneCurcuma longaHaCaT-HedgehogTNF-α, IL-8, IL-6, IL-1β[107]
    BaicalinScutellaria baicalensis-MiceTh17 cell differentiationIL-17, IL-22, IL-23, TNF-α[33]
    Betulinic acid-T cellsMiceTh17 cell differentiationIL-17, IL-10, IFN-γ[34]
    ChebulaninTerminalia chebula Retz.HaCaTMiceNF-κBIL-17, IL-23, TNF-α, MMP-9[53]
    Chrysin-NHEKMiceMAPK; JAK/STATIL-17, IL-22, TNF-α, CCL20[91]
    cis-Khellactone-RAW264.7MiceNF-κB; Th17 cell differentiationIL-17, IL-23, TNF-α, IL-1β, IL-6[50]
    CryptotanshinoneSalvia miltiorrhiza Bunge.HaCaTMiceSTAT3PCNA[63]
    CurcuminCurcuma longaHaCaT-NF-κBIL-6, IL-8[94]
    PBMCsTh17 cell differentiationIL-17, IFN-γ[96]
    T cellsMiceTh17 cell differentiationIL-17, IL-22, IFN-γ, IL-2, IL-8, TNF-α[95]
    Fisetin-Human keratinocytes-PI3K/Akt/mTOR; MAPKIL-17, IFN-γ[108]
    Gambogic acidGarcinia harburyiHaCaT; HUVECMiceNF-κB; VEGFICAM-1, IL-17, IL-22, VEGFR2[97]
    GlucosidesPaeonia lactiflora Pall-MiceSTAT1/3; Th17 cell differentiationIL-17, IL-22, IL-23, TNF-α, IL-1β, IL-12, IL-6, IFN-γ, PCNA[75]
    GlycyrrhizinGlycyrrhiza glabraHaCaTMiceNF-κB; MAPKICAM-1[92]
    HonokiolMagnolia officinalisHUVEC; splenocytesMiceNF-κB; VEGFTNF-α, IFN-γ, VEGFR2[98]
    ImperatorinAngelica hirsutifloraNeutrophils; bEnd.3MiceAkt; MAPKKi67, MPO, Ly6G[109]
    Indigodole DStrobilanthes cusiaT cells-Th17 cell differentiationIL-17[35]
    IsogarcinolGarcinia mangostana L.HaCaTMiceTh17 cell differentiationIL-17, IL-22, IL-23, TNF-α, IL-2, IFN-γ[36]
    Luteolin-HaCaT; NHEK-NF-κBIL-6, IL-8, VEGF[52]
    Marumoside A
    Niazirin
    Sitosterol-3-O-β-d-glucoside
    Moringa oleifera L.THP-1MiceTh17 cell differentiationIL-12, IL-17, IL-22, IL-23[38]
    PaeoniflorinPaeonia lactiflora PallHaCaTMiceNF-κBIL-17, IL-22, IL-23, TNF-α[49]
    Saikosaponin ABupleurum ChinenseHEKaMiceNF-κB; NLRP3TNF-α, IL-1β, IL-6, IL-8[99]
    ShikoninLeptospermum erythrorhizonHaCaTMiceJAK/STAT3Caspase-3, cyclin E[65]
    -Cyclin D1, p21, p53[64]
    TussilagononeTussilago farfaraHaCaTMiceNF-κB; STAT3; Nrf2IL-6, IL-23, TNF-α, S100A7, CXCL8[93]
    VanillinVanilla planifolia-MiceTh17 cell differentiationIL-17, IL-23[37]
    Withasteroid BDatura metel L.PBMCsMiceJAK/STAT3; Th17 cell differentiationIL-17, IL-10[74]
    Display full size

    BMDCs: bone-marrow derived dendritic cells; CXCL8: chemokine ligand 8; HUVEC: human umbilical vein endothelial cell; MAPK: mitogen-activated protein kinase; MMP-9: matrix metalloproteinase-9; MPO: myeloperoxidase; mTOR: mammalian target of rapamycin; NHEK: normal human epidermal keratinocyte; Nrf2: nuclear factor-erythroid 2-related factor 2; PBMCs: peripheral blood mononuclear cells; VEGF: vascular endothelial growth factor; VEGFR2: vascular endothelial growth factor receptor 2; -: not applicable

    The effects of herbal formulas on psoriasis

    FormulaComposition PlantExtract methodIn vitroIn vivoSignaling pathway(s)Target(s)Reference
    PlantUsed part
    Bai Xuan Xia Ta Re PianEuphorbiae humifusae L.Aerial part--MiceTh17 cell differentiationIL-17, IL-23[40]
    Terminalia chebula Retz.Young fruit
    Ripe fruit
    Terminalia bellirica Roxb.Fruit
    Aloe veraLeaf
    Convolvulus scammoniaResin
    Dang Gui Liu Huang TangAngelica acutiloba Kitag.RootEtOHHaCaTMiceMAPK; STAT3IL-22, CXCL10, K16, K17, Ki67[76]
    Rehmannia glutinosa Libosch.
    Scutellaria baicalensis Georgi.
    Astragalus membranaceus Bunge.
    Coptis chinensis Franch.
    Phellodendron amurense Rupr.Peel
    Gold lotionCitrus sinensisPeelEtOHDCsMiceTh17 cell differentiationIL-17, IL-22[39]
    Citrus hassaku
    Citrus limon
    Citrus natsudaidai
    Citrus miyauchi Iyo
    Citrus unshiu
    PAMsCarthamus tinctorius-EtOHHaCaTMiceNF-κBIL-8, TNF-α, ICAM-1, IL-23[54]
    Lithospermum erythrorhizon
    Solanum indicum
    Cymbopogon distans
    PSORI-CM01Curcuma zedoariaRhizome-HaCaTMiceNF-κB; Th17 cell differentiationIL-6, IL-8, CXCL10, CCL20[100]
    Sarcandra glabraAerial part
    Smilax glabra Roxb.Rhizome
    Prunus mumeFruit
    Arnebia euchroma (Royle) Johnst.Root
    Paeonia lactiflora
    Glycyrrhiza uralensis
    PSORI-CM02Smilax glabra Roxb.RhizomeWaterHaCaTMicePI3K/Akt/mTORBeclin1, Atg 7, Atg 16L1, Atg 3[78]
    Sarcandra glabra (Thunb.) NakaiLeaf
    Paeonia lactiflora PallRootRAW264.7STAT1; STAT6TNF-α, iNOS, IL-1β[77]
    Curcuma phaeocaulis Val.
    Prunus mume Sieb. et Zucc.Fruit
    Psoriasis 1Smilacis glabraeRhizome-HaCaTRatNF-κB; STATTNF-α, IFN-γ, IL-22, IL-17, IL-1β, IL-4[102]
    Folium isatidisLeaf
    Isatis tinctoria L.Root
    Angelica sinensis
    Hedyotis diffusaAerial part
    Ligusticum striatumRhizome
    Plantago majorLeafPBMCs-STAT4IL-17, IL-23, TNF-α, IFN-γ, IL-2, IL-6, IL-4, TGF-β[101]
    Kochia scopariaFruit
    Lobelia chinensisAerial part
    Alisma orientaleRhizome
    Dictamnus dasycarpus Turcz.Cortex
    Glycyrrhiza uralensisRoot
    Display full size

    Atg7: autophagy related 7; PAMs: plant antimicrobial solutions; TGF-β: transforming growth factor-beta; -: not applicable

    Classical signaling pathways

    Th17 cell differentiation pathway

    Psoriasis has been considered an immune-mediated skin disease. Emerging evidence suggested the crucial role of Th17 cells in the pathogenesis of psoriasis. TGF-β in combination with proinflammatory cytokines, including IL-23, IL-6, and IL-1β, can drive the differentiation of naive T cells to Th17 cells. IL-23 further promotes the survival and proliferation of Th17 cells, as well as the migration of these cells into psoriatic skin lesions [23]. Th17 cells are considered a distinct subset of CD4+ Th cells by the ability to secrete IL-17, however, Th17 cells can also produce various inflammatory cytokines, such as IL-22, IL-21, IL-6, and TNF-α to promote inflammation and keratinocyte proliferation in psoriatic skin lesions [24]. Previous studies demonstrated that the serum levels of TGF-β, IL-17, IL-22, and IL-6 were significantly higher in patients with psoriasis compared with healthy subjects [25, 26].

    Water-processed rosin from Pinus massoniana significantly reduced the proportion of Th17 cells in the spleen and inhibited the expression of Th17-related cytokines, including IL-17, IL-22, IL-23, and TNF-α in imiquimod (IMQ)-induced psoriasis-like mouse model [27]. Lavandula angustifolia essential oil and its component linalool showed significant decreases in IL-17 and IL-22 levels in IMQ-induced skin lesions [28]. Treatment with an ethanolic extract of Solanum xanthocarpum stem inhibited the skin expression of IL-17, IL-1β, IL-6, and TNF-α in the psoriasis mouse model [29]. Antrodia cinnamomea extract exerted inhibitory effects on Th17 cell differentiation and the production of IL-17, IL-22, and TNF-α in IMQ-treated mice [30]. Indigo naturalis, an extract from leaves of Baphicacanthus cusia (Ness) Bremek significantly decreased the expression of IL-1β, TNF-α, and IL-23 in keratinocytes, as well as inhibited the production of IL-17 and IL-22 in Jurkat T cells [31]. A methanolic extract of Euphorbia kansui root alleviated psoriasis symptoms by inhibiting the production of IL-23, IL-17, and IL-22 in lymph nodes from psoriatic mice [32].

    Baicalin, the major flavonoid from Scutellaria baicalensis, inhibited IL-17 production in lymph nodes and decreased the expression of IL-23, TNF-α, IL-17, and IL-22 in skin lesions of IMQ-induced psoriatic mice [33]. Betulinic acid, a natural terpenoid, showed significant downregulation in the frequency of Th17 cells as well as the production of IL-17, TNF-α, and IL-6 in IMQ-treated mice [34]. Indigodole D extracted from Strobilanthes cusia suppressed IL-17 production in Th17 cells without any cytotoxicity [35]. Isogarcinol, a natural compound derived from Garcinia mangostana L. significantly inhibited Th17 cell differentiation and the expression of Th17-related cytokines, including IL-23, IL-6, TNF-α, IL-17, and IL-22 in a mouse model of psoriasis [36]. Vanillin, a phenolic aldehyde from Vanilla planifolia, suppressed the levels of IL-23 and IL-17 in psoriatic skin lesions [37]. Niazirin, sitosterol-3-O-β-d-glucoside, and marumoside A, three components of Moringa oleifera L., inhibited the production of IL-17, IL-22, and IL-23 in vitro, and reduced IL-17 messenger RNA (mRNA) level in vivo [38].

    Gold lotion, an ethanolic extract of a mixture from peels of six citrus fruits, including Citrus sinensis (navel oranges), Citrus hassaku, Citrus limon, Citrus natsudaidai, Citrus miyauchi Iyo, and Citrus unshiu (Satsuma) decreased the ratio of Th17 cells in the spleen and reduced the expression of IL-23, IL-6, TNF-α, IL-17, and IL-22 at mRNA levels in skin lesions in IMQ-induced mouse model of psoriasis [39]. Bai Xuan Xia Ta Re Pian, a traditional herbal formula consisting of six herbs (Euphorbia Humifusae Herba, Chebula Fructus, Terminalia bellirica Fructus, Chebula Fructus Immaturus, Aloe, and Resina Scammoniae) significantly suppressed the expression of IL-23 and IL-17 in the skin of psoriatic mice [40].

    NF-κB signaling pathway

    NF-κB is a key transcription factor involved in the regulation of various cellular biological processes, including inflammatory responses [41]. Clinical studies in adult patients with moderate to severe psoriasis indicated that the level of the active form of NF-κB was significantly upregulated in psoriatic plaques, compared with non-lesional psoriatic skin and normal skin [42]. NF-κB transcription factor is a homodimer or heterodimer of NF-κB subunits (NFKBs), including p65 (RELA), RELB, p50, p52, and c-REL. In the baseline state, NF-κB dimers form a complex with the inhibitor of NF-κB (IκB) in the cytosol. Upon stimuli such as TNF-α, IκB kinase (IKK) phosphorylates IκB, leading to proteasomal degradation of IκB and releasing of NF-κB from the complex. Free NF-κB dimers translocate from the cytosol into the nucleus and bind to the promoter regions to regulate the transcription of various target genes [43]. NF-κB transcription factor is involved in the pathogenesis of psoriasis by regulating the expression of numerous cytokines, chemokines, and adhesion molecules to modulate inflammation, as well as keratinocyte proliferation and differentiation [42].

    An ethanolic extract from leaves of Gynura pseudochina (L.) showed anti-psoriatic properties by inhibiting the translocation of NFKB RELB and suppressing the expression of IL-8 in TNF-α-stimulated HaCaT cells [44]. In vitro study suggested anti-psoriatic effect of three Thai medicinal herbs, including Alpinia galanga, Curcuma longa, and Annona squamosa by reducing the expression of NF-κB signaling-related genes, such as NF-κB1 (p50), NF-κB2 (p52), and RELA in HaCaT cells [45]. Oleoresin from Copaifera langsdorffii Desf. inhibited NF-κB nuclear translocation and decreased the production of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α in lipopolysaccharide (LPS)-stimulated THP-1 monocytes, suggesting its potential in psoriasis treatment [46]. Picea mariana extract decreased the expression of inflammatory molecules IL-6, IL-8, VEGF, and ICAM-1 by promoting phosphorylation and degradation of IκB, as well as suppressing phosphorylation of NF-κB in TNF-α-treated human keratinocytes [47]. Withania somnifera Dunal seed extract inhibited skin inflammation in 12-O-tetradecanoyl phorbol-13-acetate (TPA)-induced psoriasis in mice, reduced the expression of NF-κB and decreased the production of pro-inflammatory cytokines IL-6 and TNF-α in LPS-stimulated THP-1 cells [48].

    Paeoniflorin, the major bioactive compound from Paeonia lactiflora Pall, alleviated psoriasis-like skin symptoms in IMQ-induced mice and inhibited hyperproliferation by suppressing phosphorylation of IκB-α and NF-κB in psoriatic keratinocytes [49]. cis-Khellactone, a common pyranocoumarin, reduced IMQ-induced psoriasis-like skin inflammation and decreased LPS-induced production of pro-inflammatory cytokines in macrophages by inhibiting phosphorylation of IKKα/β and NF-κB p65 [50]. Aloe polysaccharide, the main constituent of Aloe vera, decreased TNF-α-induced inflammation and proliferation in HaCaT cells by inhibiting phosphorylation of p65 and increasing the expression of IκB-α [51]. Luteolin, a common flavone, showed inhibitory effects on TNF-α-induced production of IL-6, IL-8, and VEGF, as well as hyperproliferation in HaCaT cells and NHEKs by decreasing mRNA levels of two genes (NFKB1 and RELA) and inhibiting nuclear translocation of NF-κB [52]. Chebulanin, a natural polyphenol derived from Terminalia chebula Retz., ameliorated IMQ-induced psoriatic skin lesions in mice and reduced inflammation and proliferation in HaCaT cells by decreasing phosphorylation of p65 at both mRNA and protein levels [53].

    PAMs, a mixture of ethanolic extracts from Carthamus tinctorius, Lithospermum erythrorhizon, Solanum indicum, and Cymbopogon distans reduced skin symptoms in a psoriatic mouse model and inhibited the production of inflammatory cytokines and chemokines in HaCaT cells by suppressing nuclear translocation of NF-κB [54].

    JAK/STAT signaling pathway

    JAK/STAT pathway plays an important role in immune diseases by mediating various cytokine signalings to regulate inflammation and cell proliferation. JAK protein family includes four tyrosine kinases (TYKs): JAK1–3 and TYK2. The STAT family consists of seven members: STAT1–4, STAT5A, STAT5B, and STAT6. Upon binding of type I and II cytokines to their corresponding receptors, JAKs are activated and phosphorylated, leading to the recruitment and phosphorylation of STATs. Phosphorylated STATs can form dimers and translocate to the nucleus to regulate the transcription of various target genes involved in immune responses [55, 56]. Upregulated expression of JAK1 and STAT3 has been reported in skin lesions from patients with psoriasis, compared with normal skin. In addition, STAT3 expression had a positive correlation with the severity of psoriasis [57]. A variety of inflammatory cytokines related to psoriasis, such as IL-6, IL-23 can activate JAK/STAT signaling pathway to promote the development of psoriasis by triggering inflammatory response as well as keratinocyte proliferation in skin lesions [58]. Inhibition of JAK/STAT pathway by JAK inhibitors such as tofacitinib improved disease severity in patients with moderate-to-severe psoriasis [59], suggesting that modulation of JAK/STAT signaling pathway might a potential approach for psoriasis treatment.

    An ethanolic extract of Illicium verum Hook. f. exhibited therapeutic potential for psoriasis by suppressing IFN-γ-induced ICAM-1 production in HaCaT cells via inhibiting JAK/STAT signaling pathway and decreasing the adhesion between T cells and keratinocytes [60]. Rehmannia glutinosa root extract alleviated epidermal thickening and skin levels of proinflammatory cytokines (IL-6, IL-17, IL-23, TNFα) in the IMQ-induced psoriasis mouse model by suppressing phosphorylation of JAK1, JAK2, STAT1, and STAT3 [61]. Multi-glycoside of Tripterygium wilfordii Hook. f. inhibited the expression of IL-17 and IL-22 in psoriasis mice by suppressing STAT3 signaling pathway [62]. Cryptotanshinone, a bioactive compound from Salvia miltiorrhiza Bunge. ameliorated psoriasis-like symptoms in IMQ-treated mice and reduced keratinocyte hyperproliferation by inhibiting STAT3 signaling pathway [63]. Shikonin, a main component of Leptospermum erythrorhizon improved skin severity in psoriasis mice and inhibited proliferation, promoted apoptosis, and decreased VEGF production in IL-17-stimulated HaCaT cells by suppressing activation of JAK/STAT3 signaling [64, 65].

    Multiple signaling pathways

    JAK/STAT and related signaling pathways

    Studies indicated the involvement of JAK/STAT, particularly STAT3 signaling pathway in Th17 differentiation from naive T cells, leading to the production of various inflammatory cytokines, such as TNF-α, IL-17, IL-21, and IL-22 [66, 67]. Hyperactivation of STAT3 signaling triggered Th17 differentiation, while STAT3-deficiency resulted in impairment of Th17 differentiation in T cells [67]. Tofacitinib, an inhibitor of JAK/STAT pathway significantly inhibited the production of Th17 cytokines (IL-17, IL-22) [68]. In turn, IL-22 binds to its receptor and activates several downstream pathways, including JAK/STAT3, MAPK, and PI3K/Akt/mTOR signaling pathways [6971]. Other cytokines involved in Th17 differentiation, such as IL-6 and IL-23 also contributed to JAK/STAT activation [58].

    A methanolic extract of root bark of Dictamnus dasycarpus Turcz. improved scaly skin lesions, reduced the number of inflammatory cell infiltration, and decreased epidermal thickness in IMQ-induced psoriasis mice by inhibiting STAT3 signaling pathway and reducing the number of Th17 cells as well as IL-17 production [72]. 9,19-cycloartenol glycosides G3, the main component of Cimicifuga simplex exhibited anti-psoriatic effects by suppressing the differentiation of CD4+ T cell into Th17 phenotype and inhibiting IFN-γ-induced JAK/STAT activation [73]. Withasteroid B isolated from Datura metel L. showed the inhibitory effects on JAK/STAT signaling pathway and reduced the ratio of Th17 cells as well as the production of Th17-related inflammatory cytokines [74]. Total glucosides extracted from Paeonia lactiflora Pall alleviated IMQ-induced psoriasis-like skin symptoms in mice, inhibited Th17 differentiation, and suppressed phosphorylation of STAT1 and STAT3 [75].

    Dang Gui Liu Huang Tang, a traditional herbal formula consisting of Angelica acutiloba Kitag., Rehmannia glutinosa Libosch., Scutellaria baicalensis Georgi., Astragalus membranaceus Bunge., Coptis chinensis Franch., and Phellodendron amurense Rupr., improved psoriasis symptoms in IMQ-induced mice and reduced the production of inflammatory cytokines and chemokines, as well as suppressed hyperproliferation in human keratinocytes by inhibiting the activation of STAT3 and MAPK signaling pathways [76]. PSORI-CM02, a traditional formula including five herbs, Smilax glabra Roxb., Sarcandra glabra (Thunb.) Nakai, Paeonia lactiflora Pall, Curcuma phaeocaulis Val., and Prunus mume Sieb. et Zucc., showed anti-psoriatic effects by suppressing STAT1/6 and PI3K/Akt/mTOR signaling pathways in keratinocytes and immune cells [77, 78].

    NF-κB and related signaling pathways

    NF-κB pathway can be activated by several upstream pathways, including PI3K/Akt and MAPK signalings to regulate inflammation [79, 80]. Nrf2 signaling can attenuate NF-κB activation, and in contrast, NF-κB could suppress Nrf2 activity [81]. NF-κB and JAK/STAT signaling pathways are involved in the regulation of inflammatory response in psoriasis. A previous study demonstrated that JAK/STAT signaling synergized with NF-κB to activate the transcription of various inflammatory genes in response to stimuli [82]. NF-κB activation also modulates many downstream signaling pathways which are involved in the pathogenesis of psoriasis. NF-κB signaling shows intrinsic and extrinsic effects on Th17 differentiation [83]. VEGF signaling, which is important in regulating angiogenesis (a hallmark of psoriasis), is also a downstream pathway of NF-κB [84]. Moreover, NF-κB pathway was suggested to be involved in NLRP3 inflammasome activation in psoriasis [85].

    Astragalus sinicus L. extract suppressed the production of inflammatory molecules in TNF-α/IFN-γ-stimulated human keratinocytes and IL-23-induced psoriasis-like mouse model by inhibiting NF-κB, STAT1/3, and PI3K/Akt signaling pathways [86]. An aqueous extract of Actinidia arguta shows inhibitory effects on IMQ-induced cutaneous inflammation and cytokine-induced inflammation and hyperproliferation in HaCaT cells by suppressing phosphorylation of NF-κB p65 and STAT1/3 [87]. Extracts from Sinapis Alba Linn and Datura metel L. exerted anti-psoriatic effect by inhibiting NF-κB and NLRP3 inflammasome signaling pathways [8890].

    Chrysin, a common flavone found in various natural sources, such as honey, passion flowers, or propolis, alleviated IMQ-induced psoriasis symptoms in mice and inhibited the production of inflammatory cytokines, chemokines, and antimicrobial peptides in keratinocytes by suppressing NF-κB, MAPK, and JAK/STAT signaling pathways [91]. Glycyrrhizin, a major component of Glycyrrhiza glabra, improved psoriasis-like skin lesions in IMQ-induced mice and reduced ICAM-1 production in TNF-α-stimulated HaCaT cells by inhibiting NF-κB/MAPK signaling [92]. Tussilagonone, a natural compound derived from Tussilago farfara, alleviated psoriasis symptoms in IMQ-treated mice and TNF-α-treated keratinocytes via Nrf2 activation and NF-κB/STAT3 inhibition [93]. Curcumin, a main compound of Curcuma longa, attenuated psoriasis pathology by inhibiting and NF-κB and Th17 differentiation pathways in keratinocytes and immune cells [9496]. Honokiol (a lignan isolated from Magnolia officinalis) and gambogic acid (a xanthone from Garcinia harburyi) showed the anti-psoriatic effect by suppressing NF-κB/VEGF signaling pathway [97, 98]. Saikosaponin A, a component of Bupleurum chinense, reduced psoriasis area and severity index (PASI) scores and epidermal hyperplasia in IMQ-induced mice and attenuated cytokine-induced inflammation in human keratinocytes by suppressing the phosphorylation of NF-κB and the expression of NLRP3 [99].

    PSORI-CM01, a herbal formula consisting of seven plants Curcuma zedoaria, Sarcandra glabra, Smilax glabra Roxb., Prunus mume, Arnebia euchroma (Royle) Johnst., Paeonia lactiflora, and Glycyrrhiza uralensis, exerted the anti-psoriatic effect by suppressing the translocation of NF-κB p65 and inhibiting Th17 differentiation signaling [100]. Psoriasis 1, a mixture of 12 herbs including Smilacis glabrae, Folium isatidis, Isatis tinctoria L., Angelica sinensis, Hedyotis diffusa, Ligusticum striatum, Plantago major, Kochia scoparia, Lobelia chinensis, Alisma orientale, Dictamnus dasycarpus Turcz., and Glycyrrhiza uralensis, alleviated psoriasis inflammation by inhibiting the phosphorylation of IKK, NF-κB p65, STAT3, and STAT4 in keratinocytes and T cells [101, 102].

    Other signaling pathways

    An ethanolic extract of Artemisia capillaris ameliorated IMQ-induced psoriasis-like symptoms in mice and showed antiproliferative effect by promoting apoptosis in keratinocytes [103]. The leaf extract of Vitis vinifera L. alleviated psoriatic inflammation by inhibiting the activation of AIM2 inflammasome signaling [104]. Salvia miltiorrhiza extract [also known as danshensu in traditional Chinese medicine (TCM)] suppressed epidermal hyperplasia in IMQ-induced mice and hyperproliferation in cytokine-stimulated keratinocytes by reducing the expression of yes-associated protein (YAP, an important component of Hippo signaling pathway) [105]. Andrographolide, a major component from Andrographis paniculata, exerted the anti-psoriatic effect by inhibiting TLR/MyD88 signaling in DCs [106]. Ar-turmerone, a sesquiterpenoid from Curcuma longa, suppressed TNF-α-induced inflammation and proliferation in HaCaT cells by inhibiting Hedgehog signaling pathway [107]. Fisetin (a common flavonol) and imperatorin (a furocoumarin derived from Angelica hirsutiflora) alleviated psoriasis-like symptoms by suppressing PI3K/Akt/mTOR and MAPK signaling pathways in keratinocytes and immune cells, respectively [108, 109].

    Clinical efficacy of natural products in psoriasis treatment

    Clinical studies demonstrated the anti-psoriatic effects of natural products are shown in Table 4. Topical application of extract from sea buckthorn, Indigo naturalis, Hypericum perforatum, and Gynura pseudochina showed significant reductions in skin severity compared with placebo in patients with mild to moderate psoriasis [110113]. Sea buckthorn has been traditionally used for thousands of years for treatment of skin diseases due to its anti-inflammatory and immunomodulatory effects [114]. Indigo naturalis has been widely used for psoriasis treatment in TCM for over 50 years [115]. Hypericum perforatum and Gynura pseudochina are also common medicinal plants used to treat skin symptoms [116, 117]. Herbal formula Pulian ointment (consisting of two herbs: Phellodendron amurense and Scutellaria baicalensis) and Shi Du Ruan Gao (a mixture of six herbs: Indigo naturalis, Cortex Phellodendri, Gypsum fibrosum preparatum, Calamine, Galla chinensis) also exerted anti-psoriatic activity by decreasing PASI scores without any severe adverse events after four weeks and eight weeks of topical treatment, respectively [118, 119]. Pulian ointment has been approved by Beijing Food and Drug Administration as a prescription use in hospitals in China for treatment of psoriasis [119]. Shi Du Ruan Gao has been developed and used for psoriasis treatment in hospital for over 60 years [118].

    Clinical efficacy of natural products

    Herb/formulaType of studyPatientsTreatmentEfficacy outcomeAdverse effectsReference
    Gynura pseudochina var. hispida Thv.Randomized, positive-controlledn = 25
    Mild to moderate chronic plaque psoriasis
    Topical
    4 weeks
    Significant decrease in scaling scores, epidermal thickness, NF-κB p65, and Ki-67 expressionNo laboratory abnormalities[113]
    Indigo naturalisRandomized, double-blinded, placebo-controlledn = 24
    Moderate psoriasis
    Topical
    8 weeks
    Significant reduction in PASI scores compared with placeboNot evaluated[112]
    Liang xue huo xue (Radix Rehmanniae, Sophora flower, Salvia miltiorrhiza, Rhizoma Imperatae, puccoon, red peony, Caulis Spatholobi)Multi-center, randomized, double-blind, placebo-controlledn = 50
    Blood heat syndrome psoriasis
    Oral
    6 weeks
    Significant reduction in PASI scores and serum IL-17 level compared with placeboNo abnormal vital signs[120]
    Liangxue Jiedu (Rhizoma Smilacis Chinae, Flos Sophorae, Radix Lithospermi, Rhizoma Paridis, Radix Rehmanniae, Cortex Dictamni Radicis, Radix Paeoniae Rubra, Flos Lonicerae, Rhizoma Imperatae, Radix Sophorae Flavescentis)Multicenter, randomized, controlledn = 247
    Blood heat type psoriasis
    Oral
    8 weeks
    Significant improvement in skin lesions and symptoms compared with Western medicine treatmentNo significant abnormalities[124]
    Pulian (Phellodendron amurense, Scutellaria baicalensis)Multicenter, randomized, double-blind, placebo-controlledn = 300
    Blood heat syndrome psoriasis
    Topical
    4 weeks
    Significant reduction in PASI scores compared with placeboNo adverse event[119]
    Sea buckthornSingle-blind, placebo-controlled, randomizedn = 10
    PASI score: 1–12
    Topical
    4 weeks
    Significant improvement in PASI scores and DLQI scores compared with placeboNot evaluated[110]
    Shi Du Ruan Gao (Indigo naturalis, Cortex Phellodendri, Gypsum fibrosum preparatum, Calamine, Galla chinensis)Single-center, randomized, investigator-blinded, parallel group, placebo-controlledn = 100
    Mild to moderate chronic plaque psoriasis
    Topical
    8 weeks
    Significant improvement in the TSS, IGA, and Global Subjects’ Assessment of treatment compared with placeboNo severe adverse events[118]
    St John’s wort (Hypericum perforatum L.)Single-blind, placebo-controlledn = 10
    Symmetrical plaque-type psoriasis
    Topical
    4 weeks
    Significant reduction in PASI scores compared with placeboNot evaluated[111]
    Yinxieling (Radix Rehmanniae recen, Angelica sinensis, Radix Paeoniae Rubra, Ligusticum wallichii, Lithospermi radix, Curcuma zedoaria Chloranthus spicatus, Rhizoma Smilacis glabrae, smoked plum, liquorice)Randomized controlledn = 120Oral
    8 weeks
    Significant reduction in PASI scores and serum level of TNF-α and IL-8 compared with placeboNot evaluated[121]
    Display full size

    DLQI: dermatology life quality index; IGA: investigator global assessment; TSS: total severity score

    Oral administration of herbal formula Liang xue huo xue decoction (seven herbs: Radix Rehmanniae, Sophora flower, Salvia miltiorrhiza, Rhizoma Imperatae, puccoon, red peony, Caulis Spatholobi) and Yinxieling (10 herbs: Radix Rehmanniae recen, Angelica sinensis, Radix Paeoniae Rubra, Ligusticum wallichii, Radices lithospermi, Curcuma zedoaria, Chloranthus spicatus, Rhizoma Smilacis glabrae, smoked plum, liquorice) significantly improved PASI scores and reduced serum levels of inflammatory cytokines in psoriasis patients, compared with placebo [120, 121]. Liang xue huo xue was used for psoriasis treatment due to its anti-proliferative effects on keratinocytes [122]. Yinxieling has been applied in clinical and exerted effectiveness in treatment of psoriasis [123]. Treatment with Liangxue Jiedu decoction (10 herbs: Rhizoma Smilacis Chinae, Flos Sophorae, Radix Lithospermi, Rhizoma Paridis, Radix Rehmanniae, Cortex Dictamni Radicis, Radix Paeoniae Rubra, Flos Lonicerae, Rhizoma Imperatae, Radix Sophorae Flavescentis) showed significant improvements in skin symptoms in comparison with Western medicine (cetirizine hydrochloride, vitamin C, and vitamin B complex) after eight weeks [124]. Liangxue Jiedu decoction was used for treatment of psoriasis in TCM due to its immunomodulatory activity [125].

    All the nine clinical studies mentioned in Table 1 were randomized studies with single-blind or double-blind, single-center or multi-center, and placebo-controlled or positive-controlled observation. These studies were conducted in small groups of patients (10–50 patients) or larger groups (100–300 patients). Participants were included in clinical studies consisting of both men and women, aged from 18 years old to 80 years old with skin symptoms from mild to severe. Some studies specifically targeted the patients with the blood heat syndrome based on TCM diagnosis. According to TCM, blood heat is the most common syndrome (53.8%) in patients with psoriasis, compared with blood-dryness (27.4%) and blood-stasis syndrome (18.1%) [126]. Hence, the number of blood heat type psoriasis patients might be large enough for studies rather than other types. Moreover, blood heat type patients also exhibited typical features of psoriasis with elevated levels of Th1/Th17-related cytokines, IFN-γ, IL-17, IL-23, and TNF-α [127]. Duration of treatments ranged from four weeks to eight weeks for topical application and from six weeks to eight weeks for oral administration. Both oral and topical treatment showed therapeutic effects on psoriasis in comparison with placebo or positive control drugs with no significant adverse events.

    Herbal products were also used in combination with other therapies in the treatment of psoriasis. Oral administration of Curcuma longa extract combined with ultraviolet A (UVA) therapy showed higher effects on skin severity compared with psoralen plus UVA [128]. Treatment with total glucosides of paeony, a bioactive component derived from dry paeony root in combination with acitretin significantly improved PASI50 (50% reduction of PASI scores) in patients with moderate-to-severe plaque psoriasis, in comparison with placebo plus acitretin [129]. Oral treatment of a Korean herbal formula Yangdokbagho-tang (a mixture of six ingredients: Gypsum fibrosum, Rehmanniae Radix Crudus, Anemarrhenae Rhizoma, Schizonepetae Spica, Saposhnikoviae Radix, Arctii Semen) combined with acupuncture, probiotics, and phototherapy reduced PASI scores in two cases of moderate and severe psoriasis [130].

    Several clinical trials also demonstrated that there were no significant differences between the effects of natural products and placebo or drug treatment on psoriasis symptoms. Application of ointment with silver fir (Abies alba) bark showed no significant effects compared with placebo [131]. The anti-psoriatic effects of Tripterygium wilfordii extract were not significantly different in comparison with acitretin [132]. Oral treatment with a TCM formula consisting of 16 herbs (Herba ephedrae, Radix aconiti lateralis preparate, Semen sinapis, Cortex cinnamomi, Rhizoma zingiberis, Cornu cervi degelatinatum, Radix rehmanniae preparate, Rhizoma Smilacis Glabrae, Cortex Dictamni, Rhizoma Imperatae, Radix salviae miltiorrhizae, Caulis spatholobi, Arnebiae Radix, Flos Sophorae, Radix glycyrrhizae, Indigo naturalis) for six months showed less effective outcomes compared with both placebo and methotrexate [133].

    Discussion

    Natural products have been used to treat psoriasis for centuries with significant effectiveness and few adverse events. In the aspect of TCM, psoriasis includes three phenotypes: blood heat in the active stage, blood dryness in the regression stage, and blood stasis in the resting stage. Blood heat phenotype is characterized by the continuous appearance of spot-like skin rash and skin itching. Blood dryness features include coin-like skin rash with light red color. The symptoms of blood stasis are thickened dark red skin lesions. Among these three phenotypes, blood heat is the most common with over 50% of patients suffering from this syndrome [126]. Several herbal formulas listed in Table 4 exerted efficacy on the treatment of blood heat type of psoriasis. Among the clinical trials listed in Table 4, studies investigating the clinical efficacy of Liangxue Jiedu and Pulian might be considered most valuable due to the large numbers of participants in these studies. However, since the scientific evidence for traditional herbal medicines or natural products is still limited, large-scale clinical trials (1,000 participants or more) to examine their efficacies have not been conducted. This review organized and summarized the underlying mechanisms for anti-psoriatic effects of natural products, which support the scientific base for future clinical trials.

    Medicinal herbs and traditional herbal formulas consist of various active compounds with multiple targets and multiple related signaling pathways. This characteristic might lead to the higher effects of natural products but might be an obstacle to investigate their mechanisms of action for psoriasis treatment. Moreover, the chemical composition of a plant might vary in the number of compounds, as well as the amount of each compound under different growth environments, leading to the difficulty in the repetition of experiments. Therefore, clarification of the major component in the plants is important in the study of herbal medicines. In this review, we summarized the anti-psoriatic effects of natural products, including natural compounds, herb extracts, and herbal prescriptions. These natural products target numerous psoriasis-related signaling pathways, such as Th17 differentiation, JAK/STAT, NF-κB, MAPK, PI3K/Akt/mTOR, and other signaling pathways to alleviate inflammatory response and reducing keratinocyte hyperproliferation, thus improving psoriasis symptoms. Based on the current review, inhibiting Th17 differentiation pathway as well as related targets, such as IL-17, IL-22, IL-23, and TNF-α was the most common mechanism of action of natural products against psoriasis. These targets might be considered the most credible targets and might be applied in psoriasis studies using natural products.

    Most animal studies utilized IMQ-induced mice as a model of psoriasis. Application of IMQ (a ligand of TLR7/8) to mouse skin can induce inflammatory skin lesions, resembling psoriasis symptoms in humans with activation of IL-23/IL-17 axis [134]. After the first report in 2009, IMQ-induced psoriasis-like skin inflammation in mice was widely used to investigate new underlying mechanisms in the pathogenesis of psoriasis, as well as to examine the therapeutic effects of potential agents. However, this model has certain limitations. There are several critical differences between mouse and human skin, including permeability, thickness, cutaneous immunity, and renewal process of epidermis and hair follicles, leading to the differences in drug absorption and immune response in the mouse model, compared with the human [135]. Therefore, the use of other models, which more resemble human skin, such as human three-dimensional skin equivalents is necessary and appropriate to investigate the anti-psoriatic effect of natural products.

    In conclusion, natural products show promising application in the treatment of psoriasis. The underlying mechanisms of action for the anti-psoriatic effect of natural compounds and herbal products are complex with the involvement of multiple signaling pathways (Figure 2). Further studies to evaluate the therapeutic effects of natural products in more relevant psoriasis models and larger-scale clinical trials should be conducted in the future.

    The signaling pathways underlying anti-psoriatic effects of natural products. Act1: NF-κB activator 1; P: phosphorylated; RIP: ribosome inactivating protein: TAK1: transforming growth factor-β-activated kinase 1; TRADD: Tumor necrosis factor receptor type 1-associated death domain; TRAF6: tumor necrosis factor receptor-associated factor 6

    Abbreviations

    AIM2:

    absent in melanoma 2

    DCs:

    dendritic cells

    HaCaT:

    human immortalized keratinocytes

    ICAM-1:

    intercellular adhesion molecule-1

    IFN:

    interferon

    IKK:

    inhibitor of nuclear factor-kappa B kinase

    IL:

    interleukin

    IMQ:

    imiquimod

    IκB:

    inhibitor of nuclear factor-kappa B

    JAK:

    Janus kinase

    LPS:

    lipopolysaccharide

    MAPK:

    mitogen-activated protein kinase

    mRNA:

    messenger RNA

    mTOR:

    mammalian target of rapamycin

    MyD88:

    myeloid differentiation primary response 88

    NF-κB:

    nuclear factor-kappa B

    NFKBs:

    nuclear factor-kappa B subunits

    NHEK:

    normal human epidermal keratinocyte

    NLRP3:

    nucleotide-binding oligomerization domain-like receptor protein 3

    Nrf2:

    nuclear factor-erythroid 2-related factor 2

    PAMs:

    plant antimicrobial solutions

    PASI:

    psoriasis area and severity index

    PI3K:

    phosphatidylinositol 3-kinase

    STAT:

    signal transducer and activator of transcription

    TCM:

    traditional Chinese medicine

    TGF-β:

    transforming growth factor-beta

    Th17:

    T helper 17

    THP-1:

    Tohoku hospital pediatrics-1

    TLR:

    toll-like receptor

    TNF:

    tumor necrosis factor

    VEGF:

    vascular endothelial growth factor

    Declarations

    Author contributions

    The author contributed solely to the work.

    Conflicts of interest

    The author declares that there are no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2022.

    References

    Di Meglio P, Villanova F, Nestle FO. Psoriasis. Cold Spring Harb Perspect Med. 2014;4:a015354. [DOI] [PubMed] [PMC]
    Kimmel GW, Lebwohl M. Psoriasis: overview and diagnosis. In: Bhutani T, Liao W, Nakamura M, editors. Evidence-based psoriasis: diagnosis and treatment. Cham: Springer; 2018. pp. 116. [DOI] [PMC]
    Parisi R, Symmons DP, Griffiths CE, Ashcroft DM; Identification and Management of Psoriasis and Associated ComorbidiTy (IMPACT) project team. Global epidemiology of psoriasis: a systematic review of incidence and prevalence. J Invest Dermatol. 2013;133:37785. [DOI] [PubMed]
    Danielsen K, Olsen AO, Wilsgaard T, Furberg AS. Is the prevalence of psoriasis increasing? A 30-year follow-up of a population-based cohort. Br J Dermatol. 2013;168:130310. [DOI] [PubMed]
    Egeberg A, Andersen YMF, Thyssen JP. Prevalence and characteristics of psoriasis in Denmark: findings from the Danish skin cohort. BMJ Open. 2019;9:e028116. [DOI] [PubMed] [PMC]
    Takeshita J, Grewal S, Langan SM, Mehta NN, Ogdie A, Van Voorhees AS, et al. Psoriasis and comorbid diseases: epidemiology. J Am Acad Dermatol. 2017;76:37790. [DOI] [PubMed] [PMC]
    Chu CC, Di Meglio P, Nestle FO. Harnessing dendritic cells in inflammatory skin diseases. Semin Immunol. 2011;23:2841. [DOI] [PubMed] [PMC]
    Rendon A, Schäkel K. Psoriasis pathogenesis and treatment. Int J Mol Sci. 2019;20:1475. [DOI] [PubMed] [PMC]
    Wang Y, Edelmayer R, Wetter J, Salte K, Gauvin D, Leys L, et al. Monocytes/Macrophages play a pathogenic role in IL-23 mediated psoriasis-like skin inflammation. Sci Rep. 2019;9:5310. [DOI] [PubMed] [PMC]
    Gong X, Wang W. Profiles of innate immune cell infiltration and related core genes in psoriasis. Biomed Res Int. 2021;2021:6656622. [DOI] [PubMed] [PMC]
    Kleyn EC, Morsman E, Griffin L, Wu JJ, Cm van de Kerkhof P, Gulliver W, et al. Review of international psoriasis guidelines for the treatment of psoriasis: recommendations for topical corticosteroid treatments. J Dermatolog Treat. 2019;30:3119. [DOI] [PubMed]
    Koo K, Jeon C, Bhutani T. Beyond monotherapy: a systematic review on creative strategies in topical therapy of psoriasis. J Dermatolog Treat. 2017;28:7028. [DOI] [PubMed]
    Lebwohl M, Ali S. Treatment of psoriasis. Part 2. Systemic therapies. J Am Acad Dermatol. 2001;45:64964. [DOI] [PubMed]
    Radi G, Campanati A, Diotallevi F, Bianchelli T, Offidani A. Novel therapeutic approaches and targets for treatment of psoriasis. Curr Pharm Biotechnol. 2020;22:731. [DOI] [PubMed]
    Yost J, Gudjonsson JE. The role of TNF inhibitors in psoriasis therapy: new implications for associated comorbidities. F1000 Med Rep. 2009;1:30. [DOI] [PubMed] [PMC]
    Kemény L, Varga E, Novak Z. Advances in phototherapy for psoriasis and atopic dermatitis. Expert Rev Clin Immunol. 2019;15:120514. [DOI] [PubMed]
    Yasir M, Goyal A, Bansal P, Sonthalia S. Corticosteroid adverse effects. Treasure Island (FL): StatPearls Publishing; 2022. [PubMed]
    Kremer JM. Major side effects of low-dose methotrexate [Internet]; c2021 [cited 2021 Apr 23]. Available from: https://www.uptodate.com/contents/major-side-effects-of-low-dose-methotrexate
    Mease PJ, McInnes IB, Kirkham B, Kavanaugh A, Rahman P, van der Heijde D, et al.; FUTURE 1 Study Group. Secukinumab inhibition of interleukin-17A in patients with psoriatic arthritis. N Engl J Med. 2015;373:132939. [DOI] [PubMed]
    Damevska K, Neloska L, Nikolovska S, Gocev G, Duma S. Complementary and alternative medicine use among patients with psoriasis. Dermatol Ther. 2014;27:2813. [DOI] [PubMed]
    Deng S, May BH, Zhang AL, Lu C, Xue CC. Plant extracts for the topical management of psoriasis: a systematic review and meta-analysis. Br J Dermatol. 2013;169:76982. [DOI] [PubMed]
    Svendsen MT, Jeyabalan J, Andersen KE, Andersen F, Johannessen H. Worldwide utilization of topical remedies in treatment of psoriasis: a systematic review. J Dermatolog Treat. 2017;28:37483. [DOI] [PubMed]
    Elloso MM, Gomez-Angelats M, Fourie AM. Targeting the Th17 pathway in psoriasis. J Leukoc Biol. 2012;92:118797. [DOI] [PubMed]
    Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol. 2007;25:82152. [DOI] [PubMed]
    Zaher H, Shaker OG, EL-Komy M, El-Tawdi A, Fawzi M, Kadry D. Serum and tissue expression of transforming growth factor beta 1 in psoriasis. J Eur Acad Dermatol Venereol. 2009;23:4069. [DOI] [PubMed]
    Oliveira PSSd, Cardoso PRG, Lima EVdA, Pereira MC, Duarte ALBP, Pitta IdR, et al. IL-17A, IL-22, IL-6, and IL-21 serum levels in plaque-type psoriasis in Brazilian patients. Mediators Inflamm. 2015;2015:819149. [DOI] [PubMed] [PMC]
    Li XQ, Chen Y, Zhou HM, Shi HL, Yan XN, Lin LP, et al. Anti-psoriasis effect of water-processed rosin in mice. J Ethnopharmacol. 2019;242:112073. [DOI] [PubMed]
    Rai VK, Sinha P, Yadav KS, Shukla A, Saxena A, Bawankule DU, et al. Anti-psoriatic effect of Lavandula angustifolia essential oil and its major components linalool and linalyl acetate. J Ethnopharmacol. 2020;261:113127. [DOI] [PubMed]
    Parmar KM, Itankar PR, Joshi A, Prasad SK. Anti-psoriatic potential of Solanum xanthocarpum stem in imiquimod-induced psoriatic mice model. J Ethnopharmacol. 2017;198:15866. [DOI] [PubMed]
    Li MH, Wu HC, Yao HJ, Lin CC, Wen SF, Pan IH. Antrodia cinnamomea extract inhibits Th17 cell differentiation and ameliorates imiquimod-induced psoriasiform skin inflammation. Am J Chin Med. 2015;43:140117. [DOI] [PubMed]
    Cheng HM, Kuo YZ, Chang CY, Chang CH, Fang WY, Chang CN, et al. The anti-TH17 polarization effect of Indigo naturalis and tryptanthrin by differentially inhibiting cytokine expression. J Ethnopharmacol. 2020;255:112760. [DOI] [PubMed]
    Kim SJ, Jang YW, Hyung KE, Lee DK, Hyun KH, Park SY, et al. Therapeutic effects of methanol extract from Euphorbia kansui radix on imiquimod-induced psoriasis. J Immunol Res. 2017;2017:7052560. [DOI] [PubMed] [PMC]
    Hung CH, Wang CN, Cheng HH, Liao JW, Chen YT, Chao YW, et al. Baicalin ameliorates imiquimod-induced psoriasis-like inflammation in mice. Planta Med. 2018;84:11107. [DOI] [PubMed]
    Liu CH, Chen YC, Lu CJ, Chen HM, Deng JW, Yan YH, et al. Betulinic acid suppresses Th17 response and ameliorates psoriasis-like murine skin inflammation. Int Immunopharmacol. 2019;73:34352. [DOI] [PubMed]
    Lee CL, Wang CM, Kuo YH, Yen HR, Song YC, Chou YL, et al. IL-17A inhibitions of indole alkaloids from traditional Chinese medicine Qing Dai. J Ethnopharmacol. 2020;255:112772. [DOI] [PubMed] [PMC]
    Chen S, Han K, Li H, Cen J, Yang Y, Wu H, et al. Isogarcinol Extracted from Garcinia mangostana L. ameliorates imiquimod-induced psoriasis-like skin lesions in mice. J Agric Food Chem. 2017;65:84657. [DOI] [PubMed]
    Cheng HM, Chen FY, Li CC, Lo HY, Liao YF, Ho TY, et al. Oral administration of vanillin improves imiquimod-induced psoriatic skin inflammation in mice. J Agric Food Chem. 2017;65:1023342. [DOI] [PubMed]
    Ma N, Tang Q, Wu WT, Huang XA, Xu Q, Rong GL, et al. Three constituents of Moringa oleifera seeds regulate expression of Th17-relevant cytokines and ameliorate TPA-induced psoriasis-like skin lesions in Mice. Molecules. 2018;23:3256. [DOI] [PubMed] [PMC]
    Lin CC, Wu JJ, Pan YG, Chao YH, Lin FC, Lee YR, et al. Gold lotion from citrus peel extract ameliorates imiquimod-induced psoriasis-like dermatitis in murine. J Sci Food Agric. 2018;98:550917. [DOI] [PubMed]
    Pang X, Zhang K, Huang J, Wang H, Gao L, Wang T, et al. Decryption of Active constituents and action mechanism of the traditional uighur prescription (BXXTR) alleviating IMQ-induced psoriasis-like skin inflammation in BALB/c Mice. Int J Mol Sci. 2018;19:1822. [DOI] [PubMed] [PMC]
    Lawrence T, Gilroy DW, Colville-Nash PR, Willoughby DA. Possible new role for NF-κB in the resolution of inflammation. Nat Med. 2001;7:12917. [DOI] [PubMed]
    Lizzul PF, Aphale A, Malaviya R, Sun Y, Masud S, Dombrovskiy V, et al. Differential expression of phosphorylated NF-κB/RelA in normal and psoriatic epidermis and downregulation of NF-κB in response to treatment with etanercept. J Invest Dermatol. 2005;124:127583. [DOI] [PubMed]
    Oeckinghaus A, Ghosh S. The NF-κB family of transcription factors and its regulation. Cold Spring Harb Perspect Biol. 2009;1:a000034. [DOI] [PubMed] [PMC]
    Sukadeetad K, Nakbanpote W, Heinrich M, Nuengchamnong N. Effect of drying methods and solvent extraction on the phenolic compounds of Gynura pseudochina (L.) DC. leaf extracts and their anti-psoriatic property. Ind Crops Prod. 2018;120:3446. [DOI]
    Saelee C, Thongrakard V, Tencomnao T. Effects of Thai medicinal herb extracts with anti-psoriatic activity on the expression on NF-κB signaling biomarkers in HaCaT keratinocytes. Molecules. 2011;16:390832. [DOI] [PubMed] [PMC]
    Gelmini F, Beretta G, Anselmi C, Centini M, Magni P, Ruscica M, et al. GC-MS profiling of the phytochemical constituents of the oleoresin from Copaifera langsdorffii Desf. and a preliminary in vivo evaluation of its antipsoriatic effect. Int J Pharm. 2013;440:1708. [DOI] [PubMed]
    García-Pérez ME, Allaeys I, Rusu D, Pouliot R, Janezic TS, Poubelle PE. Picea mariana polyphenolic extract inhibits phlogogenic mediators produced by TNF-α-activated psoriatic keratinocytes: impact on NF-κB pathway. J Ethnopharmacol. 2014;151:26578. [DOI] [PubMed]
    Balkrishna A, Nain P, Chauhan A, Sharma N, Gupta A, Ranjan R, et al. Super critical fluid extracted fatty acids from Withania somnifera seeds repair psoriasis-like skin lesions and attenuate pro-inflammatory cytokines (TNF-α and IL-6) release. Biomolecules. 2020;10:185. [DOI] [PubMed] [PMC]
    Bai X, Yu C, Yang L, Luo Y, Zhi D, Wang G, et al. Anti-psoriatic properties of paeoniflorin: suppression of the NF-kappaB pathway and Keratin 17. Eur J Dermatol. 2020;30:24350. [DOI] [PubMed]
    Feng L, Song P, Xu F, Xu L, Shao F, Guo M, et al. cis-Khellactone inhibited the proinflammatory macrophages via promoting autophagy to ameliorate imiquimod-induced psoriasis. J Invest Dermatol. 2019;139:194656.E3. [DOI] [PubMed]
    Leng H, Pu L, Xu L, Shi X, Ji J, Chen K. Effects of aloe polysaccharide, a polysaccharide extracted from Aloe vera, on TNF-α-induced HaCaT cell proliferation and the underlying mechanism in psoriasis. Mol Med Rep. 2018;18:353743. [DOI] [PubMed]
    Weng Z, Patel AB, Vasiadi M, Therianou A, Theoharides TC. Luteolin inhibits human keratinocyte activation and decreases NF-κB induction that is increased in psoriatic skin. Plos One. 2014;9:e90739. [DOI] [PubMed] [PMC]
    An J, Li T, Dong Y, Li Z, Huo J. Terminalia chebulanin attenuates psoriatic skin lesion via regulation of heme oxygenase-1. Cell Physiol Biochem. 2016;39:53143. [DOI] [PubMed]
    Dou R, Liu Z, Yuan X, Xiangfei D, Bai R, Bi Z, et al. PAMs ameliorates the imiquimod-induced psoriasis-like skin disease in mice by inhibition of translocation of NF-κB and production of inflammatory cytokines. PLoS One. 2017;12:e0176823. [DOI] [PubMed] [PMC]
    Villarino AV, Kanno Y, Ferdinand JR, O’Shea JJ. Mechanisms of Jak/STAT signaling in immunity and disease. J Immunol. 2015;194:217. [DOI] [PubMed] [PMC]
    Welsch K, Holstein J, Laurence A, Ghoreschi K. Targeting JAK/STAT signalling in inflammatory skin diseases with small molecule inhibitors. Eur J Immunol. 2017;47:1096107. [DOI] [PubMed]
    Farag AGA, Samaka R, Elshafey EN, Shehata WA, El Sherbiny EG, Hammam MA. Immunohistochemical study of Janus kinase 1/signal transducer and activator of transcription 3 in psoriasis vulgaris. Clin Cosmet Investig Dermatol. 2019;12:497508. [DOI] [PubMed] [PMC]
    Nogueira M, Puig L, Torres T. JAK inhibitors for treatment of psoriasis: focus on selective TYK2 inhibitors. Drugs. 2020;80:34152. [DOI] [PubMed]
    Papp KA, Krueger JG, Feldman SR, Langley RG, Thaci D, Torii H, et al. Tofacitinib, an oral Janus kinase inhibitor, for the treatment of chronic plaque psoriasis: long-term efficacy and safety results from 2 randomized phase-III studies and 1 open-label long-term extension study. J Am Acad Dermatol. 2016;74:84150. [DOI] [PubMed]
    Sung YY, Kim HK. Illicium verum extract suppresses IFN-γ-induced ICAM-1 expression via blockade of JAK/STAT pathway in HaCaT human keratinocytes. J Ethnopharmacol. 2013;149:62632. [DOI] [PubMed]
    Zhang HX, Dang MY, Chen XM, Yan X. Rehmannia radix extract ameliorates imiquimod-induced psoriasis-like skin inflammation in a mouse model via the Janus-kinase signal transducer and activator of transcription pathway. Pharmacogn Mag. 2020;16:6139. [DOI]
    Zhao J, Di T, Wang Y, Liu X, Liang D, Zhang G, et al. Multi-glycoside of Tripterygium wilfordii Hook. f. ameliorates imiquimod-induced skin lesions through a STAT3-dependent mechanism involving the inhibition of Th17-mediated inflammatory responses. Int J Mol Med. 2016;38:74757. [DOI] [PubMed] [PMC]
    Tang L, He S, Wang X, Liu H, Zhu Y, Feng B, et al. Cryptotanshinone reduces psoriatic epidermal hyperplasia via inhibiting the activation of STAT3. Exp Dermatol. 2018;27:26875. [DOI] [PubMed]
    Xu Y, Xu X, Gao X, Chen H, Geng L. Shikonin suppresses IL-17-induced VEGF expression via blockage of JAK2/STAT3 pathway. Int Immunopharmacol. 2014;19:32733. [DOI] [PubMed]
    Yu YJ, Xu YY, Lan XO, Liu XY, Zhang XL, Gao XH, et al. Shikonin induces apoptosis and suppresses growth in keratinocytes via CEBP-δ upregulation. Int Immunopharmacol. 2019;72:51121. [DOI] [PubMed]
    Chen Z, Laurence A, O’Shea JJ. Signal transduction pathways and transcriptional regulation in the control of Th17 differentiation. Semin Immunol. 2007;19:4008. [DOI] [PubMed] [PMC]
    Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, et al. STAT3 regulates cytokine- mediated generation of inflammatory helper T cells. J Biol Chem. 2007;282:935863. [DOI] [PubMed]
    Ghoreschi K, Jesson MI, Li X, Lee JL, Ghosh S, Alsup JW, et al. Modulation of innate and adaptive immune responses by tofacitinib (CP-690,550). J Immunol. 2011;186:423443. [DOI] [PubMed] [PMC]
    Rutz S, Eidenschenk C, Ouyang W. IL-22, not simply a Th17 cytokine. Immunol Rev. 2013;252:11632. [DOI] [PubMed]
    Mitra A, Raychaudhuri SK, Raychaudhuri SP. IL-22 induced cell proliferation is regulated by PI3K/Akt/mTOR signaling cascade. Cytokine. 2012;60:3842. [DOI] [PubMed]
    Lejeune D, Dumoutier L, Constantinescu S, Kruijer W, Schuringa JJ, Renauld JC. Interleukin-22 (IL-22) activates the JAK/STAT, ERK, JNK, and p38 MAP kinase pathways in a rat hepatoma cell line: pathways that are shared with and distinct from IL-10. J Biol Chem. 2002;277:3367682. [DOI] [PubMed]
    Choi M, Yi JK, Kim SY, Ryu JH, Lee J, Kwon W, et al. Anti-inflammatory effects of a methanol extract of Dictamnus dasycarpus Turcz. root bark on imiquimod-induced psoriasis. BMC Complement Altern Med. 2019;19:347. [DOI] [PubMed] [PMC]
    Su Y, Wu L, Mu G, Wang Q, Yang B, Cheng G, et al. 9,19-Cycloartenol glycoside G3 from Cimicifuga simplex regulates immune responses by modulating Th17/Treg ratio. Bioorg Med Chem. 2017;25:491723. [DOI] [PubMed]
    Su Y, Wang Q, Yang B, Wu L, Cheng G, Kuang H. Withasteroid B from D. metel L. regulates immune responses by modulating the JAK/STAT pathway and the IL-17+RORγt+/IL-10+FoxP3+ ratio. Clin Exp Immunol. 2017;190:4053. [DOI] [PubMed] [PMC]
    Li B, He S, Liu R, Huang L, Liu G, Wang R, et al. Total glucosides of paeony attenuates animal psoriasis induced inflammatory response through inhibiting STAT1 and STAT3 phosphorylation. J Ethnopharmacol. 2019;243:112121. [DOI] [PubMed]
    Nguyen LTH, Ahn SH, Nguyen UT, Yang IJ. Dang-Gui-Liu-Huang Tang a traditional herbal formula, ameliorates imiquimod-induced psoriasis-like skin inflammation in mice by inhibiting IL-22 production. Phytomedicine. 2018;47:4857. [DOI] [PubMed]
    Li L, Zhang HY, Zhong XQ, Lu Y, Wei J, Li L, et al. PSORI-CM02 formula alleviates imiquimod-induced psoriasis via affecting macrophage infiltration and polarization. Life Sci. 2020;243:117231. [DOI] [PubMed]
    Yue L, Ailin W, Jinwei Z, Leng L, Jianan W, Li L, et al. PSORI-CM02 ameliorates psoriasis in vivo and in vitro by inducing autophagy via inhibition of the PI3K/Akt/mTOR pathway. Phytomedicine. 2019;64:153054. [DOI] [PubMed]
    Reddy SA, Huang JH, Liao WS. Phosphatidylinositol 3-kinase as a mediator of TNF-induced NF-κB activation. J Immunol. 2000;164:135563. [DOI] [PubMed]
    Schulze-Osthoff K, Ferrari D, Riehemann K, Wesselborg S. Regulation of NF-κB activation by MAP kinase cascades. Immunobiology. 1997;198:3549. [DOI] [PubMed]
    Li W, Khor TO, Xu C, Shen G, Jeong WS, Yu S, et al. Activation of Nrf2-antioxidant signaling attenuates NFκB-inflammatory response and elicits apoptosis. Biochem Pharmacol. 2008;76:14859. [DOI] [PubMed] [PMC]
    Ivashkiv LB. Crosstalk with the Jak-STAT pathway in inflammation. In: Decker T, Müller M, editors. Jak-Stat signaling: from basics to disease. Vienna: Springer; 2012. pp. 35370. [DOI]
    Park SH, Cho G, Park SG. NF-κB activation in T helper 17 cell differentiation. Immune Netw. 2014;14:1420. [DOI] [PubMed] [PMC]
    Goldminz AM, Au SC, Kim N, Gottlieb AB, Lizzul PF. NF-κB: an essential transcription factor in psoriasis. J Dermatol Sci. 2013;69:8994. [DOI] [PubMed]
    Irrera N, Vaccaro M, Bitto A, Pallio G, Pizzino G, Lentini M, et al. BAY 11-7082 inhibits the NF-κB and NLRP3 inflammasome pathways and protects against IMQ-induced psoriasis. Clin Sci (Lond). 2017;131:48798. [DOI] [PubMed]
    Kim BH, Oh I, Kim JH, Jeon JE, Jeon B, Shin J, et al. Anti-inflammatory activity of compounds isolated from Astragalus sinicus L. in cytokine-induced keratinocytes and skin. Exp Mol Med. 2014;46:e87. [DOI] [PubMed] [PMC]
    Kim HK, Bae MJ, Lim S, Lee W, Kim S. A water-soluble extract from Actinidia arguta ameliorates psoriasis-like skin inflammation in mice by inhibition of neutrophil infiltration. Nutrients. 2018;10:1399. [DOI] [PubMed] [PMC]
    Hu J, Yang R, Wen C, Li H, Zhao H. Expression of NLRP3 inflammasome in BALB/c mice with imiquimod-induced psoriasis-like inflammation and therapeutic effect of mustard seed (Sinapis Alba Linn). Nan Fang Yi Ke Da Xue Xue Bao. 2013;33:13948. Chinese. [PubMed]
    Yang BY, Cheng YG, Liu Y, Liu Y, Tan JY, Guan W, et al. Datura metel L. ameliorates imiquimod-induced psoriasis-like dermatitis and inhibits inflammatory cytokines production through TLR7/8–MyD88– NF-κB–NLRP3 inflammasome pathway. Molecules. 2019;24:2157. [DOI] [PubMed] [PMC]
    Yang R, Zhou Q, Wen C, Hu J, Li H, Zhao M, et al. Mustard seed (Sinapis Alba Linn) attenuates imiquimod-induced psoriasiform inflammation of BALB/c mice. J Dermatol. 2013;40:54352. [DOI] [PubMed]
    Li HJ, Wu NL, Pu CM, Hsiao CY, Chang DC, Hung CF. Chrysin alleviates imiquimod-induced psoriasis-like skin inflammation and reduces the release of CCL20 and antimicrobial peptides. Sci Rep. 2020;10:2932. [DOI] [PubMed] [PMC]
    Xiong H, Xu Y, Tan G, Han Y, Tang Z, Xu W, et al. Glycyrrhizin ameliorates imiquimod-induced psoriasis-like skin lesions in BALB/c mice and inhibits TNF-α-induced ICAM-1 expression via NF-κB/MAPK in HaCaT cells. Cell Physiol Biochem. 2015;35:133546. [DOI] [PubMed]
    Lee J, Song K, Hiebert P, Werner S, Kim TG, Kim YS. Tussilagonone ameliorates psoriatic features in keratinocytes and imiquimod-induced psoriasis-like lesions in mice via NRF2 activation. J Invest Dermatol. 2020;140:122332.E4. [DOI] [PubMed]
    Sun J, Han J, Zhao Y, Zhu Q, Hu J. Curcumin induces apoptosis in tumor necrosis factor-alpha-treated HaCaT cells. Int Immunopharmacol. 2012;13:1704. [DOI] [PubMed]
    Kang D, Li B, Luo L, Jiang W, Lu Q, Rong M, et al. Curcumin shows excellent therapeutic effect on psoriasis in mouse model. Biochimie. 2016;123:7380. [DOI] [PubMed]
    Skyvalidas DN, Mavropoulos A, Tsiogkas S, Dardiotis E, Liaskos C, Mamuris Z, et al. Curcumin mediates attenuation of pro-inflammatory interferon γ and interleukin 17 cytokine responses in psoriatic disease, strengthening its role as a dietary immunosuppressant. Nutr Res. 2020;75:95108. [DOI] [PubMed]
    Wen J, Pei H, Wang X, Xie C, Li S, Huang L, et al. Gambogic acid exhibits anti-psoriatic efficacy through inhibition of angiogenesis and inflammation. J Dermatol Sci. 2014;74:24250. [DOI] [PubMed]
    Wen J, Wang X, Pei H, Xie C, Qiu N, Li S, et al. Anti-psoriatic effects of Honokiol through the inhibition of NF-κB and VEGFR-2 in animal model of K14-VEGF transgenic mouse. J Pharmacol Sci. 2015;128:11624. [DOI] [PubMed]
    Liu M, Zhang G, Naqvi S, Zhang F, Kang T, Duan Q, et al. Cytotoxicity of saikosaponin A targets HEKa cell through apoptosis induction by ROS accumulation and inflammation suppression via NF-κB pathway. Int Immunopharmacol. 2020;86:106751. [DOI] [PubMed]
    Han L, Sun J, Lu CJ, Zhao RZ, Lu Y, Lin HJ, et al. Formula PSORI-CM01 inhibits the inflammatory cytokine and chemokine release in keratinocytes via NF-κB expression. Int Immunopharmacol. 2017;44:22633. [DOI] [PubMed]
    Gao Y, Sun W, Cha X, Wang H. ‘Psoriasis 1’ reduces T-lymphocyte-mediated inflammation in patients with psoriasis by inhibiting vitamin D receptor-mediated STAT4 inactivation. Int J Mol Med. 2020;46:153850. [DOI] [PubMed] [PMC]
    Sun W, Gao Y, Yu X, Yuan Y, Yi J, Zhang Z, et al. ‘Psoriasis 1’ reduces psoriasis-like skin inflammation by inhibiting the VDR-mediated nuclear NF-κB and STAT signaling pathways. Mol Med Rep. 2018;18:273343. [DOI] [PubMed] [PMC]
    Lee SY, Nam S, Hong IK, Kim H, Yang H, Cho HJ. Antiproliferation of keratinocytes and alleviation of psoriasis by the ethanol extract of Artemisia capillaris. Phytother Res. 2018;32:92332. [DOI] [PubMed]
    Chung IC, Yuan SN, OuYang CN, Hu SI, Lin HC, Huang KY, et al. EFLA 945 restricts AIM2 inflammasome activation by preventing DNA entry for psoriasis treatment. Cytokine. 2020;127:154951. [DOI] [PubMed]
    Jia J, Mo X, Liu J, Yan F, Wang N, Lin Y, et al. Mechanism of danshensu-induced inhibition of abnormal epidermal proliferation in psoriasis. Eur J Pharmacol. 2020;868:172881. [DOI] [PubMed]
    Shao F, Tan T, Tan Y, Sun Y, Wu X, Xu Q. Andrographolide alleviates imiquimod-induced psoriasis in mice via inducing autophagic proteolysis of MyD88. Biochem Pharmacol. 2016;115:94103. [DOI] [PubMed]
    Yang S, Liu J, Jiao J, Jiao L. Ar-turmerone exerts anti-proliferative and anti-inflammatory activities in HaCaT keratinocytes by inactivating Hedgehog pathway. Inflammation. 2020;43:47886. [DOI] [PubMed]
    Chamcheu JC, Esnault S, Adhami VM, Noll AL, Banang-Mbeumi S, Roy T, et al. Fisetin, a 3,7,3’,4’-tetrahydroxyflavone inhibits the PI3K/Akt/mTOR and MAPK pathways and ameliorates psoriasis pathology in 2D and 3D organotypic human inflammatory skin models. Cells. 2019;8:1089. [DOI] [PubMed] [PMC]
    Tsai YF, Chen CY, Lin IW, Leu YL, Yang SC, Syu YT, et al. Imperatorin alleviates psoriasiform dermatitis by blocking neutrophil respiratory burst, adhesion, and chemotaxis through selective phosphodiesterase 4 inhibition. Antioxid Redox Signal. 2021;35:885903. [DOI] [PubMed]
    Boca AN, Ilies RF, Saccomanno J, Pop R, Vesa S, Tataru AD, et al. Sea buckthorn extract in the treatment of psoriasis. Exp Ther Med. 2019;17:10203. [DOI] [PubMed] [PMC]
    Najafizadeh P, Hashemian F, Mansouri P, Farshi S, Surmaghi MS, Chalangari R. The evaluation of the clinical effect of topical St Johns wort (Hypericum perforatum L.) in plaque type psoriasis vulgaris: a pilot study. Australas J Dermatol. 2012;53:1315. [DOI] [PubMed]
    Cheng HM, Wu YC, Wang Q, Song M, Wu J, Chen D, et al. Clinical efficacy and IL-17 targeting mechanism of Indigo naturalis as a topical agent in moderate psoriasis. BMC Complement Altern Med. 2017;17:439. [DOI] [PubMed] [PMC]
    Rerknimitr P, Nitinawarat J, Weschawalit S, Wititsuwannakul J, Wongtrakul P, Jutiviboonsuk A, et al. The efficacy of Gynura pseudochina DC. var. hispida Thv. ointment in treating chronic plaque psoriasis: a randomized controlled trial. J Altern Complement Med. 2016;22:66975. [DOI] [PubMed]
    Suryakumar G, Gupta A. Medicinal and therapeutic potential of sea buckthorn (Hippophae rhamnoides L.). J Ethnopharmacol. 2011;138:26878. [DOI] [PubMed]
    Yuan ZZ, Yuan X, Xu ZX. Studies on tabellae Indigo naturalis in treatment of psoriasis. J tradit Chin Med. 1982;2:306. [PubMed]
    Feißt C, Albert D, Verotta L, Werz O. Evaluation of hyperforin analogues for inhibition of 5-lipoxygenase. Med Chem. 2005;1:28791. [DOI] [PubMed]
    Lemmens RHMJ, Bunyapraphatsara N editors. Medicinal and poisonous plants 3. Leiden: Backhuys Publishers; 2003.
    Yan Y, Liu W, Andres P, Pernin C, Chantalat L, Briantais P, et al. Exploratory clinical trial to evaluate the efficacy of a topical traditional chinese herbal medicine in psoriasis vulgaris. Evid Based Complement Alternat Med. 2015;2015:719641. [DOI] [PubMed] [PMC]
    Li N, Zhao W, Xing J, Liu J, Zhang G, Zhang Y, et al. Chinese herbal Pulian ointment in treating psoriasis vulgaris of blood-heat syndrome: a multi-center, double-blind, randomized, placebo-controlled trial. BMC Complement Altern Med. 2017;17:264. [DOI] [PubMed] [PMC]
    Mao CL, Wu YY, Zhou DM, Xu WJ, Wang JS. The efficiency and safety of the Chinese herbal medicine liang xue huo xue decoction (LXHXD) in patients with psoriasis vulgaris of blood heat syndrome. Int J Clin Exp Med. 2019;12:60205.
    Dai YJ, Li YY, Zeng HM, Liang XA, Xie ZJ, Zheng ZA, et al. Effect of Yinxieling decoction on PASI, TNF-α and IL-8 in patients with psoriasis vulgaris. Asian Pac J Trop Med. 2014;7:66870. [DOI] [PubMed]
    Sun L, Deng B, Wang H, Chen K. Influence of Liang-Xue-Huo-Xue (LXHX) capsule on apoptosis of cultured keratinocytes. Chin J Dermatol. 2003;36:5835.
    Lu CJ, Xiang Y, Xie XL, Xuan ML, He ZH. A randomized controlled single-blind clinical trial on 84 outpatients with psoriasis vulgaris by auricular therapy combined with optimized Yinxieling Formula. Chin J Integr Med. 2012;18:18691. [DOI] [PubMed]
    Sun L, Li T, Zhou D, Yang X, Tian J, Zhao J, et al. “Efficacy and safety of Liangxue Jiedu decoction for the treatment of progressive psoriasis vulgaris: a multicenter, randomized, controlled study”. J Tradit Chin Med. 2020;40:296304. [PubMed]
    Qiu S, Tan S, Zhang J, Liu P, Ran L, Lei X. Effect of liangxue huoxue xiaoyin tang on serum levels of TNF-alpha, IFN-gamma and IL-6 in psoriasis of blood-heat type. J Tradit Chin Med. 2005;25:2925. [PubMed]
    Zhang GZ, Wang JS, Wang P, Jiang CY, Deng BX, Li P, et al. Distribution and development of the TCM syndromes in psoriasis vulgaris. J Tradit Chin Med. 2009;29:195200. [DOI] [PubMed]
    Li X, Xiao QQ, Li FL, Xu R, Fan B, Wu MF, et al. Immune signatures in patients with psoriasis vulgaris of blood-heat syndrome: a systematic review and meta-analysis. Evid Based Complement Alternat Med. 2016;2016:9503652. [DOI] [PubMed] [PMC]
    Ramírez-Boscá A, Navarro-López V, Carrión-Gutiérrez M, Martínez-Andrés A, Vilata-Corell JJ, Asín-Llorca M, et al. Efficiency and safety of a Curcuma extract combined with visible blue light phototherapy on adults with plaque psoriasis: a phase IV, randomized, open pilot clinical trial. J Dermatol. 2017;44:11778. [DOI] [PubMed]
    Yu C, Fan X, Li Z, Liu X, Wang G. Efficacy and safety of total glucosides of paeony combined with acitretin in the treatment of moderate-to-severe plaque psoriasis: a double-blind, randomised, placebo-controlled trial. Eur J Dermatol. 2017;27:1504. [DOI] [PubMed]
    Lee SR, Kim S, Park CE, Lee JH, Lee DH. Effect of Korean medicine as add-on therapy to phototherapy for psoriasis: two case reports. Medicine. 2019;98:e14526. [DOI] [PubMed] [PMC]
    Zorko MS, Štrukelj B, Švajger U, Kreft S, Lunder T. Efficacy of a polyphenolic extract from silver fir (Abies alba) bark on psoriasis: a randomised, double-blind, placebo-controlled trial. Pharmazie. 2018;73:5660. [DOI] [PubMed]
    Wu C, Jin HZ, Shu D, Li F, He CX, Qiao J, et al. Efficacy and safety of Tripterygium wilfordii Hook f versus acitretin in moderate to severe psoriasis vulgaris: a randomized clinical trial. Chin Med J (Engl). 2015;128:4439. [DOI] [PubMed] [PMC]
    Ho SGY, Yeung CK, Chan HHL. Methotrexate versus traditional Chinese medicine in psoriasis: a randomized, placebo-controlled trial to determine efficacy, safety and quality of life. Clin Exp Dermatol. 2010;35:71722. [DOI] [PubMed]
    van der Fits L, Mourits S, Voerman JS, Kant M, Boon L, Laman JD, et al. Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J Immunol. 2009;182:583645. [DOI] [PubMed]
    Dellambra E, Odorisio T, D’Arcangelo D, Failla CM, Facchiano A. Non-animal models in dermatological research. ALTEX. 2019;36:177202. [DOI] [PubMed]