Skip to main content

Advances in the modulation of ROS and transdermal administration for anti-psoriatic nanotherapies

Abstract

Reactive oxygen species (ROS) at supraphysiological concentration have a determinate role in contributing to immuno-metabolic disorders in the epithelial immune microenvironment (EIME) of psoriatic lesions. With an exclusive focus on the gene-oxidative stress environment interaction in the EIME, a comprehensive strategy based on ROS-regulating nanomedicines is greatly anticipated to become the mainstay of anti-psoriasis treatment. This potential therapeutic modality could inhibit the acceleration of psoriasis via remodeling the redox equilibrium and reshaping the EIME. Herein, we present a marked overview of the current progress in the pathomechanisms of psoriasis, with particular concerns on the potential pathogenic role of ROS, which significantly dysregulates redox metabolism of keratinocytes (KCs) and skin-resident or -infiltrating cells. Meanwhile, the emergence of versatile nanomaterial-guided evolution for transdermal drug delivery has been attractive for the percutaneous administration of antipsoriatic therapies in recent years. We emphasize the underlying molecular mechanism of ROS-based nanoreactors for improved therapeutic outcomes against psoriasis and summarize up-to-date progress relating to the advantages and limitations of nanotherapeutic application for transdermal administration, as well as update an insight into potential future directions for nanotherapies in ROS-related skin diseases.

Graphical Abstract

Introduction

Psoriasis (Ps) is a multifaceted disease related to chronic dysimmunity and genetic disease, which manifests in skin symptoms of demarcated erythematous and scaly lesions, accompanied by other systemic inflammatory comorbidities, like psychological illness, metabolic disturbance, arthritis, and cardiovascular disorders [1]. It has been affecting appropriately 125 million people worldwide [2, 3], in which the age group of 60–69 years is recognized as a weighty psoriasis burden according to the Global Burden of Disease (GBD) 2019 study [4, 5]. According to the clinical features, psoriasis is classified into cutaneous psoriasis and systemic psoriasis. Among the variants in cutaneous psoriasis, plaque psoriasis, also known as psoriasis vulgaris, is the most common phenotype, affecting 85–90% of patients with psoriasis [6]. The histopathological feature of psoriatic lesions is parakeratosis in the thickened stratum corneum, the remarkably thickened epidermis with elongations into the dermis, and an abundance of different immune cells from dermis infiltration into the epidermis. Numerous studies have currently revealed that the direct or indirect cross-talking among different cell types in epithelial immune niches, plays a vital role in the pathogenesis of psoriasis and predominately emphasized the trigger role of oxidative stress in these cell types dysfunctions. Oxidative metabolites, namely reactive species, such as ROS/RNS, including superoxide anion hydroxyl radical (•OH), radical (•O2), hydrogen peroxide (H2O2), singlet molecular oxygen (1O2), as well as nitric oxide, hydrogen sulfide, and oxidized lipids, prominently originates from mitochondrial electron transport chain (ETC), NADPH oxidases, other oxidases like peroxisome, several superoxide dismutases (SOD1–SOD3) and so on [7,8,9,10]. The physiological concentration of reactive species is significant to orchestrate cellular redox signaling and guarantee diverse normal cell processes. Inversely, the supraphysiological level of these metabolites has the opposite pleiotropy. Therefore, it is imperative to deeply understand the role of detrimental ROS in the dyshomeostasis of keratinocytes (KCs) and immune cells in the epithelial immune microenvironment (EIME), ultimately leading to the generation and perpetuation of the inflamed cascade reaction in psoriasis [7].

The conventional medications for psoriasis such as corticosteroids, vitamin D derivatives, targeting biologics, folic acid antagonists and calcineurin inhibitors are failing far to fulfill the current clinical need due to the systemic adverse reaction and the lower drug penetration [11, 12]. Over the past decades, we have witnessed great success in medical nanomaterial, which has provided more and more nano-drugs and possible solutions for transdermal administration to improve psoriasis. The application of biomaterials to locally deliver conventional medications for psoriasis therapy can achieve an enhanced local drug concentration and circumvent system adverse reactions. Among the various nanotechnologies, several nanomaterials, e.g., microneedle and hydrogel, have demonstrated to be promising in clinical applications which are already on the market. In this review, we stay organized around the following two topics: firstly, we review how specific ROS perturbs and reprograms redox signaling pathways in KCs and immune cells, as well as provide a comprehensive understanding value of ROS as a promising therapeutic target for the applications in the treatment of psoriasis. In the end, we summarize the state-of-the-art ROS-regulating nano-medicines and nanomaterial-based therapies with distinctive transdermal delivery features used in anti-psoriatic therapies.

Oxidative stress and its roles in different cell types dysfunctions of psoriasis

As the outermost immune and barrier organ of the human body, the skin is most vulnerable to be attacked by external insults, such as pathogen, toxication, pollution, trauma, UV rays, etc., concomitantly with an increased baleful ROS, consequently disturbing cutaneous defense mechanism and priming skin immune responses maintained by EIME [13], which is composed of cellular communications among KCs, skin-resident and skin-infiltrating immune cells via interactions with a gradient of various chemo-attractants, such as chemokines, cytokines, vesicles and exosomes in the epidermis and papillary dermis [13, 14], as shown as in Fig. 1. In the past decades, a dramatic increase in the numbers of evidence has highlighted that turbulence of EIME evokes the initiation and chronic inflammation in dermatoses significantly associated with oxidative stress [15,16,17]. In addition to direct skin abnormality, systemic-based perturbations of metabolome also have appreciable effects on the pathogenesis of psoriatic inflammation [18]. As the pathogenic roles of increased oxidative stress, proinflammatory cytokines, adipocytokines, endoplasmic reticulum (ER) stress unbalance, and gut microbiota dysbiosis in the development of psoriasis with metabolic comorbidities are decoded, thus evaluating the metabolite profiles of psoriasis contributes to indicating biomarkers or novel therapeutic targets for prognosis and monitor response to the treatment [6, 19]. What’s more, numerous discoveries exploring the pathogenic mechanism of psoriasis have shed light on intricately interwoven effects among keratinocyte, innate and adaptive immune cells to form clusters termed inducible skin-associated lymphoid tissue (iSALT) [20,21,22,23,24] in the pathophysiological EIME of cutaneous inflammation, especially in psoriasis [14, 25]. Deleterious reactive metabolites like ROS have a harmful role in inducing DNA mutations, epigenetic alterations, post-translational modifications of protein kinase (cysteine residues)[10], lipid peroxidation, and other key cellular components irreversible damage to these cells, thereby reprogramming their metabolic pathways of development, proliferation, activation and function, ultimately giving rise to psoriasis [15, 26, 27]. Therefore, disturbances in the oxidant-antioxidant system of the skin bring a dominant role in the pathogenesis of psoriasis [28], and keeping the dynamic equilibrium of the redox system is the most significant factor to sustain a myriad of normal biological processes in cells of EIME. Intracellular sophisticated antioxidative systems can counteract oxidative stress-induced ROS compounds, maintain redox homeostasis with a physiological threshold of ROS, and protect cells from an oxidative stress injury. The antioxidant capacity of the various skin cells is armed with the main cellular antioxidant pathways, including the main components of glutathione (GSH) pathways [29] and transcriptional regulator NF-E2-related factor 2 (NRF2) [29,30,31], which translocate to the nucleus and binds to DNA promoters to initiate transcription of many antioxidant genes and cytoprotective proteins, to balance the level of oxidative metabolites, as shown as Fig. 2. Hence, we elucidate the focus role of ROS and molecular mechanism in skin KCs and immune-resident or –infiltrating cells under psoriasis conditions.

Fig. 1
figure 1

Dysfunctional different cell types (KCs, skin-resident and -infiltrating immune cells function) mediate the propagation of inflammatory loops in EIME of psoriasis: turbulence of EIME evokes the initiation and chronic inflammation in psoriasis significantly associated with oxidative stress. Deleterious reactive metabolites ROS have a harmful role in inducing irreversible damage to these cells in EIME, thereby reprogramming their metabolic pathways involved in the development, proliferation, activation and function. Subsequently, intricately interwoven effects among these cells form clusters of inflammatory circuits in the pathophysiological EIME of cutaneous inflammation, ultimately giving rise to psoriasis

Oxidative stress-induced pathological signaling in KCs

It is admitted that KCs as amplifiers contribute to cell-mediated psoriatic IL-23/IL-17 axis inflammation cascade effect in psoriasis. That is, cytokines, derived from IL23/IL-17 axis, induce ROS accumulation and cause redox dyshomeostasis of KCs, resulting in impairing the proliferation, differentiation and function of KCs via dysregulating phosphorylation/dephosphorylation key transcription factors and signal transductions in these cells, including NF-κB, STAT3, and others [30, 32, 33]. These “activated” KCs exert a core pathogenic role in the cytokine-mediated various inflammation cascades [34,35,36], not merely serving as immune response triggers but also as proinflammatory non-immune cell effectors, which are capable of amplifying cytokine signal pathways from innate and adaptive immune cells to create a self-perpetuating autoimmune cytokine loop further so that persisting inward recruitment of leukocytes subsets into psoriatic lesions [37,38,39], e.g., macrophages, neutrophils, myeloid DCs and T subsets. Young CN et al. found that the crucial psoriatic cytokine TNF-α could stimulate the activation of the mTOR-NF-κB pathway by ROS generation and ultimately production of inflammatory cytokines in KCs to initiate and maintain the progression of psoriasis; these ROS-induced cytokines could be attenuated by antioxidant enzyme and catalase, including taurine and N-acetyl-cysteine [28]. Besides, rapamycin, an inhibitor of mTOR, could exert antiproliferative properties in the imiquimod (IMQ)-induced mice psoriasis via activating NRF2 signaling and restraining NOX2/4 from decreasing ROS generation [40]. Likewise, inhibiting the activity of NOX1/NOX4 in KCs could abrogate detrimental oxidative stress and rescue high levels of signature cytokines in a 2D model of atopic dermatitis and psoriasis [16]. CHF6001, a PD4 inhibitor, was reported to repress ROS through inactivating p47 (a subunit of the NOX complex 1) and then inhibit translocation of phosphorylated NF-κB, promoting the loss of cyclin D1 to alleviate redox-inflammatory crosstalk of psoriasis [41]. Apart from NOX isoforms, dual oxidase 2 (DUOX2) homologues can also generate ROS. A study reported by Nadeem A et al. had shown that GPR43 agonists could activate epidermal GPR43-mediated DUOX2 and IL-6 signaling pathways to give rise to pernicious ROS, leading to Th17 polarization immune responses and deterioration of psoriasis [42]. Besides, Kumari S et al. uncovered that TNF-α induced the ROS-ERK pathway-dependent upregulation of IL-24 and activation of STAT3 signaling in stressed KCs upon KCs stimulated by endogenous and exogenous insults [36]. STAT3, as an essential transcription factor, leads to the production of many cytokines in inflammatory processes of KCs [43, 44], which in turn not only have an impact on disturbing the oxidant-antioxidant system but also recruiting a more deal of immune cells into the skin lesions to perpetuate a positive feedback inflammatory loop and remodeling extracellular matrix [28, 36]. Supraphysiological level of ROS makes the KCs be the state of ‘oxidative distress’, which can induce the generation or modification of functional reductant protein networks under regulating the redox signaling pathways, as mentioned already, to control ROS production and availability [7]. Among them, SIRT1, as a NAD-dependent deacetylase, plays a salient role in regulating the cellular pathological process of oxidative stress and autoimmune inflammation[17, 32, 45, 46]. In psoriasis, SIRT1 has been reported as a vital detoxifier of ROS-mediated redox signaling pathways, including MAPK, NF-κB, and STAT3, with downregulation of psoriatic inflammatory cytokines, suppression of keratinocyte hyperproliferation, and inhibition of angiogenesis [32, 46,47,48,49,50]. In addition, IL6/IL22-induced STAT3 activation in KCs was controlled by HO-1 induction and activation of protein tyrosine phosphatase SHP-1, accompanied by reduction of KCs hyperproliferation [51].

Similarly, the KEAP1/NRF2 system, as cytoprotective and antioxidative gene transcription, is critical in the redox signaling pathway with a core role in regulating inflammation, maintenance of epidermal differentiation and keratinization in response to ROS challenge [52, 53]. The accumulated research has shown that a significant increase in detrimental ROS impairs the well-balanced cellular redox signaling pathways. It generates harmful protein oxidation products, leading to cell dysfunction and disease initiation. The expression of NRF2 is reduced and its downstream regulatory genes in psoriatic skin tissues. In the IMQ-induced psoriasis-like mice model, NRF2/HO-1 in the skin lesion was decreased. The accumulation of excessive ROS activated the NF-κB pathway, concomitantly with the secretion of proinflammatory cytokines IL-17, IL-23, IL-1β and VEGF expression [54, 55]. The reduction of other prototypical examples of redox signaling-mediated antioxidative enzymes is also involved in the pathomechanism of psoriasis, such as GSH, Px, CAT, and SOD [56, 57]. In addition, several aquaporins (AQP3, AQP8 and AQP9), referred to as ‘peroxiporins’, facilitate the transportation of H2O2 across cellular membranes to regulate downstream intracellular signalings [58, 59]. The study of Hara-Chikuma M et al. demonstrated that AQP3-facilitated H2O2 transport was the precondition of NF-κB activation in KCs participating in the acceleration of psoriasis; In AQP3 knockout mice AQP3 (-/-), IL-23-mediated psoriasiform skin inflammation was reduced [58]. Taken together, the abovementioned studies of dysfunctional KCs suggest that oxidative stress-related signaling pathways make a difference in the pathogenesis of psoriasis, and it is worthy of decreasing cytokines gene expression and obstructing the autoimmune loop for the treatment of psoriasis effectively via quenching generation and traffic of triggers-induced pernicious ROS with ROS-depletion or -blockade approaches.

Oxidative stress-mediated abnormal immunometabolism in immune cells of psoriasis

The role of oxidative stress in macrophage dysfunction

It is well established that macrophages derived from monocytes lineage cells are the main component cells of innate immunity. Most human and animal studies have emphasized the crucial role of macrophages in the pathogenesis of psoriasis [60,61,62]. ROS/RNS contributes to rearranging macrophage differentiation and exerting their effector functions in response to tissue environments via intermediating the main cellular oxidation-reduction (redox) pathways, including glutathione (GSH) pathways, and NF-E2-related factor 2 (NRF2) [30, 63, 64]. Myeloid-derived suppressor cells (MDSCs) have been demonstrated involved in the progress of psoriasis. GSH synthesis in MDSCs isolated from the bone marrow of IMQ-induced psoriatic mice model with ROS accumulation was reduced, resulting in interruption of MDSCs differentiation into CD11c+MHC II+ dendritic cells and CD206+ M2 macrophages to exacerbate skin inflammation [65]. In murine macrophages, LPS/IMQ could induce ROS/RNS-NF-κB/ERK/JNK signaling pathway and decrease the expression of NRF2, increasing iNOS and other inflammatory cytokines to exacerbate psoriasiform skin inflammation [66]. It is admitted that the major endogenous enzymatic sources of O2 and H2O2 are transmembrane NADPH oxidases and NADPH oxidase 2 complexes (NOX2) complex-generated ROS can participate in regulating the metabolism and oxidation-reduction signaling pathways of macrophages and neutrophils involved in chronic inflammation, such as mannan-induced Ps and PsA (MIP), rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) [7]. Zhong J et al. demonstrated that Nos2-derived NO modulated the pathogenic IL-1α secretion from the local macrophages, which acted to downstream target innate lymphoid cell 3 (ILC3), resulting in the up-regulation of IL-17 A to trigger and accelerate the development of MIP [67]. Moreover, mitochondria are also the source of cellular ROS [68]. Once the antioxidant defense mechanism is compromised, the aggravation of mitochondrial malfunction-induced ROS could provoke the onset of chronic inflammatory diseases [69, 70]. Mitochondrial ROS is capable of NLRP3 inflammasome activation [64, 71], which is a crucial reactor to trigger innate immune defenses by maturing proinflammatory cytokines such as interleukin (IL-1β and IL-18) [71, 72]. In the peripheral blood of untreated patients with psoriasis, the expression levels of inflammasome sensors, IL-1β and IL-18 were enhanced; Verma D et al. demonstrated that TNF-α upregulated pro-IL-1β and pro-IL18 and stimulated these inflammasome activities via increasing ROS and activation of NLRP3 signaling pathways [73]. A previous study reported that administration of propranolol (the nonselective beta-blocker) was relevant with exacerbation of psoriasis, ascribed to inhibition of autophagic flux, with an abundance of ROS-producing mitochondria in cutaneous LCs, leading to IL23A production [74]. Additionally, HO-1, considered an antioxidative enzyme, is responsible for cytoprotective molecules against oxidative damage and inflammation. Recent shreds of evidence have mentioned that drugs with the property of increased HO-1 expression are protective in animal models of psoriasis, such as curcumin, carnosol, DMF and hemin [54, 75, 76]. Elevated HO-1 expression could attenuate psoriasiform inflammation via inhibiting iNOS in macrophages and maintaining DCs immune tolerance phenotypes [70, 75, 77]. Oppositely, some conflict data suggested that variation of HO-1 system expression in macrophages not only presented beneficial roles, but detrimental outcomes in other diseases, such as cancer and infection [78, 79]. Based on the abovementioned research, it should be realized that macrophages, as the main effector components of innate immunity, are activated by intrinsically and extrinsically oxidative stress through tissue-specific signals to promote the secretion of disease context-specific cytokines [80,81,82]. Therefore, the treatment of unspecific antioxidants could alleviate disease depending on the situation of specific pathogenesis. Furthermore, a full elucidation of oxidative stress in the pathogenesis and progression mechanisms of disease-specific is a precondition for their use as therapeutic antioxidants in medical applications. In psoriasis, proinflammatory macrophages are essential contributors to the pathophysiological inflammatory cascade by forming immunological clusters termed inducible skin-associated lymphoid tissue (iSALT) in the dermis of cutaneous inflammation [14, 23,24,25, 83], which is indispensable for elicitation of adaptive immunity and ultimately orchestrated immune-related signal pathways in KCs, causing a switch into keratinocyte hyperplasia and aberrant differentiation in chronic psoriasiform skin inflammation [61]. Thus, inhibition of the proinflammatory phenotypes of macrophages could be of therapeutic benefit in the psoriasis context. Emerging selective targets against oxidative stress of macrophages and skin inflammation in dermatologic diseases are given by the above multiple specific ROS-mediated signaling pathways and offer a perspective for better-refined redox medicine.

The role of oxidative stress in neutrophil dysfunction

Psoriasis has a wide range of clinical subtypes, which are determined by complicated fine-tuning of innate and adaptive immune responses [43]. General pustular psoriasis (GPP) is an acute and severe systemic inflammation characterized by neutrophilic-rich dysfunction, leading to sterile pustules in skin lesions. It was triggered by neutrophil extracellular traps (NETs) formation (termed as NETosis, a cell death process), which is implicated in autoimmune inflammatory reactions and induced by neutrophil activation and respiratory burst, to release the non-specific effects of CitH3, enzymatic proteins (like neutrophil elastase and MPO), cytosolic proteins (such as S100 calcium-binding proteins) and recruit pro-inflammatory immune cells [84,85,86,87]. The process of NETosis mediated by reactive oxygen species (ROS)-derived from mitochondria and NADPH oxidase could induce autoantibody production, resulting in uncontrolled inflammatory response and tissue pathology [88]. In the onset of psoriasis, KCs are attacked and stressed upon various stimuli, such as trauma, drugs, and infections, followed by the release of damaged DNA/RNA, LL-37, AMPs, DAMPs and other cytokines/chemokines from these activated KCs, which could initiate innate immune responses and attract more neutrophils infiltration into the epidermis to form Munro or Kogoj abscesses, this sterile pustules constitutes typical pathological manifestations of GPP. Meanwhile, these stressed neutrophils produce weblike NETs under ROS-induced respiratory burst, and the release of MPO, elastase and hydrolase from NETs are known to transform inactive precursors of the IL-1β and IL-36 family released from KCs into more biologically active mature bodies, leading to the characteristic pro-inflammatory imbalance of the IL-36 autocrine and autoinflammatory circuits in generalized pustular psoriasis [87, 89, 90]. In the meantime, activated neutrophils secrete psoriatic cytokines such as IL-17 A and IFN-γ members, which could aggravate the self-perpetuating autoimmune cytokine loop in KCs so that persisting inward recruitment of leukocytes subsets into psoriatic lesions and promotion of KCs proliferation [33,34,35]. There is mounting evidence of NETs formation at obvious risk of autoimmune diseases, an inflammatory neutrophil subset with characteristics of aged CD10negCD16lowCD11blow neutrophils appeared in lesional skin and circulation of psoriasis and these aged neutrophils increased IL-17 expression by T cells in a NETosis-dependent way [91]; immature CD10negCD16negCD11bneg/low neutrophils from patients detected a higher ROS level under TNF-α plus f-MLF stimulation as compared with those of healthy controls [91]. Noting that the enzyme MPO is induced by exposure of neutrophils to various forms of oxidative stress, which is one of the important markers of NETosis [87]. this pro-oxidative and pro-inflammatory hemeprotein is recognized to provide a preponderant role in NETs formation; MPO-deficient neutrophils from MPO-deficient individuals cumulatively associated with GPP, the formation of NETs was predominately reduced compared to healthy donors [90]. Similarly, serum MPO levels displayed a significant increase and caused the injury of antioxidative defenses in psoriasis children [92]. Notably, in the IMQ-induced psoriatic mouse model, levels of MPO and oxidative stress were also upregulated [93]. In combination, these accumulations of evidence supported that redox imbalance between oxidant–antioxidants occurred very early in neutrophils, thereby oxidative burst, activation and degranulation of neutrophils involved in the process of NETosis, which implicated in the prolonged persistence of neutrophils in the affected psoriatic individuals and the inability of resolvable inflammation. Conclusively, these data implicate that detrimental ROS contributes to the induction of NETs and the application of ROS-elimination drugs could restore the potential occurrence of NETs formation, thereby shifting the balance to predominant anti-inflammatory signals to counteracting many neutrophil-mediated diseases, in particular GPP. Therefore, targeted NETs degradation biological treatment may be conducive to the containment of sustained neutrophil-mediated skin inflammation.

The role of oxidative stress in DC dysfunction

Much substantial evidence from clinical studies and experimental models has emphasized the critical role of DCs in the pathogenesis of autoimmune diseases, especially psoriasis [94]. The aberrant hyperactivation of DCs could bridge the innate and adaptive immune responses, sufficient to induce psoriasis. it is well appreciated that the cellular immunometabolism changes and redox signaling pathways of immune cells are tightly interwoven and interdependent to regulate their differentiation, proliferation and function [30]. Mizuguchi S et al. unveiled that in a psoriatic mouse model, the suppression of mtROS attenuated the exacerbation of IMQ stimulation psoriasiform dermatitis and IMQ-induced DC activation in vitro was suppressed by inhibition of the generation of mtROS [95]. A similar result, reported by Al-Harbi NO et al. that activation of BTK signaling in CD11c+ DCs upregulated oxidative stress, associated with significant elevation of inflammatory mediators, which are crucial factors in the pathogenesis of IMQ-induced psoriasis-like inflammation in mice [96]. Asides from these data, the cellular redox disequilibrium of DCs could adversely affect their ability to induce activation of T-cells and regulate the polarity of the immune response via glutathione depletion interfering in DC maturation and IL-12 production [97]. As a consequence, these advances suggest that ROS homeostasis is inseparable from maintaining the well-balanced cellular immunometabolism of DCs. Potential therapeutic strategies by neutralizing the excess of ROS could open up new insight into prevention in psoriasis.

The role of oxidative stress in T cell dysfunction

The pivotal role of T cells in the pathogenesis of psoriasis is evidenced by substantial studies. Dysfunctional different T cells subpopulations and their associated cytokines are crucially involved in the onset or exacerbation of psoriasis, and blockade of these cytokine-mediated inflammations could be identified as potential therapeutic targets. Strikingly, dynamic cellular redox reactions are obbligato for ensuring and regulating the homeostatic maintenance of different T cells subsets differentiation and cellular functions. The disruption of redox homeostasis in T cell subsets provides susceptibility to numerous immunopathies [30, 98]. Esmaeili B et al. demonstrated antioxidant defense mechanisms were disordered by elevated ROS in stimulated memory CD4+ T cells from psoriasis patients [99]. It is well-known that regulatory T cells (Tregs) are regarded as protect effect on preventing psoriasis, and excessive ROS would reduce the ratio of Treg: Th17 cells by promoting the proliferation and differentiation of pro-inflammatory Th17/Th1/Th22 cells and reversely suppression of the frequency of Treg to sustain the process of psoriasis [100, 101]. Furthermore, detrimental cellular ROS-induced oxidized 8-oxo-dGTP and DNA also could amplify Th17 subset cells, along with striking elevation of IL-17-producing γδ T cells in lymph nodes [102]. Considering the essential role of the dermal IL-17-producing γδ T cells in psoriasis, its redox regulation engaged in immunometabolism gains more attention as the pivotal player in developing psoriasis [103]. Recent advances demonstrated that mTORC2 constrained mitoROS production in γδ T cells, causing impairment of γδ T17 differentiation, which is critical innate dermal predominate IL-17-producing cells in the development and aggravation of psoriasis [104]. These previous researches make us conscious that more efforts should be paid to comprehensively decipher the definite role of ROS mediated in metabolic rewiring and impaired functions of T cells in disease-specific pathogenesis. It conduces accelerating the discovery of more advanced treatment modalities to restore the balance of ROS levels in T cells for combating autoimmune diseases, particularly psoriasis.

The role of oxidative stress in other immune cells dysfunction

Similar to what is discovered in the abovementioned immune cells involved in the occurrence of psoriasis, extensive research has been performed to detail the crucial role of skin-resident ILCs-associated cytokines IL-17 and IL-22 in contributing to driving dermal inflammation, particularly in psoriasis [105, 106]. ILCs belong to a family of innate immune cells lacking antigen-specific receptors and are classified into three subgroups (ILC1, ILC2, and ILC3) according to their key transcription factors expression and cytokines production [106, 107]. Among them, type 3 ILCs (ILC3s) play a central role in the etiology and disease severity of psoriasis, which was ascribed to the elevated number of IL-22- and IL-17 A/F-producing ILC3s induced by their expression of RORγt transcription factors in psoriatic skin and blood [106, 108,109,110]. RORγt+ ILC and γδ T cells are also prerequisites for driving psoriasiform plaque formation in the IMQ-induced disease models through the aggregation delivery of IL-17 A, IL-17 F, and IL-22 into the skin inflammation [111]. Similar to the immunometablism of other immune cells, ILC plasticity could be supervised by redox metabolic pathways and cytokine milieu. The deficiency of NOX2 shifted Tbet+ ILC1s transdifferentiation into RORγt+ ILC3s in a redox-dependent manner through IL-1β production and aggravated the inflamed joints of Ncf1−/− mice [112]. Likewise, one study also found that Nos2-derived NO upregulated IL-17-producing ILC3 by IL-1α stimulation from the local macrophages participated in triggering and progressing the development of MIP. In addition to the better-studied pathogenesis of ILCs in psoriasis, contributions of NK cell-mediated innate immune responses to inflammatory skin diseases, especially psoriasis, have shown increasingly emerging [113,114,115]. Different subsets of NK cells take part in dysregulating the imbalance of immune response to many autoimmune diseases through the induction of their cytokines and cytotoxic functions [116]. A study reported by Gilhar A et al. illuminated that NK and NKT cells from autologous human lymphocytes were injected into nonlesional skin grafts from psoriatic patients on mice could give rise to representative psoriatic skin inflammation with the expression of inflammatory epidermis signatures [117]. Besides, NKT cells with IFN-γ/CCR5 expression in psoriatic skin were relevant to the severity of psoriasiform hyperplasia and microabscess [118]. Certainly, analogous to the effect of redox-associated metabolic pathways on ILC development and function, the probabilities of NK cell-fate transitions at different stages are also shifted upon autophagy perturbations-inducing ROS disequilibrium [119]. The excessive ROS production under the condition of disrupting dysfunctional mitochondria elimination caused by the deletion of Atg5 or Atg7, severely compromised homeostasis and the maturity of NK cells. Additionally, progressive research in mast cells (MCs) enables satisfactory characterization of cells and their delicate roles in the complex network of psoriasis. Gaudenzio N et al. reported that IFN-γ-primed human MCs caused abundant immunologic synapses with CD4+ T cells, concomitantly with an enhancement of the production of Th22 and IL-22/IFN-γ-producing Th cells from the circulating memory CD4+ T-cell pool; a productive infiltration of IL-22+CD4+ T cells observed in contact with mast cells in human psoriatic skin biopsies [120]. Strikingly, the proportion of IL-22-producing mast cells occupied 20–80% in patients with psoriasis, and skin mast cells expressed IL-22 and IL-17 mRNA [121]. Furthermore, IL-24 from activated T cell-derived microvesicles motivated MCs and excessive MCs activation in psoriasis could produce IL-24, subsequently provoking STAT3 phosphorylation of KCs [122, 123].

Fig. 2
figure 2

ROS contributes to the rearranging immunometablism of different cell types, accompanied by exerting their effector functions in response to tissue environments via intermediating the main cellular oxidation-reduction (redox) signaling pathways

Advances in understanding MCs activation and degranulation have shown that the role of mitochondrial translocation and ROS involved in activating MCs of allergic inflammatory diseases is overwhelming [124,125,126,127]. Skin biopsies from AD revealed that mitochondrial translocation was present in the degranulation and TNF secretion of human skin mast cells [125]. However, the causal relationship between ROS-stimulated MCs activation and psoriasis is needed to be done to expand our basic knowledge. Overall, a disordered oxidant-antioxidant system, in combination with the turbulence of cellular ROS homeostasis from enhanced activation of redox signaling pathways, renders the disturbed immunometablism of immune cells particularly vulnerable to trigger and exacerbation of psoriasis. Comprehensively studying the pathophysiological role played by ROS in the abovementioned immune cells related to the pathogenesis of psoriasis would help to design potential dysfunctional effector cells-targeted anti-inflammatory and anti-psoriatic drugs.

Therapeutic drugs targeting oxidative stress in EIME of psoriasis

To date, the therapeutic efficacies of various agents depend on how well these cycles of inflammation mediated by the abovementioned dysfunctional cells in EIMEs of psoriasis are broken [38]. In consideration of the aforementioned multi-faceted influences of oxidative stress present in the dysfunctional different cell types in EIME of psoriatic inflammation (summarized in Table 1), considerable research has demonstrated disorganized cellular redox signaling pathways in these dysfunctional cells whose induced multiple inflammatory networks could be sophisticatedly modulated and blocked by a variety of chemical agents or drugs. As shown in Table 2, DMF has been previously reported as a broad-spectrum anti-inflammatory drug. It could be used to treat psoriasis via modulating the phenotypic switch of immune cell types through glutathione depletion and reprogramming the cellular redox balance, particularly the modulation of macrophages and type II dendritic cells [76, 128]. Alongside these mechanisms, DMF could also impair NETs formation in polymorphonuclear granulocytes isolated from psoriasis patients via limiting oxidative burst capacity, mediated by depletion of intracellular GSH levels [129]. Building on a study reporting that DMF could cause short-term oxidative stress and activate the antioxidant signaling response of transcription factor NRF2, increasing the antioxidant protein expression and modulating cellular redox state to alter the expression of key genes or proteins related to calcium signaling of immune cell activation [128]. In type II DCs, DMF performed its therapeutic effect via inducing glutathione (GSH) depletion of DCs, followed by increasing the expression of antioxidant hemoxygenase-1 (HO-1) gene and impaired phosphorylation of STAT1 to ameliorate psoriasis and MS (Multiple Sclerosis) [76]. CBD (Cannabidiol), as a wide spectrum of antioxidant and anti-inflammatory modulators, is studied for application in preventing and treating redox imbalance and inflammation-associated diseases [130,131,132]. Indeed, CBD could be considered a potential anti-NETotic factor to inhibit NETosis formation by reducing NADPH oxidase and MPO expression [87]. Ibrutinib, a BTK inhibitor, could ameliorate psoriasiform inflammation by attenuating ROS and inflammatory mediators in CD11c+ DCs [96]. Apremilast, a PDE4 inhibitor, improvement of intracellular cAMP, could augment IL-10-producing Bregs and its concomitant decrease in Th1 cells, IFNγ-producing NKT cells and IL-17-producing NKT cells and suppress IFNγ+CD3+ T cells and IL-17+CD3+ T cells for combating PsA and Ps [133,134,135,136]. Other natural immunomodulatory compounds, such as curcumin [75], proanthocyanidins [100, 137], and galanin [54] perform their anti-proliferative and anti-inflammatory effects in different cell types via utilization of their important pharmacological properties of antioxidant to neutralize baleful ROS, interrupt pro-inflammatory MAPK, NF-κB, and STAT3 signalings and potentiate anti-inflammatory NRF-2, SIRT1, and HO-1 pathways. Other non-canonical anti-inflammatory drugs, like Ambroxol [66] and MTH1 inhibitors[102] could be used as antipsoriatic drugs possessing capabilities of aiming at ROS elimination in specific diseasing-causing cell types to ameliorate psoriasis. In addition to the above-mentioned chemical and non-classical drugs as a potential treatment for psoriasis, some of the main classical traditional anti-psoriasis drugs also can regulate immune cell metabolism and keratinocyte excessive proliferation. For example, MTX, the classical anti-psoriasis drug [138], can also be regarded as an antioxidant, which can neutralize free radicals and reactive oxygen superoxide (O2), thereby inhibiting the formation of malondialdehyde acetaldehyde (MAA) adducts. Vitamin A is an indirect antioxidant that indirectly regulates many genes involved in mediating typical antioxidant responses and can prevent lipid peroxidation, thus remodeling metabolic pathways and gene expression profiles in tissues and cells [139]. However, their traditional therapeutic routes targeting the abovementioned inflammatory network are still not satisfactory due to their substantial toxicity concerning internal organs, nonspecific targeting, low effective drug concentration of skin lesions, specific risks of infection, and poor patient compliance [140, 141]. 90% of voters in the International eDelphi Consensus Meeting recommended switching the MTX route to subcutaneous administration against psoriasis for averting oral adverse events [142].

Topical therapy is the safe, convenient, and most widely used approach for the transdermal delivery of classical antipsoriatic drugs to treat mild psoriasis and consolidation treatment of moderate-to-severe psoriasis in current clinical applications. the circumvent of adverse reactions and sufficient concentration of therapeutic drug at the target lesion could be facilitated by transdermal administrations [141, 143]. A number of strategies for the transdermal delivery of bioactive drugs have been investigated for the clinic. Compared with the parenteral delivery route, topical different formulations [144], including ointment, cream, lotion, liquid, emulsions, gel formulations and nanomedicines-assisted transdermal delivery of drugs could directly repress the deterioration of psoriasis to achieve comparable therapeutic effects through a variety of mechanisms with lower drug doses. Nowadays, transdermal drug delivery of systemic drugs with particular advantages of avoiding first-pass metabolism, lesser side effects, pain-free and noninvasive self-administration for patients brings into investigation [145, 146]. Still, effectively cutaneous drug absorption becomes challenging in the local treatment of psoriasis, particularly for its thickened epidermis [141].

Table 1 The pathogenetic role of ROS in dysfunctional different cell types (KCs, skin-resident and -infiltrating immune cells functions) mediated propagation of inflammatory loops in the EIME of psoriasis
Table 2 The therapeutic effects of common natural compounds and drugs in the targeted regulation of ROS-mediated pathogenesis of psoriasis
Table 3 Nanomaterials used for transdermal drug delivery in psoriasis treatment

Latest developments of biomaterials for psoriasis therapies

For the past few years, numerous studies have explored and optimized more new and refined effective therapeutic modalities for psoriasis with drugs or nanomaterials to circumvent the drawback of conventional drugs and resolve the transdermal approaches limitation of drug diffusion or permeation to the dermis. As a result, switching the dynamic equilibrium of the oxidation-reduction system of these key pathogenetic cells is quite pertinent to providing a comprehensive strategy to reshape the immune-microenvironment in psoriasis.

Fig. 3
figure 3

Different types of nanoparticles/nanocarriers used as therapeutic modalities of ROS-related psoriasis

Mounting evidence has emphasized the critical role oxidative stress played in the pathogenesis of psoriasis, which promotes the discovery of new therapeutic modalities. Based on the abovementioned reports, ROS-mediated dysfunctional different cell types (KCs, skin-resident and -infiltrating immune cells functions) in the epithelial microenvironment (EIME) propagate multiple inflammatory loops of psoriasis. Therapies based on ROS-inhibition and -elimination targets for the blockade of inflammatory loops could be effective in the treatment of psoriasis. Besides the systemic and topical antipsoriatic drugs, recent advances in nanotechnology have promoted the emergence of numerous nanosystems, as shown as Fig. 3 and Table 3, which could resolve limitations of drug systemic side effects and transdermal drug diffusion or permeation in conventional therapies.

Self-therapeutic nanomaterials for the treatment of psoriasis

Mental nanoparticles

Ce-based nanoparticles

Ceria nanoparticles (NPs) have been regarded as typical nano-antioxidants with therapeutic effects on a range of ROS-related diseases, including hepatic ischemia-reperfusion injury [171], acute kidney injury (AKI) [172, 173], multiple CNS diseases [174, 175], rheumatoid arthritis (RA) [176], etc. Their detailed mechanism for scavenging the overproduction of ROS from pathogenic cells restores the redox homeostasis for reprogramming the immuno-environment by facilitating the transformation of cytopathogenic phenotypic transition into the cytoprotective subtype. Besides, the ceria NPs could be modified with the capability of localized into mitochondria for reduction of ROS against neuroinflammation [175]. It is well-documented that psoriasis is a disordered oxidative stress-related inflammatory disease, a feasible approach could be manufactured to downregulate oxidative stress for a detoxification effect via direct delivery of ROS-regulating nanosystems into skin lesions. On account of the above ROS-eliminating activity of ceria, it uncovers more opportunities for potential therapeutic interventions to the progress of psoriasis to reconfigure the steady-state cellular redox homeostasis in EIME. Wu L. et al. fabricated β-cyclodextrins (β-CDs) modified ceria NPs (β-CDs/CeO2 NPs) with drug-loaded and antioxidative activities for combinational psoriasis therapy in the IMQ-induced psoriatic model (Fig. 4). CeO2 with intrinsic superoxide dismutase- and catalase-mimicking capacities have been developed as therapeutic agents for cytoprotection against ROS-mediated damage [177] and provides combinational antipsoriatic efficacy for transdermal delivery of dithranol (DIT) [178]. Further research is imperative to broaden better our understanding of the ceria-based NPs and tailor their functional orientations to meet their specific needs for reversing the role of specific redox pathways in the interrelated pathology of psoriasis.

Fig. 4
figure 4

Ce NPs-based self-therapeutic nanomaterials for the topical treatment of psoriasis. β-cyclodextrin modified ceria nanoparticles were designed as a ROS scavenger nanozyme to transdermal delivery of dithranol for the combinational therapy of psoriasis. Reproduced with permission [178]. Copyright 2020, Dove Medical Press

Gold nanoparticles

Gold nanoparticles (Au NPs) have shown good biocompatibility, water-solubility, catalytic activity and great potential as self-therapeutic nanosystems for drug delivery platforms against inflammatory disorders, including AKI and RA due to their anti-inflammatory and antioxidative performances [179, 180]. It has been reported that the tunable bio-effects of Au NPs differ across research due to the application of regulatory particle sizes and surface modification [181]. Özcan A et al. found that Au NPs, as transdermal drug delivery, could facilitate MTX transcutaneous delivery into the skin across the stratum corneum barriers and lessen psoriatic skin inflammation in noninvasive manners, to avoid systemic side effects and achieve good skin penetration, ascribed to small size and immunomodulatory effects of Au NPs (Fig. 5a) [158]. Likewise, Au NPs coupled with oligonucleotides (siRNA) can be qualified to preferentially gene editing and enhance the transdermal treatment of psoriasis (Fig. 5b) [182]. Additionally, sub-15 nm Au NPs could be tailored by 30% octadecyl chains to restore the deterioration of psoriasis without an excipient and the side effects of hair loss and skin wrinkling [159]. It was attributed to the optimal core size for effective endocytosis by KCs and the assistance of epidermal delivery of Au NPs to effectively restrain the IL-17 signaling pathway mediated the epidermal hyperproliferation and inflammation in the IMQ-induced psoriasis mice model (Fig. 5c). Therefore, the decisive contributions of these studies in bespoke Au NPs for the intervention of psoriasis make a favorable difference in the biomedical application of Au NPs for the treatment of psoriasis.

Fig. 5
figure 5

Au NPs-based self-therapeutic nanomaterials for the topical treatment of psoriasis. a MTX-GNPs were prepared to inhibit the exacerbation of psoriasis via reshaping the immune infiltration and cytokine secretion of the skin. Reproduced with permission [156]. Copyright 2020, Elsevier. b siRNA conjugated with spherical nucleic acid gold nanoparticles were developed for the reduction of T cell activation and inflammatory gene expression to topically control the progress of psoriasis. Reproduced with permission [182]. Copyright 2017, Elsevier. c Alkyl-terminated Au NPs were synthesized as self-therapeutic nanomedicines for topically preventing and treating imiquimod-induced psoriasis mice via downregulation of gene expression involved in the interleukin-17 signaling pathway. Reproduced with permission [159]. Copyright 2017, American Chemical Society

Silver nanoparticles

Recently, considerable research have demonstrated that bio-friendly silver (Ag) NPs have potential properties in immunomodulatory and ROS-modulating activities by elaborately tailoring their size and shape [183, 184]. AgNPs decorate biomaterials with appropriately therapeutic window concentrations of Ag+ ions, not only can they endow AgNPs with the biological function of regulating macrophage polarization and ROS responsiveness but also optimize their biocompatibility for alleviating a wide variety of preclinical inflammatory diseases such as RA and diabetic wound [183,184,185,186,187]. Ag NPs extracted from natural herbs efficiently suppressed NF-κB activation of macrophage in vitro and human psoriasis plaques, eventually resulting in psoriasis resolution [188]. Furthermore, immunomodulatory Ag NPs co-decorated ZnO nanoparticles were conferred with the capability of inactivating p65 in proinflammatory macrophages and abrogating the secretion of ROS-induced adaptive cytokines in psoriatic KCs (Fig. 6). These composite nanoparticles (Ag/ZnO NPs) identified as self-therapeutic nanocarriers to deliver MTX into the stratum corneum, not only exerted their immunosuppressive effect but also combinedly augment the antipsoriatic efficacy of a low-dose MTX under the realization of sustainable MTX release [189]. Therefore, these results suggested that the appropriate concentration of Ag NPs could be designed for anti-inflammation and ROS-depletion against inflammatory disorders.

Fig. 6
figure 6

Ag NPs-based self-therapeutic nanomaterials for the topical treatment of psoriasis. The Car@NMs@MTX-ZA hydrogel was successfully fabricated as self-therapeutic nanotherapy for combined anti-inflammation with antiproliferation for the treatment of psoriasis. Reproduced with permission [189]. Copyright 2022, Springer Nature

Polymers

It is worth mentioning that multifarious polymers with different modifications are available for a wide range of biomedical applications, including drug delivery systems [190], gene targeting [191, 192], and therapeutic agents [193, 194] for targeted therapy in inflammatory diseases. Cell-free DNA (cfDNA) has been proven to be an inflammatory trigger to activate DNA sensors-induced immune responses involved in initiating and exacerbating the pathogenesis of autoimmune diseases [195, 196], such as RA, SLE and psoriasis. It presents evidence that approaches for effectively eliminating cfDNA is feasible for the remission of disease severity. Liang H et al. constructed self-assembly of PLGA-block-PDMA block copolymer, PLGA-b-PDMA463 with a high DNA-binding affinity, which could scavenge cfDNA released from dead and dying cells to restrain autoimmune inflammation against RA [194]. In psoriasis, these cationic nanoparticles were composed of the diblock copolymer of PLGA-b-PDMA474, which similarly beneficially prevented cfDNA from the formation of the DNA-LL37 immune complex via topical application against psoriasis (Fig. 7) [197]. Altogether, these data implied that the possible applications of bespoke polymers could neutralize the detrimental effects of cfDNA or RNA signature to serve as potential antipsoriatic nanomedicines.

Fig. 7
figure 7

Polymer-based self-therapeutic nanomaterials for the topical treatment of psoriasis. Cationic nanoparticles were constructed as cfDNA scavengers for topical remission of DNA-LL37-induced cell inflammation in a psoriasiform mice model and cynomolgus monkey model. Reproduced with permission [197]. Copyright 2020, American Association for the Advancement of Science

Natural bioactive compound

Natural products have gained considerable attention for psoriasis treatment due to their excellent biocompatibility and high effectiveness. Bilirubin, a highly potent anti-cancer and anti-inflammatory compound can scavenge various ROS and plays a crucial role in protecting cells from oxidative stress-mediated damage in the human body [161]. Hyeongseop Keum et al. demonstrated that the bilirubin nanoparticles (BRNPs), composed of the endogenous antioxidant bilirubin and a safe hydrophilic PEG polymer, can readily infiltrate the disrupted outer cornified skin barrier and efficiently downregulate the accumulation of intracellular ROS in KCs. Meanwhile, this novel biocompatible nanomedicine could be further expanded to treat other chronic skin inflammation diseases, including atopic dermatitis [160]. Polyphenols and flavonoids in natural products have been widely used in the treatment of inflammation-related diseases due to their excellent antioxidative properties. Recently, mung bean-derived NPs (MBNs) with a facile approach has been reported for alleviating skin inflammation. MBNs can regulate macrophage polarization and antagonize the activation of the nuclear factor kappa B (NF-κB) signaling pathway which are conducive to the subsides of inflammation in psoriasiform skin [162]. Moreover, melatonin (MLT), a natural hormone and antioxidant mainly derived from the pineal gland with the circadian rhythm of secretion, have regarded as an anti-inflammation and immunomodulator for inflammatory skin diseases [198,199,200,201], such as skin psoriasis [201] and wound healing [163]. Several studies have shown that the circadian rhythm of melatonin secretion in psoriatic patients is disappeared and melatonin-dependent redox homeostasis of the skin cells is dysregulated [201, 202]. Topical or systemic administration of melatonin could make good effective in diminishing the extensive ROS generation and proinflammatory cytokines under psoriasis and skin tissue regeneration [198, 201]. Taken together, these biologically-derived antioxidant NPs have not only significant efficacy but also high clinical translation potential.

Nanomaterial-based transdermal drug delivery platform for the treatment of psoriasis

Other than the aforesaid representatively self-therapeutic nanoparticles for the topical restoration of psoriasis. Recently, several nanocarriers, such as liposomes [151, 153], polymers [157, 197], silica nanoparticles[157, 203], metal nanoparticles [158, 159] and microneedles[12] have been introduced to favor transdermal delivery of antipsoriatic drugs and gene editing efficiency, which strategically make contributions to avoidance of their low solubility, bioavailability, and poor skin permeability to augment their antipsoriatic efficacy.

Lipid nanoparticles

It is widely recognized that lipid nanoparticles have been widely used in skin-related diseases [153, 204] and skin-based cosmetics [205], owing to their excellent bioavailability and biodegradability. Their comprehensive roles of both topical drug carriers and penetration enhancers, improve transdermal delivery of drugs [151, 206, 207], peptides [153], and oligonucleotide [154, 208] into skin lesions. Kim JY et al. designed STAT3-inhibiting peptide-encased discoidal lipid nanoparticles (DLNPs) that could contribute to promoting the penetration of peptide inhibitors into thicked stratum corneum of psoriasis (Fig. 8a). Meanwhile, these lipid formulation-based transcutaneous delivery systems exerted good biocompatibility without the side effects of conventional corticosteroid drugs [153]. In addition, Suzuki IL et al. fabricated polymer-lipid nanoparticles (PLNs) to resolve the delivery limitation of RNAi topical therapy, such as improving the biological stability of siRNA, optimizing its cellular endocytosis and sufficient endosomal release (Fig. 8b) [208]. Analogously, curcumin-loaded cellulose nanofiber (CNF) films composed of hybridized curcumin (Cur)-loaded nanostructured lipid carriers (NLCs) were constructed to enhance the deposition of curcumin into the dermis via topical treatment, conducing to amelioration of the psoriatic skin symptoms in IMQ-induced mice, almost comparable to topical corticosteroid cream [206]. Another report also demonstrated that curcumin-loaded hyaluronan (HA)-modified ethosomes could target overexpressed CD44 protein and allowed the slow release of the loaded curcumin in the inflamed epidermis [209]. Yet the limitation of lipid nanoparticles is vulnerable to oxidative degradation and exhibits poor stability, resulting in lower drug payload and inconvenient storage. These carrier systems may not have the capacity of prolonging circulation and retention, leading to a limit in the systemic bioavailability and therapeutic efficacy of cargos. More efforts should be made to optimize the facility of lipid nanoparticles.

Fig. 8
figure 8

Lipid nanomaterials-based transdermal drug delivery platform for the treatment of psoriasis. a The preparation of the DLNP transcutaneous delivery system could improve the skin penetration of STAT3-inhibiting peptides for efficiently treating psoriatic skin inflammation without causing adverse systemic events. Reproduced with permission [153]. Copyright 2018, American Chemical Society. b Hybrid polymer-lipid nanoparticles in combinational with photosensitizer TPPS2a for delivery of siRNA were aimed to topical treat psoriasis effectively through optimizing the endosomal escape of TNFα siRNA in the cytoplasm. Reproduced with permission [208]. Copyright 2021, Elsevier. c Lipid-hybridized CNF film was successfully prepared for transdermal delivery of curcumin to cure psoriasis. Reproduced with permission [206]. Copyright 2018, Elsevier

Silica nanoparticles

It is well-demonstrated that mesoporous silica nanoparticles have been considered as available drug/gene delivery carriers for their unique properties and biocompatibility. They could be functionalized with specific properties via tuning their size and surface modification/ bioconjugation for targeting and delivering therapeutic agents against a variety of inflammatory diseases [210], such as RA [176], osteoporosis [211], and atherosclerosis [212], etc. Owing to the abovementioned advantages of silica NPs, Mo C et al. employed dendritic mesoporous silica NPs as drug carriers to enhance the penetration activity of erianin across the skin in the favor of exerting an inhibitory effect on keratinocyte proliferation for the topical therapy of psoriasis (Fig. 9a) [203]. Moreover, the skin retention and permeability of silica NPs could be regulated by the particle size and polymer decoration, thereby affecting their affinity to cfDNA in the dermis along with regulation of the antipsoriatic effects (Fig. 9b) [157]. As a result of these positive results, it is encouraging that the versatile well-controlled and -modified fabrication of silica NPs has great potential to clinically apply to treat cutaneous inflammatory diseases.

Fig. 9
figure 9

Silica nanomaterials-based transdermal drug delivery platform for the treatment of psoriasis. a The synthesis of erianin-loaded dendritic mesoporous silica was employed for topical therapy of psoriasis, ascribed for their mechanisms on pro-apoptotic effect in KCs. Reproduced with permission [203]. Copyright 2020, Springer Nature. b Optimized size of silica NPs decorated with polymer could elevate the affinity of cfDNA to inhibit topical psoriasis inflammation via better penetration ability. Reproduced with permission [157]. Copyright 2021, Elsevier

Polymer/nanomicelles

It is widely known that polymer/nanomicelles can promote targeted therapy and sustained hydrophobic drug delivery with relatively high drug loading capacity, except for their performance as cfDNA scavengers. Because of their capability of prolonged circulation, reducing the initial-burst release and delivery of the hydrophobic drug, they are often utilized as a carrier system for transdermal drug delivery to resolve the restriction of drug controlled release and percutaneous absorption, thereby circumventing the drug-associated side effects [11, 189, 190]. Polycaprolactone-Polyethyleneglycol-Polycaprolactone (PCL-PEG-PCL)-based self-assembled nanomicelles were employed as a carrier system for efficient delivery and sustainable release of MTX against RA and psoriasis through the transdermal route [189, 190]. Similarly, the stable multi-component monolithic lipid-polymer hybrid nanoparticles (LPNs) were fabricated to load clobetasol propionate, a potent corticosteroid, contributing to facilitating its sustained release and penetration into deeper dermis, consequently exhibiting enhanced therapeutic effect at dose reduction without systemic toxicities absorption of the corticosteroids (Fig. 10) [11]. However, the therapeutic efficacy of topical administration is compromised by the comprehensive effect of limited penetration and skin retention. Yang Mai et al. developed the tris (hydroxymethyl) aminomethane-modified bioadhesive nanoparticles (Tris-BNPs) encapsulated with betamethasone dipropionate (BD) which showed deeper penetration and longer retention compared with commercial BD ointment. This formulation can mitigate skin inflammation after only a single administration [213]. Thus, all these present works demonstrated polymers with good drug loading capacity, biocompatibility, stability, drug controlled release and efficient cellular uptake, possessed great potential for pharmaceutical applications in the field of transdermal drug delivery systems. However, the drug capacity strongly depends on the concentrations of nanomicelles [214]. Strategies should be innovated to combine the advantages of different nanoparticles to achieve most of the benefits of improved transcutaneous antipsoriatic efficacy.

Fig. 10
figure 10

Polymer/nanomicelles-based transdermal drug delivery platform for the treatment of psoriasis. Lipid-polymer hybrid nanoparticles were fabricated to load clobetasol propionate for enhancement of its cellular uptake and skin permeability to improve antipsoriatic efficacy. Reproduced with permission [11]. Copyright 2020, Elsevier

Microneedles

Emerging nanotechnologies based-microneedles associated with efficient settlement for the dilemma of skin penetration hold tremendous promise in transdermal delivery therapy [145, 215]. Microneedles are capable of traversing the stratum corneum in a micro-invasive manner and directly translocating bioactive drugs into the dermis [12, 168,169,170, 216]. It could be equipped with various therapeutic efficacies via the incorporation of appropriate structural nanomaterials, genome editing materials as well as drug molecules or nanomedicines with tailored pharmacological properties. Wan T et al. had taken advantage of the CRISPR-Cas9–based genome editing technology for precisely targeting the inflammatory signatures of NLRP3, which mediated abnormal cross-talking of innate and adaptive immune responses and glucocorticoid resistance in psoriasis [168]. More importantly, the presence of a microneedles-mediated transdermal therapeutic strategy positively reduced off-target effects of gene editing by allowing the local release of genome editor in target lesions of psoriasis and atopic dermatitis to improve glucocorticoid sensitivity (Fig. 11a, b). Additionally, Q. Jing et al. utilized the homologous targeting functions of the HaCaT cell membrane to develop HaCaT cell membrane-coated nanocarriers for transdermal targeted delivery of shikonin in the pathological epidermis, as shown in Fig. 11c-d. This nanocomposite could be internalized by the KCs, leading to the triggering of drug release in the target lesion. Ultimately, the augmented therapeutic efficacy of shikonin against imiquimod-induced psoriatic epidermal hyperplasia was achieved [216]. As surveyed above, whereas therapeutic drug delivery through microneedles, has received considerable attention for different applications in the field of dermatology, the potential skin bacterial, fungal infection-associated risks, sensitization, and other restrictions of the costs, transportation, cargoes stability, and loading are inevitable [169, 170]. More studies should be investigated to optimize the biocompatibility of microneedles before being applied to human skin. Meanwhile, further schemes of ingredients should be facilitated to resolve the above limitation and optimize the clinical translation of formulations.

Fig. 11
figure 11

Microneedles-based transdermal drug delivery platform for the treatment of psoriasis. a Microneedle-mediated transdermal codelivery of CRISPR-Cas9–based genome editor and glucocorticoids were used for high-efficiency treatment of psoriasis. Reproduced with permission [168]. Copyright 2021, American Association for the Advancement of Science. b Characterization images of the MN patches, CP/Ad-SS-GD/Cas9 RNP nanoparticles and Dex-loaded PLGA nanoparticles; drug release of Cas9 protein and Dex from the MN patch; fluorescence images of MN patch. Reproduced with permission [168]. Copyright 2021, American Association for the Advancement of Science. c Schematic illustration of the synthesis of SKN-PMs and HCM/SKN-PMs. Reproduced with permission [216]. Copyright 2021, Elsevier. d Sketch of the MN-HCM/SKN-PM preparation process and their characterization images. Reproduced with permission [216]. Copyright 2021, Elsevier

Hydrogel

In consideration of multiple inflammatory pathways of psoriasis immunopathogenesis and optimization of topical drug bioavailability, inhibition of psoriasis activity with multiple therapeutic modalities specific to different targets outbalance single-agent approaches. Consequently, an ideal percutaneous nanocarrier needs to meet the following requirements: (1) self-therapeutic activity, with intrinsic anti-inflammatory property and improved therapeutic efficacy of extrinsic medication; (2) better drug loading capacity and controllable drug release; (3) good moisture retention, which can maintain the moist environment of the skin and reduction of drug breakage; and (4) enhanced patient compliance. Hydrogels, owing to their biochemical characteristics of good retention, avoidance of drug leakage, good hydrophilicity and adhesiveness, have been identified as the most competitive candidate for the percutaneous treatment of inflammatory diseases [164,165,166]. Considerable research has demonstrated that hydrogels can be well-appointed with tunable functions via the incorporation of various bioactive substances, such as nanoparticles and drugs and establish well-pleasing biomedical applications in transdermal drug delivery [190, 217,218,219,220]. As shown in Fig. 12, For improvement of the transdermal application of lyophobic drugs, Sun L et al. fabricated curcumin (Cur) loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) loaded into the hydrogel which was employed to topically treat IMQ-induced psoriasis-like mouse for promotion of drug permeability across skin and enhancement of anti-psoriatic activity (Fig. 12a) [219]. similarly, Qiu F et al. produced Celastrol Noisome hydrogel (Cel Nio gel) for topical administration to psoriasis. When applied in the IMQ-induced psoriatic mice model, cel was mainly accumulated in the skin other than exposure to the blood or lymphatic system, resulting in the reduction of the mRNA levels of inflammatory cytokines (Fig. 12b) [221]. Additionally, Kajal Rana et al. presented that a betamethasone-loaded topical hydrogel (B-Gel) which can efficiently entrap steroids with the properties of spreadability and sustained release drugs, provided an alternative for topical application of steroids [220]. Moreover, implementing biocompatible hydrogel micropatch probes integrated with mass spectrometry to explore the skin metabolome could be regarded as a diagnostic approach to provide information about the pathological alterations of the skin metabolome caused by psoriasis, favoring understanding of the complicated pathophysiology. However, antibiotic-immobilized hydrogels should be seriously utilized due to the problems of multidrug resistance and relatively long treatment course, while hydrogels loaded with noble metal nanoparticles often cause undesirable systemic toxicity.

Above all, It is noted that most of the existing ROS-based nanomedicines or transdermal delivery nanoplatform are engineered with some deficiency, comprehensive resolution of limitations of these nanobiotechnological carriers related to drug controlled release, drug lower loading capacity and optimizing transdermal permeation, particularly in the thickened stratum corneum of psoriasis remains intractable. Therefore, it is highly expected that address these issues in elaborately engineered redox-active nanosystems design and a more simplified way for the feasibility of clinical translation, rather than decorating sophisticated structures that may render potential biosafety issues.

Fig. 12
figure 12

Hydrogel-based transdermal drug delivery platform for the treatment of psoriasis. a Cur encapsulated into PLGA NPs were synthesized as hydrogel to optimize the dispersion, sustained release and penetration of curcumin across the skin for improvement of its anti-psoriatic efficacy. Reproduced with permission [219]. Copyright 2017, Elsevier. b Therapeutic mechanism of Cel Nio gel for the transcutaneous treatment of imiquimod-induced psoriasiform skin inflammation. Reproduced with permission [221]. Copyright 2021, Dove Medical Press

Summary and Outlook

As the significant role of oxidative stress in the molecular pathological mechanisms of psoriasis continues to be unraveled, targeting ROS in dysfunctional different cell types in EIME offers a promising methodology for psoriasis. In the future, a more major focus should be paid to investigating more effectively new-generation of therapeutics mediated precisely regulation of cellular ROS concentrations in EIEM within a physiological threshold. Meanwhile, it is appreciated that the noticeable advances in the field of nanotechnology regarding multifarious nanomaterials with ROS depletion performances have been witnessed. Most notably, besides current ROS-detoxifying self-therapeutic nanomaterials directly against psoriasis, the emergence of a nano-platform for transdermal drug delivery system greatly expands the application of nanomaterials in the field of precision medicine. Nanotechnologies dramatically facilitate the absorption and diffusion of drugs at skin barriers, especially in psoriatic conditions characterized by highly packed SC, giving rise to increased drug availability in local therapy and decreased systemic adverse effects. The incorporation of nanotechnologies offers protection for the labile therapeutically active compounds as well as the assistance of drug storage and prolonged residence time of drug molecules at the target site against skin disease. Aside from the mentioned already, it is anticipated that more comprehensive investigations related to reconstructed skin experimental models should mimic the real-time biological status of skin lesions for the achievement of accessing the permeability and pharmaceutical properties of nanomaterials. Furthermore, the skin irritation and biosafety evaluations of nanomaterials about long-term therapeutic effects should be conducted for potential clinical transformation. Finally, we envision that these nano-biotechnologies will expand more therapeutic avenues for precision medicine, especially in skin diseases.

Availability of data and materials

Not applicable.

References

  1. Armstrong AW, Read C. Pathophysiology, clinical presentation, and treatment of psoriasis: a review. JAMA. 2020;323(19):1945–60. https://doi.org/10.1001/jama.2020.4006.

    Article  CAS  PubMed  Google Scholar 

  2. Michalek IM, Loring B, John SM. A systematic review of worldwide epidemiology of psoriasis. J Eur Acad Dermatol Venereol. 2017;31(2):205–12. https://doi.org/10.1111/jdv.13854.

    Article  CAS  PubMed  Google Scholar 

  3. Parisi R, Iskandar IYK, Kontopantelis E, Augustin M, Griffiths CEM, Ashcroft DM. National, regional, and worldwide epidemiology of psoriasis: systematic analysis and modelling study. BMJ. 2020;369:m1590. https://doi.org/10.1136/bmj.m1590.

    Article  PubMed  PubMed Central  Google Scholar 

  4. GBD 2019 diseases and injuries collaborators. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the global burden of disease study 2019. Lancet (London England). 2020;396(10258):1204–22. https://doi.org/10.1016/S0140-6736(20)30925-9.

    Article  Google Scholar 

  5. Damiani G, Bragazzi NL, Karimkhani Aksut C, Wu D, Alicandro G, McGonagle D, Guo C, Dellavalle R, Grada A, Wong P, et al. The global, regional, and national burden of psoriasis: results and insights from the global burden of disease 2019 study. Front Med (Lausanne). 2021;8:743180. https://doi.org/10.3389/fmed.2021.743180.

    Article  Google Scholar 

  6. Yan B-X, Chen X-Y, Ye L-R, Chen J-Q, Zheng M, Man X-Y. Cutaneous and systemic psoriasis: classifications and classification for the distinction. Front Med (Lausanne). 2021;8:649408. https://doi.org/10.3389/fmed.2021.649408.

    Article  Google Scholar 

  7. Sies H, Jones DP. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol. 2020;21(7):363–83. https://doi.org/10.1038/s41580-020-0230-3.

    Article  CAS  PubMed  Google Scholar 

  8. Poprac P, Jomova K, Simunkova M, Kollar V, Rhodes CJ, Valko M. Targeting free radicals in oxidative stress-related human diseases. Trends Pharmacol Sci. 2017;38(7):592–607. https://doi.org/10.1016/j.tips.2017.04.005.

    Article  CAS  PubMed  Google Scholar 

  9. Kuehne A, Emmert H, Soehle J, Winnefeld M, Fischer F, Wenck H, Gallinat S, Terstegen L, Lucius R, Hildebrand J, et al. Acute activation of oxidative pentose phosphate pathway as first-line response to oxidative stress in human skin cells. Mol Cell. 2015;59(3):359–71. https://doi.org/10.1016/j.molcel.2015.06.017.

    Article  CAS  PubMed  Google Scholar 

  10. Kidane D, Chae WJ, Czochor J, Eckert KA, Glazer PM, Bothwell AL, Sweasy JB. Interplay between DNA repair and inflammation, and the link to cancer. Crit Rev Biochem Mol Biol. 2014;49(2):116–39. https://doi.org/10.3109/10409238.2013.875514.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Pukale SS, Sharma S, Dalela M, Singh AK, Mohanty S, Mittal A, Chitkara D. Multi-component clobetasol-loaded monolithic lipid-polymer hybrid nanoparticles ameliorate imiquimod-induced psoriasis-like skin inflammation in swiss albino mice. Acta Biomater. 2020;115:393–409. https://doi.org/10.1016/j.actbio.2020.08.020.

    Article  CAS  PubMed  Google Scholar 

  12. Du H, Liu P, Zhu J, Lan J, Li Y, Zhang L, Zhu J, Tao J. Hyaluronic acid-based dissolving microneedle patch loaded with methotrexate for improved treatment of psoriasis. ACS Appl Mater Interfaces. 2019;11(46):43588–98. https://doi.org/10.1021/acsami.9b15668.

    Article  CAS  PubMed  Google Scholar 

  13. Dainichi T, Kitoh A, Otsuka A, Nakajima S, Nomura T, Kaplan DH, Kabashima K. The epithelial immune microenvironment (EIME) in atopic dermatitis and psoriasis. Nat Immunol. 2018;19(12):1286–98. https://doi.org/10.1038/s41590-018-0256-2.

    Article  CAS  PubMed  Google Scholar 

  14. Jiang Y, Tsoi LC, Billi AC, Ward NL, Harms PW, Zeng C, Maverakis E, Kahlenberg JM, Gudjonsson JE. Cytokinocytes: the diverse contribution of keratinocytes to immune responses in skin. JCI Insight. 2020;5(20):e142067. https://doi.org/10.1172/jci.insight.142067.

    Article  PubMed Central  Google Scholar 

  15. Trouba KJ, Hamadeh HK, Amin RP, Germolec DR. Oxidative stress and its role in skin disease. Antioxid Redox Signal. 2002;4(4):665–73. https://doi.org/10.1089/15230860260220175.

    Article  CAS  PubMed  Google Scholar 

  16. Emmert H, Fonfara M, Rodriguez E, Weidinger S. NADPH oxidase inhibition rescues keratinocytes from elevated oxidative stress in a 2D atopic dermatitis and psoriasis model. Exp Dermatol. 2020;29(8):749–58. https://doi.org/10.1111/exd.14148.

    Article  CAS  PubMed  Google Scholar 

  17. Pleńkowska J, Gabig-Cimińska M, Mozolewski P. Oxidative stress as an important contributor to the pathogenesis of psoriasis. Int J Mol Sci. 2020. https://doi.org/10.3390/ijms21176206.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Yan D, Afifi L, Jeon C, Trivedi M, Chang HW, Lee K, Liao W. The metabolomics of psoriatic disease. Psoriasis (Auckl). 2017;7(1):1–15. https://doi.org/10.2147/PTT.S118348.

    Article  Google Scholar 

  19. Hao Y, Zhu Y-J, Zou S, Zhou P, Hu Y-W, Zhao Q-X, Gu L-N, Zhang H-Z, Wang Z, Li J. Metabolic syndrome and psoriasis: mechanisms and future directions. Front Immunol. 2021;12:711060. https://doi.org/10.3389/fimmu.2021.711060.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kabashima K, Honda T, Ginhoux F, Egawa G. The immunological anatomy of the skin. Nat Rev Immunol. 2019;19(1):19–30. https://doi.org/10.1038/s41577-018-0084-5.

    Article  CAS  PubMed  Google Scholar 

  21. Streilein JW. Skin-associated lymphoid tissues (SALT): Origins and functions. J Invest Dermatol. 1983;80(Suppl):12s–6s. https://doi.org/10.1111/1523-1747.ep12536743.

    Article  PubMed  Google Scholar 

  22. Streilein JW. Circuits and signals of the skin-associated lymphoid tissues (SALT). J Invest Dermatol. 1985;85(1 Suppl):10 s–13 s. https://doi.org/10.1111/1523-1747.ep12275413.

    Article  Google Scholar 

  23. Sontheimer RD. Perivascular dendritic macrophages as immunobiological constituents of the human dermal microvascular unit. J Invest Dermatol. 1989;93(2 Suppl):96S–101S. https://doi.org/10.1111/1523-1747.

    Article  CAS  PubMed  Google Scholar 

  24. Lowes MA, Suarez-Farinas M, Krueger JG. Immunology of psoriasis. Annu Rev Immunol. 2014;32:227–55. https://doi.org/10.1146/annurev-immunol-032713-120225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zaba LC, Cardinale I, Gilleaudeau P, Sullivan-Whalen M, Suárez-Fariñas M, Suárez Fariñas M, Fuentes-Duculan J, Novitskaya I, Khatcherian A, Bluth MJ, et al. Amelioration of epidermal hyperplasia by TNF inhibition is associated with reduced Th17 responses. J Exp Med. 2007;204(13):3183–94. https://doi.org/10.1084/jem.20071094.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Plenkowska J, Gabig-Ciminska M, Mozolewski P. Oxidative stress as an important contributor to the pathogenesis of psoriasis. Int J Mol Sci. 2020. https://doi.org/10.3390/ijms21176206.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Xian D, Song J, Yang L, Xiong X, Lai R, Zhong J. Emerging roles of redox-mediated angiogenesis and oxidative stress in dermatoses. Oxid Med Cell Longev. 2019;2019:2304018. https://doi.org/10.1155/2019/2304018.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Young CN, Koepke JI, Terlecky LJ, Borkin MS, Boyd SL, Terlecky SR. Reactive oxygen species in tumor necrosis factor-alpha-activated primary human keratinocytes: implications for psoriasis and inflammatory skin disease. J Invest Dermatol. 2008;128(11):2606–14. https://doi.org/10.1038/jid.2008.122.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mailloux RJ, McBride SL, Harper M-E. Unearthing the secrets of mitochondrial ROS and glutathione in bioenergetics. Trends Biochem Sci. 2013;38(12):592–602. https://doi.org/10.1016/j.tibs.2013.09.001.

    Article  CAS  PubMed  Google Scholar 

  30. Muri J, Kopf M. Redox regulation of immunometabolism. Nat Rev Immunol. 2021;21(6):363–81. https://doi.org/10.1038/s41577-020-00478-8.

    Article  CAS  PubMed  Google Scholar 

  31. Boo YC. Natural Nrf2 modulators for skin protection. Antioxid (Basel). 2020;9(9):812. https://doi.org/10.3390/antiox9090812.

    Article  CAS  Google Scholar 

  32. Xu F, Xu J, Xiong X, Deng Y. Salidroside inhibits MAPK, NF-κB, and STAT3 pathways in psoriasis-associated oxidative stress via SIRT1 activation. Redox Rep. 2019;24(1):70–4. https://doi.org/10.1080/13510002.2019.1658377.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Sakon S, Xue X, Takekawa M, Sasazuki T, Okazaki T, Kojima Y, Piao J-H, Yagita H, Okumura K, Doi T, et al. NF-kappaB inhibits TNF-induced accumulation of ROS that mediate prolonged MAPK activation and necrotic cell death. EMBO J. 2003;22(15):3898–909. https://doi.org/10.1093/emboj/cdg379.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kennedy-Crispin M, Billick E, Mitsui H, Gulati N, Fujita H, Gilleaudeau P, Sullivan-Whalen M, Johnson-Huang LM, Suárez-Fariñas M, Krueger JG. Human keratinocytes’ response to injury upregulates CCL20 and other genes linking innate and adaptive immunity. J Invest Dermatol. 2012;132(1):105–13. https://doi.org/10.1038/jid.2011.262.

    Article  CAS  PubMed  Google Scholar 

  35. Bernard FX, Morel F, Camus M, Pedretti N, Barrault C, Garnier J, Lecron JC. Keratinocytes under fire of proinflammatory cytokines: bona fide innate immune cells involved in the physiopathology of chronic atopic dermatitis and psoriasis. J Allergy (Cairo). 2012;2012: 718725. https://doi.org/10.1155/2012/718725

    Article  CAS  PubMed Central  Google Scholar 

  36. Kumari S, Bonnet MC, Ulvmar MH, Wolk K, Karagianni N, Witte E, Uthoff-Hachenberg C, Renauld J-C, Kollias G, Toftgard R, et al. Tumor necrosis factor receptor signaling in keratinocytes triggers interleukin-24-dependent psoriasis-like skin inflammation in mice. Immunity. 2013;39(5):899–911. https://doi.org/10.1016/j.immuni.2013.10.009.

    Article  CAS  PubMed  Google Scholar 

  37. Kashiwagi M, Hosoi J, Lai JF, Brissette J, Ziegler SF, Morgan BA, Georgopoulos K. Direct control of regulatory T cells by keratinocytes. Nat Immunol. 2017;18(3):334–43. https://doi.org/10.1038/ni.3661.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lowes MA, Russell CB, Martin DA, Towne JE, Krueger JG. The IL-23/T17 pathogenic axis in psoriasis is amplified by keratinocyte responses. Trends Immunol. 2013;34(4):174–81. https://doi.org/10.1016/j.it.2012.11.005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nestle FO, Di Meglio P, Qin J-Z, Nickoloff BJ. Skin immune sentinels in health and disease. Nat Rev Immunol. 2009;9(10):679–91. https://doi.org/10.1038/nri2622.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Kim HR, Kim JC, Kang SY, Kim HO, Park CW, Chung BY. Rapamycin alleviates 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced aggravated dermatitis in mice with imiquimod-induced psoriasis-like dermatitis by inducing autophagy. Int J Mol Sci. 2021;22(8):3968. https://doi.org/10.3390/ijms22083968.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Woodby B, Sticozzi C, Pambianchi E, Villetti G, Civelli M, Valacchi G, Facchinetti F. The PDE4 inhibitor CHF6001 affects keratinocyte proliferation via cellular redox pathways. Arch Biochem Biophys. 2020;685:108355. https://doi.org/10.1016/j.abb.2020.108355.

    Article  CAS  PubMed  Google Scholar 

  42. Nadeem A, Ahmad SF, Al-Harbi NO, El-Sherbeeny AM, Al-Harbi MM, Almukhlafi TS. GPR43 activation enhances psoriasis-like inflammation through epidermal upregulation of IL-6 and dual oxidase 2 signaling in a murine model. Cell Signal. 2017;33:59–68. https://doi.org/10.1016/j.cellsig.2017.02.014.

    Article  CAS  PubMed  Google Scholar 

  43. Schon MP. Adaptive and innate immunity in psoriasis and other inflammatory disorders. Front Immunol. 2019;10:1764. https://doi.org/10.3389/fimmu.2019.01764.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sano S, Chan KS, Carbajal S, Clifford J, Peavey M, Kiguchi K, Itami S, Nickoloff BJ, DiGiovanni J. STAT3 links activated keratinocytes and immunocytes required for development of psoriasis in a novel transgenic mouse model. Nat Med. 2005;11(1):43–9. https://doi.org/10.1038/nm1162.

    Article  CAS  PubMed  Google Scholar 

  45. Shen P, Deng X, Chen Z, Ba X, Qin K, Huang Y, Huang Y, Li T, Yan J, Tu S. Sirt1: A potential therapeutic target in autoimmune diseases. Front Immunol. 2021;12:779177. https://doi.org/10.3389/fimmu.2021.779177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Singh V, Ubaid S. Role of silent information regulator 1 (SIRT1) in regulating oxidative stress and inflammation. Inflammation. 2020;43(5):1589–98. https://doi.org/10.1007/s10753-020-01242-9.

    Article  CAS  PubMed  Google Scholar 

  47. Liu A, Zhang B, Zhao W, Tu Y, Wang Q, Li J. Catalpol ameliorates psoriasis-like phenotypes via SIRT1 mediated suppression of NF-κB and MAPKs signaling pathways. Bioengineered. 2021;12(1):183–95. https://doi.org/10.1080/21655979.2020.1863015.

    Article  CAS  PubMed  Google Scholar 

  48. Wang Y, Huo J, Zhang D, Hu G, Zhang Y. Chemerin/ChemR23 axis triggers an inflammatory response in keratinocytes through ROS-SIRT1-NF-κB signaling. J Cell Biochem. 2019;120(4):6459–70. https://doi.org/10.1002/jcb.27936.

    Article  CAS  PubMed  Google Scholar 

  49. Liu A, Zhao W, Zhang B, Tu Y, Wang Q, Li J. Cimifugin ameliorates imiquimod-induced psoriasis by inhibiting oxidative stress and inflammation via NF-κB/MAPK pathway. Biosci Rep. 2020;40(6):BSR20200471. https://doi.org/10.1042/BSR20200471.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Qiong H, Han L, Zhang N, Chen H, Yan K, Zhang Z, Ma Y, Xu J. Glycyrrhizin improves the pathogenesis of psoriasis partially through IL-17A and the SIRT1-STAT3 axis. BMC Immunol. 2021;22(1):34. https://doi.org/10.1186/s12865-021-00421-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang B, Xie S, Su Z, Song S, Xu H, Chen G, Cao W, Yin S, Gao Q, Wang H. Heme oxygenase-1 induction attenuates imiquimod-induced psoriasiform inflammation by negative regulation of STAT3 signaling. Sci Rep. 2016;6:21132. https://doi.org/10.1038/srep21132.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ishitsuka Y, Ogawa T, Roop D. The KEAP1/NRF2 signaling pathway in keratinization. Antioxid (Basel). 2020;9(8):751. https://doi.org/10.3390/antiox9080751.

    Article  CAS  Google Scholar 

  53. Cuadrado A, Rojo AI, Wells G, Hayes JD, Cousin SP, Rumsey WL, Attucks OC, Franklin S, Levonen A-L, Kensler TW, et al. Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat Rev Drug Discov. 2019;18(4):295–317. https://doi.org/10.1038/s41573-018-0008-x.

    Article  CAS  PubMed  Google Scholar 

  54. Sangaraju R, Alavala S, Nalban N, Jerald MK, Sistla R. Galangin ameliorates Imiquimod-Induced psoriasis-like skin inflammation in BALB/c mice via down regulating NF-κB and activation of Nrf2 signaling pathways. Int Immunopharmacol. 2021;96:107754. https://doi.org/10.1016/j.intimp.2021.107754.

    Article  CAS  PubMed  Google Scholar 

  55. Wang W, Yuhai, Wang H, Chasuna. Bagenna. Astilbin reduces ROS accumulation and VEGF expression through Nrf2 in psoriasis-like skin disease. Biol Res. 2019;52(1):49. https://doi.org/10.1186/s40659-019-0255-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Skutnik-Radziszewska A, Maciejczyk M, Fejfer K, Krahel J, Flisiak I, Kołodziej U, Zalewska A. Salivary antioxidants and oxidative stress in psoriatic patients: can salivary total oxidant status and oxidative status index be a plaque psoriasis biomarker? Oxid Med Cell Longev. 2020;2020:9086024. https://doi.org/10.1155/2020/9086024.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Melero JL, Andrades S, Arola L, Romeu A. Deciphering psoriasis. A bioinformatic approach. J Dermatol Sci. 2018;89(2):120–6. https://doi.org/10.1016/j.jdermsci.2017.11.010.

    Article  CAS  PubMed  Google Scholar 

  58. Hara-Chikuma M, Satooka H, Watanabe S, Honda T, Miyachi Y, Watanabe T, Verkman AS. Aquaporin-3-mediated hydrogen peroxide transport is required for NF-κB signalling in keratinocytes and development of psoriasis. Nat Commun. 2015;6:7454. https://doi.org/10.1038/ncomms8454.

    Article  CAS  PubMed  Google Scholar 

  59. Miller EW, Dickinson BC, Chang CJ. Aquaporin-3 mediates hydrogen peroxide uptake to regulate downstream intracellular signaling. Proc Natl Acad Sci U S A. 2010;107(36):15681–6. https://doi.org/10.1073/pnas.1005776107.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Kuraitis D, Rosenthal N, Boh E, McBurney E. Macrophages in dermatology: pathogenic roles and targeted therapeutics. Arch Dermatol Res. 2022;314(2):133–40. https://doi.org/10.1007/s00403-021-02207-0.

    Article  PubMed  Google Scholar 

  61. Wang H, Peters T, Kess D, Sindrilaru A, Oreshkova T, Van Rooijen N, Stratis A, Renkl AC, Sunderkötter C, Wlaschek M, et al. Activated macrophages are essential in a murine model for T cell-mediated chronic psoriasiform skin inflammation. J Clin Investig. 2006;116(8):2105–14. https://doi.org/10.1172/JCI27180.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Nickoloff BJ, Nestle FO. Recent insights into the immunopathogenesis of psoriasis provide new therapeutic opportunities. J Clin Investig. 2004;113(12):1664–75. https://doi.org/10.1172/JCI22147.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Zhang Y, Choksi S, Chen K, Pobezinskaya Y, Linnoila I, Liu Z-G. ROS play a critical role in the differentiation of alternatively activated macrophages and the occurrence of tumor-associated macrophages. Cell Res. 2013;23(7):898–914. https://doi.org/10.1038/cr.2013.75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Weinberg SE, Sena LA, Chandel NS. Mitochondria in the regulation of innate and adaptive immunity. Immunity. 2015;42(3):406–17. https://doi.org/10.1016/j.immuni.2015.02.002.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Liu P, Peng C, Chen X, Wu L, Yin M, Li J, Qin Q, Kuang Y, Zhu W. Acitretin promotes the differentiation of myeloid-derived suppressor cells in the treatment of psoriasis. Front Med (Lausanne). 2021;8:625130. https://doi.org/10.3389/fmed.2021.625130.

    Article  Google Scholar 

  66. Sunkari S, Thatikonda S, Pooladanda V, Challa VS, Godugu C. Protective effects of ambroxol in psoriasis like skin inflammation: exploration of possible mechanisms. Int Immunopharmacol. 2019;71:301–12. https://doi.org/10.1016/j.intimp.2019.03.035.

    Article  CAS  PubMed  Google Scholar 

  67. Zhong J, Scholz T, Yau ACY, Guerard S, Hüffmeier U, Burkhardt H, Holmdahl R. Mannan-induced Nos2 in macrophages enhances IL-17-driven psoriatic arthritis by innate lymphocytes. Sci Adv. 2018;4(5):eaas9864. https://doi.org/10.1126/sciadv.aas9864.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Brookes PS, Yoon Y, Robotham JL, Anders MW, Sheu S-S, Calcium. ATP, and ROS: a mitochondrial love-hate triangle. Am J Physiol Cell Physiol. 2004;287(4):C817–33. https://doi.org/10.1152/ajpcell.00139.2004.

    Article  CAS  PubMed  Google Scholar 

  69. Mills EL, Kelly B, Logan A, Costa ASH, Varma M, Bryant CE, Tourlomousis P, Däbritz JHM, Gottlieb E, Latorre I, et al. Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell. 2016. https://doi.org/10.1016/j.cell.2016.08.064.

    Article  PubMed  PubMed Central  Google Scholar 

  70. Harty LC, Biniecka M, O’Sullivan J, Fox E, Mulhall K, Veale DJ, Fearon U. Mitochondrial mutagenesis correlates with the local inflammatory environment in arthritis. Ann Rheum Dis. 2012;71(4):582–8. https://doi.org/10.1136/annrheumdis-2011-200245.

    Article  CAS  PubMed  Google Scholar 

  71. Schroder K, Tschopp J. The inflammasomes. Cell. 2010;140(6):821–32. https://doi.org/10.1016/j.cell.2010.01.040.

    Article  CAS  PubMed  Google Scholar 

  72. Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature. 2011;469(7329):221–5. https://doi.org/10.1038/nature09663.

    Article  CAS  PubMed  Google Scholar 

  73. Verma D, Fekri SZ, Sigurdardottir G, Bivik Eding C, Sandin C, Enerback C. Enhanced inflammasome activity in patients with psoriasis promotes systemic inflammation. J Invest Dermatol. 2021;141(3):586–95 e585. https://doi.org/10.1016/j.jid.2020.07.012.

    Article  CAS  PubMed  Google Scholar 

  74. Müller G, Lübow C, Weindl G. Lysosomotropic beta blockers induce oxidative stress and IL23A production in langerhans cells. Autophagy. 2020;16(8):1380–95. https://doi.org/10.1080/15548627.2019.1686728.

    Article  CAS  PubMed  Google Scholar 

  75. Campbell NK, Fitzgerald HK, Malara A, Hambly R, Sweeney CM, Kirby B, Fletcher JM, Dunne A. Naturally derived heme-oxygenase 1 inducers attenuate inflammatory responses in human dendritic cells and T cells: relevance for psoriasis treatment. Sci Rep. 2018;8(1):10287. https://doi.org/10.1038/s41598-018-28488-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Ghoreschi K, Brück J, Kellerer C, Deng C, Peng H, Rothfuss O, Hussain RZ, Gocke AR, Respa A, Glocova I, et al. Fumarates improve psoriasis and multiple sclerosis by inducing type II dendritic cells. J Exp Med. 2011;208(11):2291–303. https://doi.org/10.1084/jem.20100977.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Kirino M, Kirino Y, Takeno M, Nagashima Y, Takahashi K, Kobayashi M, Murakami S, Hirasawa T, Ueda A, Aihara M, et al. Heme oxygenase 1 attenuates the development of atopic dermatitis-like lesions in mice: implications for human disease. J Allergy Clin Immunol. 2008;122(2):290–7. https://doi.org/10.1016/j.jaci.2008.05.031. 297.e1-8 .

    Article  CAS  PubMed  Google Scholar 

  78. Mitterstiller A-M, Haschka D, Dichtl S, Nairz M, Demetz E, Talasz H, Soares MP, Einwallner E, Esterbauer H, Fang FC, et al. Heme oxygenase 1 controls early innate immune response of macrophages to Salmonella Typhimurium infection. Cell Microbiol. 2016;18(10):1374–89. https://doi.org/10.1111/cmi.12578.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Chau L-Y. Heme oxygenase-1: emerging target of cancer therapy. J Biomed Sci. 2015;22:22. https://doi.org/10.1186/s12929-015-0128-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Bambouskova M, Gorvel L, Lampropoulou V, Sergushichev A, Loginicheva E, Johnson K, Korenfeld D, Mathyer ME, Kim H, Huang L-H, et al. Electrophilic properties of itaconate and derivatives regulate the IκBζ-ATF3 inflammatory axis. Nature. 2018;556(7702):501–4. https://doi.org/10.1038/s41586-018-0052-z.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Yu X, Lan P, Hou X, Han Q, Lu N, Li T, Jiao C, Zhang J, Zhang C, Tian Z. HBV inhibits LPS-induced NLRP3 inflammasome activation and IL-1β production via suppressing the NF-κB pathway and ROS production. J Hepatol. 2017;66(4):693–702. https://doi.org/10.1016/j.jhep.2016.12.018.

    Article  CAS  PubMed  Google Scholar 

  82. Feng L, Song P, Xu F, Xu L, Shao F, Guo M, Huang W, Kong L, Wu X, Xu Q. Cis-khellactone inhibited the proinflammatory macrophages via promoting autophagy to ameliorate imiquimod-induced psoriasis. J Invest Dermatol. 2019;139(9):1946-1956 e1943. https://doi.org/10.1016/j.jid.2019.02.021.

    Article  CAS  PubMed  Google Scholar 

  83. Natsuaki Y, Egawa G, Nakamizo S, Ono S, Hanakawa S, Okada T, Kusuba N, Otsuka A, Kitoh A, Honda T, et al. Perivascular leukocyte clusters are essential for efficient activation of effector T cells in the skin. Nat Immunol. 2014;15(11):1064–9. https://doi.org/10.1038/ni.2992.

    Article  CAS  PubMed  Google Scholar 

  84. Thiam HR, Wong SL, Wagner DD, Waterman CM. Cellular mechanisms of NETosis. Annu Rev Cell Dev Biol. 2020;36:191–218. https://doi.org/10.1146/annurev-cellbio-020520-111016.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Lood C, Blanco LP, Purmalek MM, Carmona-Rivera C, De Ravin SS, Smith CK, Malech HL, Ledbetter JA, Elkon KB, Kaplan MJ. Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease. Nat Med. 2016;22(2):146–53. https://doi.org/10.1038/nm.4027.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Hamam HJ, Khan MA, Palaniyar N. Histone acetylation promotes neutrophil extracellular trap formation. Biomolecules. 2019;9(1):32. https://doi.org/10.3390/biom9010032.

    Article  CAS  PubMed Central  Google Scholar 

  87. Wójcik P, Garley M, Wroński A, Jabłońska E, Skrzydlewska E. Cannabidiol modifies the formation of NETs in neutrophils of psoriatic patients. Int J Mol Sci. 2020. https://doi.org/10.3390/ijms21186795.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Mutua V, Gershwin LJ. A review of neutrophil extracellular traps (NETs) in disease: potential anti- NETs therapeutics. Clin Rev Allergy Immunol. 2021;61(2):194–211. https://doi.org/10.1007/s12016-020-08804-7.

    Article  CAS  PubMed  Google Scholar 

  89. Uppala R, Tsoi LC, Harms PW, Wang B, Billi AC, Maverakis E, Michelle Kahlenberg J, Ward NL, Gudjonsson JE. “Autoinflammatory psoriasis”-genetics and biology of pustular psoriasis. Cell Mol Immunol. 2021;18(2):307–17. https://doi.org/10.1038/s41423-020-0519-3.

    Article  CAS  PubMed  Google Scholar 

  90. Haskamp S, Bruns H, Hahn M, Hoffmann M, Gregor A, Lohr S, Hahn J, Schauer C, Ringer M, Flamann C, et al. Myeloperoxidase modulates inflammation in generalized pustular psoriasis and additional rare pustular skin diseases. Am J Hum Genet. 2020;107(3):527–38. https://doi.org/10.1016/j.ajhg.2020.07.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Rodriguez-Rosales YA, Langereis JD, Gorris MAJ, van den Reek J, Fasse E, Netea MG, de Vries IJM, Gomez-Munoz L, van Cranenbroek B, Korber A, et al. Immunomodulatory aged neutrophils are augmented in blood and skin of psoriasis patients. J Allergy Clin Immunol. 2021;148(4):1030–40. https://doi.org/10.1016/j.jaci.2021.02.041.

    Article  CAS  PubMed  Google Scholar 

  92. Bacchetti T, Simonetti O, Ricotti F, Offidani A, Ferretti G. Plasma oxidation status and antioxidant capacity in psoriatic children. Arch Dermatol Res. 2020;312(1):33–9. https://doi.org/10.1007/s00403-019-01976-z.

    Article  CAS  PubMed  Google Scholar 

  93. Baek J-O, Byamba D, Wu WH, Kim T-G, Lee M-G. Assessment of an imiquimod-induced psoriatic mouse model in relation to oxidative stress. Arch Dermatol Res. 2012;304(9):699–706. https://doi.org/10.1007/s00403-012-1272-y.

    Article  CAS  PubMed  Google Scholar 

  94. Ganguly D, Haak S, Sisirak V, Reizis B. The role of dendritic cells in autoimmunity. Nat Rev Immunol. 2013;13(8):566–77. https://doi.org/10.1038/nri3477.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Mizuguchi S, Gotoh K, Nakashima Y, Setoyama D, Takata Y, Ohga S, Kang D. Mitochondrial reactive oxygen species are essential for the development of psoriatic inflammation. Front Immunol. 2021;12:714897. https://doi.org/10.3389/fimmu.2021.714897.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Al-Harbi NO, Nadeem A, Ahmad SF, Bakheet SA, El-Sherbeeny AM, Ibrahim KE, Alzahrani KS, Al-Harbi MM, Mahmood HM, Alqahtani F, et al. Therapeutic treatment with Ibrutinib attenuates imiquimod-induced psoriasis-like inflammation in mice through downregulation of oxidative and inflammatory mediators in neutrophils and dendritic cells. Eur J Pharmacol. 2020;877:173088. https://doi.org/10.1016/j.ejphar.2020.173088.

    Article  CAS  PubMed  Google Scholar 

  97. Kim H-J, Barajas B, Chan RC-F, Nel AE. Glutathione depletion inhibits dendritic cell maturation and delayed-type hypersensitivity: implications for systemic disease and immunosenescence. J Allergy Clin Immunol. 2007;119(5):1225–33.

    Article  CAS  Google Scholar 

  98. Amico D, Spadoni T, Rovinelli M, Serafini M, D’Amico G, Campelli N, Svegliati Baroni S, Gabrielli A. Intracellular free radical production by peripheral blood T lymphocytes from patients with systemic sclerosis: role of NADPH oxidase and ERK1/2. Arthritis Res Ther. 2015;17:68. https://doi.org/10.1186/s13075-015-0591-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Esmaeili B, Mansouri P, Doustimotlagh AH, Izad M. Redox imbalance and IL-17 responses in memory CD4 T cells from patients with psoriasis. Scand J Immunol. 2019;89(1):e12730. https://doi.org/10.1111/sji.12730.

    Article  CAS  PubMed  Google Scholar 

  100. Lai R, Xian D, Xiong X, Yang L, Song J, Zhong J. Proanthocyanidins: novel treatment for psoriasis that reduces oxidative stress and modulates Th17 and Treg cells. Redox Rep. 2018;23(1):130–5. https://doi.org/10.1080/13510002.2018.1462027.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Kim B-H, Oh I, Kim J-H, Jeon J-E, Jeon B, Shin J, Kim T-Y. Anti-inflammatory activity of compounds isolated from Astragalus sinicus L. in cytokine-induced keratinocytes and skin. Exp Mol Med. 2014;46:e87. https://doi.org/10.1038/emm.2013.157.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Bivik Eding C, Köhler I, Verma D, Sjögren F, Bamberg C, Karsten S, Pham T, Scobie M, Helleday T, Warpman Berglund U, et al. MTH1 inhibitors for the treatment of psoriasis. J Invest Dermatol. 2021;141(8):2037–48. https://doi.org/10.1016/j.jid.2021.01.026.

    Article  CAS  PubMed  Google Scholar 

  103. Cai Y, Shen X, Ding C, Qi C, Li K, Li X, Jala VR, Zhang H-g, Wang T, Zheng J, et al. Pivotal role of dermal IL-17-producing γδ T cells in skin inflammation. Immunity. 2011;35(4):596–610. https://doi.org/10.1016/j.immuni.2011.08.001.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Yang Q, Liu X, Liu Q, Guan Z, Luo J, Cao G, Cai R, Li Z, Xu Y, Wu Z, et al. Roles of mTORC1 and mTORC2 in controlling γδ T1 and γδ T17 differentiation and function. Cell Death Differ. 2020;27(7):2248–62. https://doi.org/10.1038/s41418-020-0500-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Bielecki P, Riesenfeld SJ, Hütter J-C, Torlai Triglia E, Kowalczyk MS, Ricardo-Gonzalez RR, Lian M, Amezcua Vesely MC, Kroehling L, Xu H, et al. Skin-resident innate lymphoid cells converge on a pathogenic effector state. Nature. 2021;592(7852):128–32. https://doi.org/10.1038/s41586-021-03188-w.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Ebbo M, Crinier A, Vély F, Vivier E. Innate lymphoid cells: major players in inflammatory diseases. Nat Rev Immunol. 2017;17(11):665–78. https://doi.org/10.1038/nri.2017.86.

    Article  CAS  PubMed  Google Scholar 

  107. Walker JA, Barlow JL, McKenzie ANJ. Innate lymphoid cells–how did we miss them? Nat Rev Immunol. 2013;13(2):75–87. https://doi.org/10.1038/nri3349.

    Article  CAS  PubMed  Google Scholar 

  108. Teunissen MBM, Munneke JM, Bernink JH, Spuls PI, Res PCM, Te Velde A, Cheuk S, Brouwer MWD, Menting SP, Eidsmo L, et al. Composition of innate lymphoid cell subsets in the human skin: enrichment of NCR(+) ILC3 in lesional skin and blood of psoriasis patients. J Invest Dermatol. 2014;134(9):2351–60. https://doi.org/10.1038/jid.2014.146.

    Article  CAS  PubMed  Google Scholar 

  109. Villanova F, Flutter B, Tosi I, Grys K, Sreeneebus H, Perera GK, Chapman A, Smith CH, Di Meglio P, Nestle FO. Characterization of innate lymphoid cells in human skin and blood demonstrates increase of NKp44 + ILC3 in psoriasis. J Invest Dermatol. 2014;134(4):984–91. https://doi.org/10.1038/jid.2013.477.

    Article  CAS  PubMed  Google Scholar 

  110. Ward NL, Umetsu DT. A new player on the psoriasis block: IL-17A- and IL-22-producing innate lymphoid cells. J Invest Dermatol. 2014;134(9):2305–7. https://doi.org/10.1038/jid.2014.216.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Pantelyushin S, Haak S, Ingold B, Kulig P, Heppner FL, Navarini AA, Becher B. Rorγt + innate lymphocytes and γδ T cells initiate psoriasiform plaque formation in mice. J Invest Dermatol. 2012;122(6):2252–6. https://doi.org/10.1172/JCI61862.

    Article  CAS  Google Scholar 

  112. Chan T-Y, Yen C-L, Huang Y-F, Lo P-C, Nigrovic PA, Cheng C-Y, Wang W-Z, Wu S-Y, Shieh C-C. Increased ILC3s associated with higher levels of IL-1β aggravates inflammatory arthritis in mice lacking phagocytic NADPH oxidase. Eur J Immunol. 2019;49(11):2063–73. https://doi.org/10.1002/eji.201948141.

    Article  CAS  PubMed  Google Scholar 

  113. von Bubnoff D, Andrès E, Hentges F, Bieber T, Michel T, Zimmer J. Natural killer cells in atopic and autoimmune diseases of the skin. J Allergy Clin Immunol. 2010;125(1):60–8. https://doi.org/10.1016/j.jaci.2009.11.020.

    Article  CAS  Google Scholar 

  114. Polese B, Zhang H, Thurairajah B, King IL. Innate lymphocytes in psoriasis. Front Immunol. 2020;11:242. https://doi.org/10.3389/fimmu.2020.00242.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Sato Y, Ogawa E, Okuyama R. Role of innate immune cells in psoriasis. Int J Mol Sci. 2020;21(18):6604. https://doi.org/10.3390/ijms21186604.

    Article  CAS  PubMed Central  Google Scholar 

  116. Kucuksezer UC, Aktas Cetin E, Esen F, Tahrali I, Akdeniz N, Gelmez MY, Deniz G. The role of natural killer cells in autoimmune diseases. Front Immunol. 2021;12:622306. https://doi.org/10.3389/fimmu.2021.622306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Gilhar A, Ullmann Y, Kerner H, Assy B, Shalaginov R, Serafimovich S, Kalish RS. Psoriasis is mediated by a cutaneous defect triggered by activated immunocytes: induction of psoriasis by cells with natural killer receptors. J Invest Dermatol. 2002;119(2):384–91. https://doi.org/10.1046/j.1523-1747.2002.01812.x.

    Article  CAS  PubMed  Google Scholar 

  118. Kono F, Honda T, Aini W, Manabe T, Haga H, Tsuruyama T. Interferon-γ/CCR5 expression in invariant natural killer T cells and CCL5 expression in capillary veins of dermal papillae correlate with development of psoriasis vulgaris. Br J Dermatol. 2014;170(5):1048–55. https://doi.org/10.1111/bjd.12812.

    Article  CAS  PubMed  Google Scholar 

  119. López-Soto A, Bravo-San Pedro JM, Kroemer G, Galluzzi L, Gonzalez S. Involvement of autophagy in NK cell development and function. Autophagy. 2017;13(3):633–6. https://doi.org/10.1080/15548627.2016.1274486.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Gaudenzio N, Laurent C, Valitutti S, Espinosa E. Human mast cells drive memory CD4 + T cells toward an inflammatory IL-22 + phenotype. J Allergy Clin Immunol. 2013;131(5):1400–7.e11. https://doi.org/10.1016/j.jaci.2013.01.029.

    Article  CAS  PubMed  Google Scholar 

  121. Mashiko S, Bouguermouh S, Rubio M, Baba N, Bissonnette R, Sarfati M. Human mast cells are major IL-22 producers in patients with psoriasis and atopic dermatitis. J Allergy Clin Immunol. 2015;136(2):351-9.e1. https://doi.org/10.1016/j.jaci.2015.01.033.

    Article  CAS  PubMed  Google Scholar 

  122. Shefler I, Pasmanik-Chor M, Kidron D, Mekori YA, Hershko AY. T cell-derived microvesicles induce mast cell production of IL-24: relevance to inflammatory skin diseases. J Allergy Clin Immunol. 2014; 133(1):217–24.e1-3. https://doi.org/10.1016/j.jaci.2013.04.035.

  123. Zhang Y, Shi Y, Lin J, Li X, Yang B, Zhou J. Immune cell infiltration analysis demonstrates excessive mast cell activation in psoriasis. Front Immunol. 2021;12:773280. https://doi.org/10.3389/fimmu.2021.773280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Tagen M, Elorza A, Kempuraj D, Boucher W, Kepley CL, Shirihai OS, Theoharides TC. Mitochondrial uncoupling protein 2 inhibits mast cell activation and reduces histamine content. J Immunol. 2009;183(10):6313–9. https://doi.org/10.4049/jimmunol.0803422.

    Article  CAS  PubMed  Google Scholar 

  125. Zhang B, Alysandratos K-D, Angelidou A, Asadi S, Sismanopoulos N, Delivanis D-A, Weng Z, Miniati A, Vasiadi M, Katsarou-Katsari A, et al. Human mast cell degranulation and preformed TNF secretion require mitochondrial translocation to exocytosis sites: relevance to atopic dermatitis. J Allergy Clin Immunol. 2011;127(6):1522–31.e8. https://doi.org/10.1016/j.jaci.2011.02.005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Chelombitko MA, Averina OA, Vasilyeva TV, Pletiushkina OY, Popova EN, Fedorov AV, Chernyak BV, Shishkina VS, Ilinskaya OP. Mitochondria-targeted antioxidant skq1 (10-(6´-plastoquinonyl)decyltriphenylphosphonium bromide) inhibits mast cell degranulation in vivo and in vitro. Biochem (Mosc). 2017;82(12):1493–503. https://doi.org/10.1134/S0006297917120082.

    Article  CAS  Google Scholar 

  127. Swindle EJ, Metcalfe DD. The role of reactive oxygen species and nitric oxide in mast cell-dependent inflammatory processes. Immunol Rev. 2007;217:186–205. https://doi.org/10.1111/j.1600-065X.2007.00513.x.

    Article  CAS  PubMed  Google Scholar 

  128. Herrmann A-K, Wüllner V, Moos S, Graf J, Chen J, Kieseier B, Kurschus FC, Albrecht P, Vangheluwe P, Methner A. Dimethyl fumarate alters intracellular ca handling in immune cells by redox-mediated pleiotropic effects. Free Radic Biol Med. 2019;141:338–47. https://doi.org/10.1016/j.freeradbiomed.2019.07.005.

    Article  CAS  PubMed  Google Scholar 

  129. Hoffmann JHO, Schaekel K, Hartl D, Enk AH, Hadaschik EN. Dimethyl fumarate modulates neutrophil extracellular trap formation in a glutathione- and superoxide-dependent manner. Br J Dermatol. 2018;178(1):207–14. https://doi.org/10.1111/bjd.15839.

    Article  CAS  PubMed  Google Scholar 

  130. Millar SA, Stone NL, Bellman ZD, Yates AS, England TJ, O’Sullivan SE. A systematic review of cannabidiol dosing in clinical populations. Br J Clin Pharmacol. 2019;85(9):1888–900. https://doi.org/10.1111/bcp.14038.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Atalay S, Jarocka-Karpowicz I, Skrzydlewska E. Antioxidative and anti-inflammatory properties of cannabidiol. Antioxid (Basel). 2019;9(1):21. https://doi.org/10.3390/antiox9010021.

    Article  CAS  Google Scholar 

  132. Rajesh M, Mukhopadhyay P, Bátkai S, Patel V, Saito K, Matsumoto S, Kashiwaya Y, Horváth B, Mukhopadhyay B, Becker L, et al. Cannabidiol attenuates cardiac dysfunction, oxidative stress, fibrosis, and inflammatory and cell death signaling pathways in diabetic cardiomyopathy. J Am Coll Cardiol. 2010;56(25):2115–25. https://doi.org/10.1016/j.jacc.2010.07.033.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Sakkas LI, Mavropoulos A, Zafiriou E, Roussaki-Schulze A, Bogdanos DP. The effect of apremilast on signal transduction and IL-10 production in CD39high regulatory B cells in patients with psoriatic arthritis. Mediterr J Rheumatol. 2018;29(1):59–61. https://doi.org/10.31138/mjr.29.1.59.

    Article  PubMed  PubMed Central  Google Scholar 

  134. Mavropoulos A, Zafiriou E, Simopoulou T, Brotis AG, Liaskos C, Roussaki-Schulze A, Katsiari CG, Bogdanos DP, Sakkas LI. Apremilast increases IL-10-producing regulatory B cells and decreases proinflammatory T cells and innate cells in psoriatic arthritis and psoriasis. Rheumatol (Oxford). 2019;58(12):2240–50. https://doi.org/10.1093/rheumatology/kez204.

    Article  CAS  Google Scholar 

  135. Schafer PH, Parton A, Capone L, Cedzik D, Brady H, Evans JF, Man HW, Muller GW, Stirling DI, Chopra R. Apremilast is a selective PDE4 inhibitor with regulatory effects on innate immunity. Cell Signal. 2014;26(9):2016–29. https://doi.org/10.1016/j.cellsig.2014.05.014.

    Article  CAS  PubMed  Google Scholar 

  136. Keating GM. Apremilast. A review in psoriasis and psoriatic arthritis. Drugs. 2017;77(4):459–72. https://doi.org/10.1007/s40265-017-0709-1.

    Article  CAS  PubMed  Google Scholar 

  137. Toda K, Tsukayama I, Nagasaki Y, Konoike Y, Tamenobu A, Ganeko N, Ito H, Kawakami Y, Takahashi Y, Miki Y, et al. Red-kerneled rice proanthocyanidin inhibits arachidonate 5-lipoxygenase and decreases psoriasis-like skin inflammation. Arch Biochem Biophys. 2020;689:108307. https://doi.org/10.1016/j.abb.2020.108307.

    Article  CAS  PubMed  Google Scholar 

  138. Patel RV, Clark LN, Lebwohl M, Weinberg JM. Treatments for psoriasis and the risk of malignancy. J Am Acad Dermatol. 2009;60(6):1001–17. https://doi.org/10.1016/j.jaad.2008.12.031.

    Article  PubMed  Google Scholar 

  139. Blaner WS, Shmarakov IO, Traber MG. Vitamin A and vitamin E: will the real antioxidant please stand up? Annu Rev Nutr. 2021;41:105–31. https://doi.org/10.1146/annurev-nutr-082018-124228.

    Article  CAS  PubMed  Google Scholar 

  140. Ghoreschi K, Balato A, Enerbäck C, Sabat R. Therapeutics targeting the IL-23 and IL-17 pathway in psoriasis. Lancet. 2021;397(10275):754–66. https://doi.org/10.1016/s0140-6736(21)00184-7.

    Article  CAS  PubMed  Google Scholar 

  141. Tiwari N, Osorio Blanco E, Sonzogni A, Esporrin-Ubieto D, Wang H, Calderon M. Nanocarriers for skin applications: where do we stand? Angew Chem Int Ed Engl. 2021;61(3):e202107960. https://doi.org/10.1002/anie.202107960.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. van Huizen AM, Menting SP, Gyulai R, Iversen L, van der Kraaij GE, Middelkamp-Hup MA, Warren RB, Spuls PI, Schejtman AA, Egeberg A, et al. International edelphi study to reach consensus on the methotrexate dosing regimen in patients with psoriasis. JAMA Dermatol. 2022;158(5):561–72. https://doi.org/10.1001/jamadermatol.2022.0434.

    Article  PubMed  Google Scholar 

  143. Wollina U, Tirant M, Vojvodic A, Lotti T. Treatment of psoriasis: novel approaches to topical delivery. Open Access Maced J Med Sci. 2019;7(18):3018–25. https://doi.org/10.3889/oamjms.2019.414.

    Article  PubMed  PubMed Central  Google Scholar 

  144. Dadwal A, Mishra N, Narang RK. Novel topical nanocarriers for treatment of psoriasis: an overview. Curr Pharm Des. 2018;24(33):3934–50. https://doi.org/10.2174/1381612824666181102151507.

    Article  CAS  PubMed  Google Scholar 

  145. Prausnitz MR, Langer R. Transdermal drug delivery. Nat Biotechnol. 2008;26(11):1261–8. https://doi.org/10.1038/nbt.1504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Anselmo AC, Gokarn Y, Mitragotri S. Non-invasive delivery strategies for biologics. Nat Rev Drug Discov. 2018;18(1):19–40. https://doi.org/10.1038/nrd.2018.183.

    Article  CAS  PubMed  Google Scholar 

  147. Lee YJ, Bae JH, Kang S-G, Cho SW, Chun D-I, Nam SM, Kim CH, Nam HS, Lee SH, Lee SH, et al. Pro-oxidant status and Nrf2 levels in psoriasis vulgaris skin tissues and dimethyl fumarate-treated HaCaT cells. Arch Pharmacal Res. 2017;40(9):1105–16. https://doi.org/10.1007/s12272-017-0955-5.

    Article  CAS  Google Scholar 

  148. Gesser B, Rasmussen MK, Iversen L. Dimethyl fumarate targets MSK1, RSK1, 2 and IKKα/β Kinases and regulates NF-κB /p65 activation in psoriasis: a demonstration of the effect on peripheral blood mononuclear cells, drawn from two patients with severe psoriasis before and after treatment with dimethyl fumarate. Psoriasis (Auckl). 2020;10:1–11. https://doi.org/10.2147/PTT.S234151.

    Article  CAS  Google Scholar 

  149. Kornberg MD, Bhargava P, Kim PM, Putluri V, Snowman AM, Putluri N, Calabresi PA, Snyder SH. Dimethyl fumarate targets GAPDH and aerobic glycolysis to modulate immunity. Science. 2018;360(6387):449–53. https://doi.org/10.1126/science.aan4665.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Landeck L, Asadullah K, Amasuno A, Pau-Charles I, Mrowietz U. Dimethyl fumarate (DMF) vs. monoethyl fumarate (MEF) salts for the treatment of plaque psoriasis: a review of clinical data. Arch Dermatol Res. 2018;310(6):475–83. https://doi.org/10.1007/s00403-018-1825-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Lin CY, Hsu CY, Elzoghby AO, Alalaiwe A, Hwang TL, Fang JY. Oleic acid as the active agent and lipid matrix in cilomilast-loaded nanocarriers to assist PDE4 inhibition of activated neutrophils for mitigating psoriasis-like lesions. Acta Biomater. 2019;90:350–61. https://doi.org/10.1016/j.actbio.2019.04.002.

    Article  CAS  PubMed  Google Scholar 

  152. Tripathi P, Kumar A, Jain PK, Patel JR. Carbomer gel bearing methotrexate loaded lipid nanocontainers shows improved topical delivery intended for effective management of psoriasis. Int J Biol Macromol. 2018;120(Pt A):1322–34. https://doi.org/10.1016/j.ijbiomac.2018.08.136.

    Article  CAS  PubMed  Google Scholar 

  153. Kim JY, Ahn J, Kim J, Choi M, Jeon H, Choe K, Lee DY, Kim P, Jon S. Nanoparticle-assisted transcutaneous delivery of a signal transducer and activator of transcription 3-inhibiting peptide ameliorates psoriasis-like skin inflammation. ACS Nano. 2018;12(7):6904–16. https://doi.org/10.1021/acsnano.8b02330.

    Article  CAS  PubMed  Google Scholar 

  154. Liu H, Kang RS, Bagnowski K, Yu JM, Radecki S, Daniel WL, Anderson BR, Nallagatla S, Schook A, Agarwal R, et al. Targeting the IL-17 receptor using liposomal spherical nucleic acids as topical therapy for psoriasis. J Invest Dermatol. 2020;140(2):435–44 e434. https://doi.org/10.1016/j.jid.2019.06.146.

    Article  CAS  PubMed  Google Scholar 

  155. Wu K, Wu X, Guo J, Jiao Y, Zhou C. Facile polyphenol-europium assembly enabled functional poly(l-lactic acid) nanofiber mats with enhanced antioxidation and angiogenesis for accelerated wound healing. Adv Healthc Mater. 2021;10(19):e2100793. https://doi.org/10.1002/adhm.202100793.

    Article  CAS  PubMed  Google Scholar 

  156. Shah PP, Desai PR, Patel AR, Singh MS. Skin permeating nanogel for the cutaneous co-delivery of two anti-inflammatory drugs. Biomaterials. 2012;33(5):1607–17. https://doi.org/10.1016/j.biomaterials.2011.11.011.

    Article  CAS  PubMed  Google Scholar 

  157. Yan Y, Liang H, Liu X, Liu L, Chen Y. Topical cationic hairy particles targeting cell free DNA in dermis enhance treatment of psoriasis. Biomaterials. 2021;276:121027. https://doi.org/10.1016/j.biomaterials.2021.121027.

    Article  CAS  PubMed  Google Scholar 

  158. Ozcan A, Sahin D, Impellizzieri D, Nguyen TT, Hafner J, Yawalkar N, Kurzbach D, Tan G, Akdis CA, Nilsson J, et al. Nanoparticle-coupled topical methotrexate can normalize immune responses and induce tissue remodeling in psoriasis. J Invest Dermatol. 2020;140(5):1003–14 e1008. https://doi.org/10.1016/j.jid.2019.09.018.

    Article  CAS  PubMed  Google Scholar 

  159. Han R, Ho LWC, Bai Q, Chan CKW, Lee LKC, Choi PC-L, Choi CHJ. Alkyl-terminated gold nanoparticles as a self-therapeutic treatment for psoriasis. Nano Lett. 2021;21(20):8723–33. https://doi.org/10.1021/acs.nanolett.1c02899.

    Article  CAS  PubMed  Google Scholar 

  160. Keum H, Kim TW, Kim Y, Seo C, Son Y, Kim J, Kim D, Jung W, Whang C-H, Jon S. Bilirubin nanomedicine alleviates psoriatic skin inflammation by reducing oxidative stress and suppressing pathogenic signaling. J Control Release. 2020;325:359–69. https://doi.org/10.1016/j.jconrel.2020.07.015.

    Article  CAS  PubMed  Google Scholar 

  161. Lee Y, Kim H, Kang S, Lee J, Park J, Jon S. Bilirubin nanoparticles as a nanomedicine for anti-inflammation therapy. Angew Chem Int Ed. 2016;55(26):7460–3. https://doi.org/10.1002/anie.201602525.

    Article  CAS  Google Scholar 

  162. Sun H, Zhao Y, Zhang P, Zhai S, Li W, Cui J. Transcutaneous delivery of mung bean-derived nanoparticles for amelioration of psoriasis-like skin inflammation. Nanoscale. 2022;14(8):3040–8. https://doi.org/10.1039/d1nr08229a.

    Article  CAS  PubMed  Google Scholar 

  163. Lopes Rocha Correa V, Assis Martins J, Ribeiro de Souza T, de Castro Nunes Rincon G, Pacheco Miguel M, Borges de Menezes L. Correa Amaral A. Melatonin loaded lecithin-chitosan nanoparticles improved the wound healing in diabetic rats. Int J Biol Macromol. 2020;162:1465–75. https://doi.org/10.1016/j.ijbiomac.2020.08.027.

    Article  CAS  PubMed  Google Scholar 

  164. Liang Y, He J, Guo B. Functional hydrogels as wound dressing to enhance wound healing. ACS Nano. 2021. https://doi.org/10.1021/acsnano.1c04206.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Yan X, Fang WW, Xue J, Sun TC, Dong L, Zha Z, Qian H, Song YH, Zhang M, Gong X, et al. Thermoresponsive in situ forming hydrogel with sol-gel irreversibility for effective methicillin-resistant staphylococcus aureus infected wound healing. ACS Nano. 2019;13(9):10074–84. https://doi.org/10.1021/acsnano.9b02845.

    Article  CAS  PubMed  Google Scholar 

  166. Gan D, Xing W, Jiang L, Fang J, Zhao C, Ren F, Fang L, Wang K, Lu X. Plant-inspired adhesive and tough hydrogel based on ag-lignin nanoparticles-triggered dynamic redox catechol chemistry. Nat Commun. 2019;10(1):1487. https://doi.org/10.1038/s41467-019-09351-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Batheja P, Sheihet L, Kohn J, Singer AJ, Michniak-Kohn B. Topical drug delivery by a polymeric nanosphere gel: formulation optimization and in vitro and in vivo skin distribution studies. J Control Release. 2011;149(2):159–67. https://doi.org/10.1016/j.jconrel.2010.10.005.

    Article  CAS  PubMed  Google Scholar 

  168. Wan T, Pan Q, Ping Y. Microneedle-assisted genome editing: a transdermal strategy of targeting by CRISPR-Cas9 for synergistic therapy of inflammatory skin disorders. Sci Adv. 2021;7(11):eabe2888. https://doi.org/10.1126/sciadv.abe2888.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Ye Y, Yu J, Wen D, Kahkoska AR, Gu Z. Polymeric microneedles for transdermal protein delivery. Adv Drug Deliv Rev. 2018;127:106–18. https://doi.org/10.1016/j.addr.2018.01.015.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Yang D, Chen M, Sun Y, Jin Y, Lu C, Pan X, Quan G, Wu C. Microneedle-mediated transdermal drug delivery for treating diverse skin diseases. Acta Biomater. 2021;121:119–33. https://doi.org/10.1016/j.actbio.2020.12.004.

    Article  CAS  PubMed  Google Scholar 

  171. Ni D, Wei H, Chen W, Bao Q, Rosenkrans ZT, Barnhart TE, Ferreira CA, Wang Y, Yao H, Sun T, et al. Ceria nanoparticles meet hepatic ischemia-reperfusion injury: the perfect imperfection. Adv Mater. 2019;31(40):e1902956. https://doi.org/10.1002/adma.201902956.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Yu H, Jin F, Liu D, Shu G, Wang X, Qi J, Sun M, Yang P, Jiang S, Ying X, et al. Ros-responsive nano-drug delivery system combining mitochondria-targeting ceria nanoparticles with atorvastatin for acute kidney injury. Theranostics. 2020;10(5):2342–57. https://doi.org/10.7150/thno.40395.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Weng Q, Sun H, Fang C, Xia F, Liao H, Lee J, Wang J, Xie A, Ren J, Guo X, et al. Catalytic activity tunable ceria nanoparticles prevent chemotherapy-induced acute kidney injury without interference with chemotherapeutics. Nat Commun. 2021;12(1):1436. https://doi.org/10.1038/s41467-021-21714-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Zeng F, Wu Y, Li X, Ge X, Guo Q, Lou X, Cao Z, Hu B, Long NJ, Mao Y, et al. Custom-made ceria nanoparticles show a neuroprotective effect by modulating phenotypic polarization of the microglia. Angew Chem Int Ed Engl. 2018;57(20):5808–12. https://doi.org/10.1002/anie.201802309.

    Article  CAS  PubMed  Google Scholar 

  175. Kwon HJ, Cha M-Y, Kim D, Kim DK, Soh M, Shin K, Hyeon T, Mook-Jung I. Mitochondria-targeting ceria nanoparticles as antioxidants for alzheimer’s disease. ACS Nano. 2016;10(2):2860–70. https://doi.org/10.1021/acsnano.5b08045.

    Article  CAS  PubMed  Google Scholar 

  176. Kim J, Kim HY, Song SY, Go SH, Sohn HS, Baik S, Soh M, Kim K, Kim D, Kim HC, et al. Synergistic oxygen generation and reactive oxygen species scavenging by manganese ferrite/ceria co-decorated nanoparticles for rheumatoid arthritis treatment. ACS Nano. 2019;13(3):3206–17. https://doi.org/10.1021/acsnano.8b08785.

    Article  CAS  PubMed  Google Scholar 

  177. Kwon HJ, Kim D, Seo K, Kim YG, Han SI, Kang T, Soh M, Hyeon T. Ceria nanoparticle systems for selective scavenging of mitochondrial, intracellular, and extracellular reactive oxygen species in parkinson’s disease. Angew Chem Int Ed Engl. 2018;57(30):9408–12. https://doi.org/10.1002/anie.201805052.

    Article  CAS  PubMed  Google Scholar 

  178. Wu L, Liu G, Wang W, Liu R, Liao L, Cheng N, Li W, Zhang W, Ding D. Cyclodextrin-modified ceo2 nanoparticles as a multifunctional nanozyme for combinational therapy of psoriasis. Int J Nanomedicine. 2020;15:2515–27. https://doi.org/10.2147/IJN.S246783.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Li J, Chen L, Xu X, Fan Y, Xue X, Shen M, Shi X. Targeted combination of antioxidative and anti-inflammatory therapy of rheumatoid arthritis using multifunctional dendrimer-entrapped gold nanoparticles as a platform. Small. 2020;16(49):e2005661. https://doi.org/10.1002/smll.202005661.

    Article  CAS  PubMed  Google Scholar 

  180. Zhang D-Y, Tu T, Younis MR, Zhu KS, Liu H, Lei S, Qu J, Lin J, Huang P. Clinically translatable gold nanozymes with broad spectrum antioxidant and anti-inflammatory activity for alleviating acute kidney injury. Theranostics. 2021;11(20):9904–17. https://doi.org/10.7150/thno.66518.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Moyano DF, Liu Y, Ayaz F, Hou S, Puangploy P, Duncan B, Osborne BA, Rotello VM. Immunomodulatory effects of coated gold nanoparticles in LPS-stimulated and murine model systems. Chem. 2016;1(2):320–7. https://doi.org/10.1016/j.chempr.2016.07.007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Nemati H, Ghahramani M-H, Faridi-Majidi R, Izadi B, Bahrami G, Madani S-H, Tavoosidana G. Using siRNA-based spherical nucleic acid nanoparticle conjugates for gene regulation in psoriasis. J Control Release. 2017;268:259–68. https://doi.org/10.1016/j.jconrel.2017.10.034.

    Article  CAS  PubMed  Google Scholar 

  183. Ninan N, Goswami N, Vasilev K. The impact of engineered silver nanomaterials on the immune system. Nanomaterials (Basel). 2020;10(5):967. https://doi.org/10.3390/nano10050967.

    Article  CAS  Google Scholar 

  184. Yang Y, Guo L, Wang Z, Liu P, Liu X, Ding J, Zhou W. Targeted silver nanoparticles for rheumatoid arthritis therapy via macrophage apoptosis and re-polarization. Biomaterials. 2021;264:120390. https://doi.org/10.1016/j.biomaterials.2020.120390.

    Article  CAS  PubMed  Google Scholar 

  185. Chen Y, Guan M, Ren R, Gao C, Cheng H, Li Y, Gao B, Wei Y, Fu J, Sun J, et al. Improved immunoregulation of ultra-low-dose silver nanoparticle-loaded TiO2 nanotubes via M2 macrophage polarization by regulating GLUT1 and autophagy. Int J Nanomedicine. 2020;15:2011–26. https://doi.org/10.2147/IJN.S242919.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Rao K, Roome T, Aziz S, Razzak A, Abbas G, Imran M, Jabri T, Gul J, Hussain M, Sikandar B, et al. Bergenin loaded gum xanthan stabilized silver nanoparticles suppress synovial inflammation through modulation of the immune response and oxidative stress in adjuvant induced arthritic rats. J Mater Chem B. 2018;6(27):4486–501. https://doi.org/10.1039/c8tb00672e.

    Article  CAS  PubMed  Google Scholar 

  187. Choudhury H, Pandey M, Lim YQ, Low CY, Lee CT, Marilyn TCL, Loh HS, Lim YP, Lee CF, Bhattamishra SK, et al. Silver nanoparticles: advanced and promising technology in diabetic wound therapy. Mater Sci Eng C Mater Biol Appl. 2020;112:110925. https://doi.org/10.1016/j.msec.2020.110925.

    Article  CAS  PubMed  Google Scholar 

  188. Crisan D, Scharffetter-Kochanek K, Crisan M, Schatz S, Hainzl A, Olenic L, Filip A, Schneider LA, Sindrilaru A. Topical silver and gold nanoparticles complexed with cornus mas suppress inflammation in human psoriasis plaques by inhibiting NF-κB activity. Exp Dermatol. 2018;27(10):1166–9. https://doi.org/10.1111/exd.13707.

    Article  CAS  PubMed  Google Scholar 

  189. Xu J, Chen H, Chu Z, Li Z, Chen B, Sun J, Lai W, Ma Y, He Y, Qian H, et al. A multifunctional composite hydrogel as an intrinsic and extrinsic coregulator for enhanced therapeutic efficacy for psoriasis. J Nanobiotechnol. 2022;20(1):155. https://doi.org/10.1186/s12951-022-01368-y.

    Article  CAS  Google Scholar 

  190. Qindeel M, Khan D, Ahmed N, Khan S, Asim Ur R. Surfactant-free, self-assembled nanomicelles-based transdermal hydrogel for safe and targeted delivery of methotrexate against rheumatoid arthritis. ACS Nano. 2020;14(4):4662–81. https://doi.org/10.1021/acsnano.0c00364.

    Article  CAS  PubMed  Google Scholar 

  191. Nguyen DN, Roth TL, Li PJ, Chen PA, Apathy R, Mamedov MR, Vo LT, Tobin VR, Goodman D, Shifrut E, et al. Polymer-stabilized Cas9 nanoparticles and modified repair templates increase genome editing efficiency. Nat Biotechnol. 2020;38(1):44–9. https://doi.org/10.1038/s41587-019-0325-6.

    Article  CAS  PubMed  Google Scholar 

  192. Lee K, Conboy M, Park HM, Jiang F, Kim HJ, Dewitt MA, Mackley VA, Chang K, Rao A, Skinner C, et al. Nanoparticle delivery of Cas9 ribonucleoprotein and donor DNA in vivo induces homology-directed DNA repair. Nat Biomed Eng. 2017;1:889–901. https://doi.org/10.1038/s41551-017-0137-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Peng B, Liang H, Li Y, Dong C, Shen J, Mao HQ, Leong KW, Chen Y, Liu L. Tuned cationic dendronized polymer: molecular scavenger for rheumatoid arthritis treatment. Angew Chem Int Ed Engl. 2019;58(13):4254–8. https://doi.org/10.1002/anie.201813362.

    Article  CAS  PubMed  Google Scholar 

  194. Liang H, Peng B, Dong C, Liu L, Mao J, Wei S, Wang X, Xu H, Shen J, Mao H-Q, et al. Cationic nanoparticle as an inhibitor of cell-free DNA-induced inflammation. Nat Commun. 2018;9(1):4291. https://doi.org/10.1038/s41467-018-06603-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Coimbra S, Catarino C, Costa E, Oliveira H, Figueiredo A, Rocha-Pereira P, Santos-Silva A. Circulating cell-free DNA levels in portuguese patients with psoriasis vulgaris according to severity and therapy. Br J Dermatol. 2014;170(4):939–42. https://doi.org/10.1111/bjd.12738.

    Article  CAS  PubMed  Google Scholar 

  196. Mondelo-Macía P, Castro-Santos P, Castillo-García A, Muinelo-Romay L, Diaz-Peña R. Circulating free DNA and its emerging role in autoimmune diseases. J Pers Med. 2021;11(2):151. https://doi.org/10.3390/jpm11020151.

    Article  PubMed  PubMed Central  Google Scholar 

  197. Liang H, Yan Y, Wu J, Ge X, Wei L, Liu L, Chen Y. Topical nanoparticles interfering with the DNA-LL37 complex to alleviate psoriatic inflammation in mice and monkeys. Sci Adv. 2020;6(31):eabb5274. https://doi.org/10.1126/sciadv.abb5274.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Ragothaman M, Kannan Villalan A, Dhanasekaran A, Palanisamy T. Bio-hybrid hydrogel comprising collagen-capped silver nanoparticles and melatonin for accelerated tissue regeneration in skin defects. Mater Sci Eng C Mater Biol Appl. 2021;128:112328. https://doi.org/10.1016/j.msec.2021.112328.

    Article  CAS  PubMed  Google Scholar 

  199. Chitimus DM, Popescu MR, Voiculescu SE, Panaitescu AM, Pavel B, Zagrean L, Zagrean A-M. Melatonin’s impact on antioxidative and anti-inflammatory reprogramming in homeostasis and disease. Biomolecules. 2020;10(9):1211. https://doi.org/10.3390/biom10091211.

    Article  CAS  PubMed Central  Google Scholar 

  200. Slominski A, Fischer TW, Zmijewski MA, Wortsman J, Semak I, Zbytek B, Slominski RM, Tobin DJ. On the role of melatonin in skin physiology and pathology. Endocrine. 2005;27(2):137–48. https://doi.org/10.1385/ENDO:27:2:137.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Scuderi SA, Cucinotta L, Filippone A, Lanza M, Campolo M, Paterniti I, Esposito E. Effect of melatonin on psoriatic phenotype in human reconstructed skin model. Biomedicines. 2022;10(4):752. https://doi.org/10.3390/biomedicines10040752.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Slominski AT, Zmijewski MA, Semak I, Kim T-K, Janjetovic Z, Slominski RM, Zmijewski JW. Melatonin, mitochondria, and the skin. Cell Mol Life Sci. 2017;74(21):3913–25. https://doi.org/10.1007/s00018-017-2617-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Mo C, Lu L, Liu D, Wei K. Development of erianin-loaded dendritic mesoporous silica nanospheres with pro-apoptotic effects and enhanced topical delivery. J Nanobiotechnol. 2020;18(1):55. https://doi.org/10.1186/s12951-020-00608-3.

    Article  CAS  Google Scholar 

  204. Damiani G, Pacifico A, Linder DM, Pigatto PDM, Conic R, Grada A, Bragazzi NL. Nanodermatology-based solutions for psoriasis: state-of-the art and future prospects. Dermatol Ther. 2019;32(6):e13113. https://doi.org/10.1111/dth.13113.

    Article  PubMed  Google Scholar 

  205. Khezri K, Saeedi M, Maleki Dizaj S. Application of nanoparticles in percutaneous delivery of active ingredients in cosmetic preparations. Biomed Pharmacother. 2018;106:1499–505. https://doi.org/10.1016/j.biopha.2018.07.084.

    Article  CAS  PubMed  Google Scholar 

  206. Kang N-W, Kim M-H, Sohn S-Y, Kim K-T, Park J-H, Lee S-Y, Lee J-Y, Kim D-D. Curcumin-loaded lipid-hybridized cellulose nanofiber film ameliorates imiquimod-induced psoriasis-like dermatitis in mice. Biomaterials. 2018;182:245–58. https://doi.org/10.1016/j.biomaterials.2018.08.030.

    Article  CAS  PubMed  Google Scholar 

  207. Yu F, Zhang Y, Yang C, Li F, Qiu B, Ding W. Enhanced transdermal efficiency of curcumin-loaded peptide-modified liposomes for highly effective antipsoriatic therapy. J Mater Chem B. 2021;9(24):4846–56. https://doi.org/10.1039/d1tb00557j.

    Article  CAS  PubMed  Google Scholar 

  208. Suzuki IL, de Araujo MM, Bagnato VS, Bentley MVLB. TNFα siRNA delivery by nanoparticles and photochemical internalization for psoriasis topical therapy. J Control Release. 2021;338:316–29. https://doi.org/10.1016/j.jconrel.2021.08.039.

    Article  CAS  PubMed  Google Scholar 

  209. Zhang Y, Xia Q, Li Y, He Z, Li Z, Guo T, Wu Z, Feng N. CD44 assists the topical anti-psoriatic efficacy of curcumin-loaded hyaluronan-modified ethosomes: a new strategy for clustering drug in inflammatory skin. Theranostics. 2019;9(1):48–64. https://doi.org/10.7150/thno.29715.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Tang F, Li L, Chen D. Mesoporous silica nanoparticles: synthesis, biocompatibility and drug delivery. Adv Mater. 2012;24(12):1504–34. https://doi.org/10.1002/adma.201104763.

    Article  CAS  PubMed  Google Scholar 

  211. Mora-Raimundo P, Lozano D, Benito M, Mulero F, Manzano M, Vallet-Regí M. Osteoporosis remission and new bone formation with mesoporous silica nanoparticles. Adv Sci. 2021;8(16):e2101107. https://doi.org/10.1002/advs.202101107.

    Article  CAS  Google Scholar 

  212. Pham LM, Kim E-C, Ou W, Phung CD, Nguyen TT, Pham TT, Poudel K, Gautam M, Nguyen HT, Jeong J-H, et al. Targeting and clearance of senescent foamy macrophages and senescent endothelial cells by antibody-functionalized mesoporous silica nanoparticles for alleviating aorta atherosclerosis. Biomaterials. 2021;269:120677. https://doi.org/10.1016/j.biomaterials.2021.120677.

    Article  CAS  PubMed  Google Scholar 

  213. Mai Y, Ouyang Y, Yu M, Qin Y, Girardi M, Saltzman WM, Cocco E, Zhao C, Yu L, Jia Y, et al. Topical formulation based on disease-specific nanoparticles for single-dose cure of psoriasis. J Control Release. 2022;349:354–66. https://doi.org/10.1016/j.jconrel.2022.07.006.

    Article  CAS  PubMed  Google Scholar 

  214. Singh AP, Biswas A, Shukla A, Maiti P. Targeted therapy in chronic diseases using nanomaterial-based drug delivery vehicles. Signal Transduct Target Ther. 2019;4:33. https://doi.org/10.1038/s41392-019-0068-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Shravanth SH, Osmani RAM, Anupama LJS, Rahamathulla VP, Gangadharappa M. HV. Microneedles-based drug delivery for the treatment of psoriasis. J Drug Deliv Sci Technol. 2021;64:102668. https://doi.org/10.1016/j.jddst.2021.102668.

    Article  CAS  Google Scholar 

  216. Jing Q, Ruan H, Li J, Wang Z, Pei L, Hu H, He Z, Wu T, Ruan S, Guo T, et al. Keratinocyte membrane-mediated nanodelivery system with dissolving microneedles for targeted therapy of skin diseases. Biomaterials. 2021;278:121142. https://doi.org/10.1016/j.biomaterials.2021.121142.

    Article  CAS  PubMed  Google Scholar 

  217. Kharaziha M, Baidya A, Annabi N. Rational design of immunomodulatory hydrogels for chronic wound healing. Adv Mater. 2021;33(39):e2100176. https://doi.org/10.1002/adma.202100176.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  218. Gong C, Wu Q, Wang Y, Zhang D, Luo F, Zhao X, Wei Y, Qian Z. A biodegradable hydrogel system containing curcumin encapsulated in micelles for cutaneous wound healing. Biomaterials. 2013;34(27):6377–87. https://doi.org/10.1016/j.biomaterials.2013.05.005.

    Article  CAS  PubMed  Google Scholar 

  219. Sun L, Liu Z, Wang L, Cun D, Tong HHY, Yan R, Chen X, Wang R, Zheng Y. Enhanced topical penetration, system exposure and anti-psoriasis activity of two particle-sized, curcumin-loaded plga nanoparticles in hydrogel. J Control Release. 2017;254:44–54. https://doi.org/10.1016/j.jconrel.2017.03.385.

    Article  CAS  PubMed  Google Scholar 

  220. Rana K, Pani T, Jha SK, Mehta D, Yadav P, Jain D, Pradhan MK, Mishra S, Kar R. G BR et al. Hydrogel-mediated topical delivery of steroids can effectively alleviate psoriasis attenuating the autoimmune responses. Nanoscale. 2022;14(10):3834–48. https://doi.org/10.1039/d1nr06001e.

    Article  CAS  PubMed  Google Scholar 

  221. Qiu F, Xi L, Chen S, Zhao Y, Wang Z, Zheng Y. Celastrol niosome hydrogel has anti-inflammatory effect on skin keratinocytes and circulation without systemic drug exposure in psoriasis mice. Int J Nanomedicine. 2021;16:6171–82. https://doi.org/10.2147/IJN.S323208.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (Nos. 82072862, and 82170572).

Author information

Authors and Affiliations

Authors

Contributions

JX defined the focus of the review and made major contributions to writing the manuscript. HC summarized the associated research studies. YX, FW and HQ guided the writing and revised the manuscript. All authors read and approved the final manuscript. JX and HC contributed equally to this work.

Corresponding authors

Correspondence to Haisheng Qian, Fei Wang or Yunsheng Xu.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing financial interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, J., Chen, H., Qian, H. et al. Advances in the modulation of ROS and transdermal administration for anti-psoriatic nanotherapies. J Nanobiotechnol 20, 448 (2022). https://doi.org/10.1186/s12951-022-01651-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12951-022-01651-y

Keywords