Abstract
Cell death is an essential process that plays a vital role in restoring and maintaining skin homeostasis. It supports recovery from acute injury and infection and regulates barrier function and immunity. Cell death can also provoke inflammatory responses. Loss of cell membrane integrity with lytic forms of cell death can incite inflammation due to the uncontrolled release of cell contents. Excessive or poorly regulated cell death is increasingly recognised as contributing to cutaneous inflammation. Therefore, drugs that inhibit cell death could be used therapeutically to treat certain inflammatory skin diseases. Programmes to develop such inhibitors are already underway. In this review, we outline the mechanisms of skin-associated cell death programmes; apoptosis, necroptosis, pyroptosis, NETosis, and the epidermal terminal differentiation programme, cornification. We discuss the evidence for their role in skin inflammation and disease and discuss therapeutic opportunities for targeting the cell death machinery.
Keywords: apoptosis, cell death, cornification, necroptosis, pyroptosis, skin
Introduction
The skin functions as our bodies first line of defence, an interactive barrier, protecting us from a constantly changing environment full of physical, chemical, and biological aggressors. Maintenance of barrier integrity, immune surveillance, and rapid response are fundamental to proper function. The epithelial barrier and immune cells orchestrate this multifaceted protection. Acute and chronic inflammatory skin diseases can arise due to abnormal over-reactions to the changing environment. Given the large variety of commensal microbial species that live on the skin surface and in hair follicles, a high degree of tolerance is required. Maintaining skin homeostasis is immensely complex and involves a highly regulated nexus of interactions between the different cellular compartments and the microbial communities (Box 1 – Structure and key cellular players in the skin).
Programmed cell death is essential for maintaining and restoring skin homeostasis
Programmed cell death (PCD) plays a vital role in forming and maintaining the cutaneous and mucosal barriers. Maintenance of the protective barrier depends on the balanced process of keratinocyte differentiation and cornification. While the process of cornification is programmed and results in the death of the cells, unlike other forms of PCD, the cornified cells become an integral part of the tissue [1].
PCD also supports the response to and recovery from acute injury and infection. Apoptosis of immune cells is critical for the resolution of acute inflammation associated with injury and infection. Inflammatory forms of cell death such as necroptosis, pyroptosis, and NETosis may have evolved to provide an opposing effect on inflammation. Induction of these cell death programmes is often in response to pathogen-associated molecular patterns (PAMPs) or to self-derived damage-associated molecular patterns (DAMPs), where enhanced inflammatory and immune responses are required to produce an adequate tissue response [2–5]. Less apparent is a potential physiological role for ferroptosis-associated inflammation. This form of cell death appears to be one of the oldest in an evolutionary sense, a way of eliminating cells that are not effectively managing the consequences of our oxygen-based metabolism. The inflammatory consequences of ferroptosis may have been repurposed to provide physiological roles in immune surveillance and tumour suppression [6].
Box 1. Structure and key cellular players of the skin
The skin can be broadly broken down into the epidermis, the dermis, and the hypodermis. The hypodermis is the subcutaneous fat layer, it consists of adipocytes in a loose connective tissue and contains larger blood vessels en-route to the dermis. The dermis consists of dermal fibroblasts in an extracellular matrix and forms a tough, irregular connective tissue. Structures such as hair follicles, sweat glands, sebaceous glands, nerves, and sensory receptors are present in this layer, as is blood supply and lymphatic vessels servicing and supporting the skin. Immune cell populations in the dermis, include macrophages, T-cells, mast cells, eosinophils, dendritic cells, and innate lymphoid cells (ILCs).
The outermost layer of the skin is the epidermis, a stratified squamous keratinised epithelium. In the epidermis the main populations of immune cells are CD8+ resident memory T-cells (TRM) and an epidermal resident population of Antigen Presenting Cells called Langerhans Cells. Keratinocytes are the main structural element of the epidermis; however, they also play a key immunomodulatory role. They are responsible for secreting antimicrobial peptides even in the absence of injury, and stimulating inflammation in response to DAMPs and PAMPs. They are necessarily one of the first participants in the inflammatory process and thus play an important role in antigen presentation and modulation of the immune response. However, as these cells are at the frontline, a degree of tolerance is required. Microorganisms colonise not only the surface of the skin but also into hair follicles, sweat glands and sebaceous glands, meaning that host exposure to microbial species is not limited to the dead cells making up the stratum corneum. Epidermal pathologies and activation of inflammatory signalling cascades in the epidermis are common in ISDs, suggesting a breakdown in immune tolerance, and highlighting the importance of keratinocytes in immune regulation in the skin.
The intertwining outcomes of cell death and inflammation
Activation of inflammatory signalling pathways is one of the earliest innate immune responses during injury and infection. Extracellular signals detected by cells in the skin initiate acute inflammation. Ligation of signalling molecules to membrane-bound receptors generates internal signalling cascades that can affect cell function by regulating genes and transcription factors. In some circumstances, these signalling molecules can also induce cell death programmes. These differing outcomes are entwined as cell death can also drive inflammatory signalling and immune cell recruitment. While the downstream consequences of inflammation can include the production of death receptor ligands leading to enhanced cell death. Lytic forms of cell death can promote inflammation due to the uncontrolled release of inflammatory cell contents [5,7–9], including pro-inflammatory DAMPs such as HMGB1 [10–16], IL33 [13,17,18], uric acid [19], S100 proteins [20], heat shock proteins (HSPs) [21], and nucleic acids/DNA [22–25]. The interplay between inflammation and cell death can contribute to inflammatory amplification loops required to generate adequate tissue and immune responses during injury or infection. However, when poorly regulated, these interactions can result in a vicious cycle leading to enhanced and ongoing inflammation and disease (Figure 1).
Figure 1. Interactions between inflammatory signals and cell death programmes can enhance both.
Acute inflammation is a healthy response to danger signals such as, trauma, stress, or infection. An inflammatory stimuli can prompt several outcomes for a cell. Most often the response will be survival signalling, which enhances the expression of proteins associated with proliferation and inflammation. In some circumstances, an inflammatory stimulus may initiate a cell death pathway. Apoptosis is typically non-inflammatory, and an important part of the resolution of acute inflammation in a tissue, however, apoptotic cells will proceed to late apoptosis when phagocytic clearance (efferocytosis) is delayed. Other lytic forms of programmed cell death, including necroptosis, pyroptosis, NETosis and ferroptosis, are thought to promote inflammation by releasing damage-associated molecular patterns (DAMPs), and other inflammatory mediators, into the extracellular environment. Inflammatory amplification loops play a physiological role in acute tissue inflammation in order to generate adequate tissue and immune responses. Lytic forms of cell death, including late apoptosis, can further enhance inflammatory amplification loops. Once an insult is dealt with, the resolution of inflammation is vital to restore cellular and tissue homeostasis. Excess inflammatory signalling can lead to ongoing amplification cycles that fail to resolve, resulting in chronic inflammation, enhanced cell death, tissue pathology and disease.
When both inflammation and cell death feature prominently in the pathology of a disease or phenotype, we must consider their relative contributions to pathogenesis. Is cell death driving inflammation, or is inflammation causing excessive cell death? Can we reduce the impact of both by targeting one or the other, or is a multi-pronged approach required? Here we discuss different types of PCD, the inflammatory consequences, and their association with inflammatory skin phenotypes in mice and humans.
Cell death modalities in skin function and dysfunction
PCD has many distinct forms, several of which are known to contribute to skin inflammation and disease. These include apoptosis, necroptosis, pyroptosis, NETosis, and ferroptosis. Additionally, while the nomenclature committee on cell death suggests that terminal differentiation should be considered distinct from the concept of PCD [26], cornification, which does result in the death of the cell, is integral to proper skin function; and management of alternative cell death programmes is essential to its proper execution. We will be discussing the role of each of these mechanisms in skin homeostasis and inflammation.
Cornification
One of the critical functions of the skin is as a mechanical barrier. Cornified keratinocytes in the epidermis primarily perform this function. The epidermis comprises several layers of keratinocytes — at the bottom is the stratum basale, a layer of proliferating keratinocytes attached firmly to a contiguous basement membrane separating the dermis from the epidermis [27]. It is from these basal keratinocytes that the cells of the upper layers are derived. Basal keratinocytes passage up through the layers of the epidermis in an ordered path, influenced by calcium gradients, while undergoing terminal differentiation. The first stage of differentiation forms the stratum spinosum, typically the thickest layer of the epidermis and the primary location of Langerhans cells (LCs). Further up is the stratum granulosum, which consists of flattened, nucleated, and highly granulated cells between which tight junctions form [1,27,28]. The cornification process occurs from this stage. A rapid decrease in intracellular pH facilitates denucleation [29] and complete DNA degradation by the keratinocyte-specific enzyme DNase1L2, leading to lysis of the nucleus and the formation of corneocytes [30]. Corneocytes are flattened, highly keratinised, non-living cells organised in layers to form the external protective layer, the stratum corneum. Corneocytes’ intracellular organelles and cell contents are replaced by a proteinaceous layer, ‘the cornified envelope,’ with cross-linking of proteins at the cell periphery [1,27,28,31,32]. As part of normal barrier function, the dead cells of the stratum corneum are continuously shed from the epidermal surface [33] (Figure 2). The rate of keratinocyte proliferation in the stratum basale must balance the rate of sloughing of the cornified cells of the stratum corneum to maintain epidermal homeostasis.
Figure 2. Overview of epidermal differentiation and cornification.
The epidermis consists of layers of keratinocytes at different stages of differentiation. Basal keratinocytes, distinguished by the expression of keratins 5 and 14, proliferate, producing new cells that will differentiate while passaging towards the skin surface. Cells of the spinous layer enter cell cycle arrest. They express typical markers of differentiation such as keratins 1 and 10, and caspase-14. NF-κB expression is also increased, inhibiting apoptosis. Pro-filaggrin and proteins of the epidermal differentiation complex are expressed as cells transition from the spinous to the granular layer. Cornification is initiated from the granular layer. Caspase-14 becomes active and contributes to the processing of filaggrin, and various anti-inflammatory proteins are expressed. Lamellar bodies are extruded, releasing serine proteases and lipids that will fill the inter-corneocyte space. Rapid intracellular acidification enables degradation of the nucleus and other organelles, at which point the cells would be considered dead. Keratins and other proteins are cross-linked by transglutaminases forming the cornified envelope. Eventually, proteases degrade the corneodemosomes that hold the corneocytes together allowing the cells to shed during desquamation.
Death by cornification does not end cell function
While no further gene expression is possible once the nucleus is degraded, functional proteins are retained and can be activated after cell death. Corneocytes are, therefore, not completely inert and can respond to environmental changes. Their responses, however, are pre-determined based on the suite of functional proteins, including keratins, ceremide, loricrin, and filaggrin preserved during cornification [33,34]. These proteins are responsible for giving strength and elasticity to the epidermis and ensuring imperviousness and water retention.
Potential danger signals inherent in cornification are actively suppressed
Cell death during cornification involves processes that have the potential to activate danger signals. Failure to effectively suppress inflammation can invoke further inflammatory signalling and risk disturbing the differentiation process of adjacent KCs. The tightly controlled gene expression programme for cornification, therefore, includes genes encoding proteins that mediate the degradation of potential DAMPs and regulate enzymatic activity [32]. For example, DNase1L2 and DNase2 completely degrade DNA upon nuclear envelope dissolution [30,35]. This is notably different from apoptosis, where DNA is degraded (fragmented) by DNases, but the fragments are retained within the apoptotic cell [36,37]. One reason for this difference may be the differing cell fates. Cornified cells are retained as part of the epidermis for days or weeks, while apoptotic cells are destined for elimination by phagocytes typically within hours of death, at which point they enter lysosomes and the cell remnants, including the fragmented DNA, are fully degraded [37,38].
Cornification requires a massive activation of epidermal proteases. Lamellar bodies released from the granular layer during cornification contain serine proteases that degrade corneodesmosomes and promote desquamation [31,32]. The SPINK5 gene encodes the serine protease inhibitor LEKTI which regulates the activity of these desquamation-involved proteases [39]. Truncating mutations in SPINK5 cause Netherton's Syndrome, a rare, autosomal recessive disorder characterised by congenital ichthyosis with defective cornification, bamboo hair, and severe atopic manifestation [39,40].
An interesting gene expression screen also identified proteins explicitly associated with keratinocyte terminal differentiation that do not appear to contribute to the cornification process at all but instead play a suppressive or anti-inflammatory role. These include modulators of pyroptosis (CARD18, NLRP10, PYDC1) and IL-1 family antagonists (IL-36RN, IL-37, IL-38) [32,41].
Apoptosis
Apoptosis is a highly regulated process that plays a vital role in restoring and maintaining skin homeostasis by eliminating unnecessary or damaged cells. It also plays a key role in hair follicle renewal and is critical to ensuring immunologic tolerance. There are two distinct apoptotic pathways, the intrinsic or mitochondrial pathway and the extrinsic or death receptor pathway. Although the initiating steps in intrinsic and extrinsic apoptosis differ, the pathways converge upon the activation of a family of proteases called caspases [26]. The process downstream of caspases is highly regulated and results in the packaging of cellular components into apoptotic bodies prior to cell degradation without the release of cell contents [42]. Thus, apoptosis is dogmatically considered to be non-inflammatory and immunologically silent.
Intrinsic apoptosis
Intrinsic apoptosis occurs in response to disruptions of cellular homeostasis, such as oxidative stress or DNA damage [26]. However, it is also induced in healthy cells as part of their natural life cycle. Intrinsic apoptosis is characterised by mitochondrial outer membrane permeabilisation (MOMP). Upon activation, pro-apoptotic members of the BCL2 family, such as BAX and BAK, oligomerise into pore-forming complexes on the mitochondrial outer membrane [43,44]. These pores enable the release of pro-apoptotic proteins, including cytochrome-c and the inhibitor of apoptosis protein (IAP) inhibitor Smac/DIABLO [45–47], into the cytosol. Cytosolic cytochrome-c associates with the adaptor protein APAF1 to form the apoptosome, a molecular platform that facilitates the activation of caspase-9, triggering the caspase cascade [26] (Figure 3A). While disruption of the mitochondrial membrane is enough to kill a cell, the apoptotic caspase cascade, which occurs well after the point of no return, functions to prevent type I interferon production [42,48] and ensures that DNA and other intracellular substrates with the potential to act as DAMPs are cleaved and inactivated [38,42,48,49].
Figure 3. Overview of intrinsic and extrinsic apoptosis.
(A) Intrinsic Apoptosis — intracellular stress activates pro-apoptotic members of the BCL2 family, BAX and BAK, which form pores in the mitochondrial outer membrane. Cytochrome-c is released into the cytosol where it binds to the adaptor protein APAF1 to promote the formation of the Apoptosome. This molecular platform facilitates caspase-9 activation, initiating the caspase cascade. Executioner caspases can then cleave hundreds of substrates limiting immunogenicity during apoptosis. Cytochrome-c is not the only protein released from the mitochondria upon membrane permeabilization. The IAP antagonist protein Smac/DIABLO is contained in the mitochondria and is released during intrinsic apoptosis. Smac/DIABLO antagonises XIAP, lifting the late-stage inhibition of caspases, and ensuring execution of the caspase cascade. (B) Extrinsic apoptosis can occur following death receptor ligation (Fas, TRAIL or TNF) and activation of the apoptotic caspase cascade, beginning with caspase-8. However, ligation of TNFR1 will preferentially activate pro-survival signals. TNF binding promotes the formation of complex I via recruitment of TRADD to the receptor's death domain. TRAF2, RIPK1 and the cIAPs can then be recruited to the receptor complex. The cIAPs recruit LUBAC, and mediate ubiquitination of RIPK1. This leads to the recruitment of a TAK1-containing complex, enabling subsequent recruitment of NEMO and the IKKs. Phosphorylation of RIPK1 via the IKKs and TAK1, prevents the formation of RIPK-dependent death complexes. Activation of canonical NF-κB can then proceed, promoting transcription of pro-inflammatory genes and genes encoding anti-apoptotic factors such as cFLIP. Ongoing activation of TNFR1 or defects in complex I regulatory components will promote dissociation and internalisation of the receptor complex to form a cytosolic death complex, known as complex II. The interaction of complex II components, enables oligomerisation and autoproteolytic cleavage of Caspase-8, initiating caspase-mediated apoptosis. Activated caspase-8 can also cleave BID to its truncated form (tBid) which can activate BAK initiating, intrinsic apoptosis. The subsequent release of Smac/DIABLO can lift the XIAP break on caspase activation ensuring completion of the death programme. Smac/DIABLO also antagonises cIAP1 and 2, thus, while intrinsic and extrinsic apoptosis operate independent of each other, they also each include mechanisms to promote activation of the other.
Defects in the regulation of intrinsic apoptosis can affect the hair follicle cycle, causing poor hair growth, premature greying, and alopecia [50]. Anti-apoptotic Bcl-2 is highly expressed in melanocyte stem cells, specifically protecting them from apoptosis during catagen. Bcl-2 deficiency in mice causes their apoptotic elimination resulting in premature hair greying [50,51]. Conditional knockout of Bcl-2 in mouse epidermis or systemic treatment with the Bcl-2 antagonist ABT-199/venetoclax during catagen causes selective loss of hair follicle-associated stem cells leading to disrupted hair follicle regeneration and delayed hair regrowth [52].
Extrinsic apoptosis
Extrinsic apoptosis is initiated upon detection of an extracellular signal, either cytokines produced by other cells locally or systemically or DAMPs or PAMPs that activate pattern recognition receptors (PRRs) [26,53]. Initiation is most often by ligation of death receptors, such as TNFR1, FAS, or TRAIL receptors 1 (TRAILR1/Death Receptor (DR)4) and 2 (TRAILR2/DR5), with their corresponding ligands (FASL, TRAIL, and TNF, respectively) [26] (Figure 3B). However, it can also be triggered upon activation of Toll-Like Receptors (TLRs) 3 and 4 via TIR domain-containing adapter-inducing interferon-β (TRIF) mediated receptor complex formation, usually during infection [53] (Figure 3B).
Death receptor–ligand binding initiates the formation and internalisation of death-inducing signalling complexes (DISCs). In Fas and TRAIL-mediated apoptosis, receptor activation enables binding of Fas-associated death domain (FADD) to the receptors’ own death domains. The death effector domain (DED) of FADD can then bind pro-caspase-8 triggering its autoproteolytic cleavage and oligomerisation to form active caspase-8, initiating the caspase cascade [54,55].
Activation of TNFR1 initiates recruitment of TNFR1-associated death domain protein (TRADD), RIPK1, and TRAF2 [56–58] to a membrane-bound signalling platform referred to as complex I. TRAF2, in turn, recruits the E3 ubiquitin ligases, cellular inhibitor of apoptosis proteins (cIAP)1 and cIAP2 [59]. The cIAPs conjugate K11, K48 and K63 ubiquitin to RIPK1, which sequesters RIPK1 at the receptor complex [60–62]. The K63 ubiquitin on RIPK1 acts as a scaffold for recruitment of the transforming growth factor-β-activated kinase 1 (TAK1)/TAK1-binding protein (TAB)1/2 complex and the IκB kinase (IKK) complex [58,63,64]. The cIAPs also recruit the linear ubiquitin-chain assembly complex (LUBAC) to complex I. LUBAC consists of three proteins, SHANK-associated RH domain-interacting protein (SHARPIN), hemeoxidized iron-regulatory protein 2 ubiquitin ligase-1 (HOIL-1), and HOIL-1-interacting protein (HOIP). LUBAC assembles linear ubiquitin chains on complex I components including RIPK1 and the IKK regulatory subunit, nuclear factor-kappa B (NF-κB) essential modulator (NEMO). This further recruits the NF-κB catalytic subunits IKK1/IKKα and IKK2/IKKβ [65–68] (Figure 3B).
Activated IKK mediates phosphorylation and degradation of Inhibitor of NF-κB (IκB) proteins resulting in the activation of NF-κB and expression of NF-κB dependent genes. These include genes encoding anti-apoptotic proteins such as the caspase-8 regulator, cellular FLICE-like inhibitory protein (cFLIP) [58,63]. cFLIP is structurally related to caspase-8 but lacks proteolytic activity. The presence of cFLIP constrains caspase-8 activation, limiting substrate cleavage to only a subset of proteins located within the complex, including RIPK1 (Figure 3B) [58,63]. An alternative cFLIP isoform (cFLIPlong) can prevent caspase-8 mediated substrate cleavage entirely, thereby blocking apoptosis but also, by failing to cleave RIPK1, promoting the formation of the necrosome (discussed later) [69].
The post-translational regulation of RIPK1 plays an essential role in shifting the response from cytokine production to cell death upon activation of TNFR1 [70–72]. Within complex I, TAK1 and the IKKs will phosphorylate RIPK1 preventing activation [73] and limiting downstream death signalling [74]. Reduced ubiquitylation of RIPK1 favours dissociation and internalisation of complex I, and recruitment of FADD, pro-caspase-8, and RIPK3, forming a RIPK1-dependent complex known as complex II (Figure 3B) [73].
The interaction of components in complex II, including activated RIPK1, prompts oligomerisation and autoproteolytic cleavage of pro-caspase-8 to its active form. Disruption of complex I components, such as TAK1, TRAF2, cIAPs, or LUBAC, affects post-translational regulation of RIPK1 [70–72] and reduces NF-κB activation resulting in low expression of cFLIP. Reduction in cFLIP levels leads to complete activation of caspase-8 [58,63] and initiation of the caspase cascade.
Caspase cascades prevent excess inflammation during intrinsic and extrinsic apoptosis
Both intrinsic and extrinsic apoptosis conclude with the initiation of a caspase cascade. Initiator caspases (e.g. caspase-8 and -9) cleave and activate executioner caspases (e.g. caspase-3, -6, and -7). X-linked inhibitor of apoptosis protein (XIAP) is a direct inhibitor of caspases-3, -7, and -9 and acts as a final break before the execution of apoptosis [75–77]. It is inhibited by the mitochondrial protein (Smac/DIABLO), which is released from the mitochondria upon the release of cytochrome-C during MOMP [46,78,79].
Once activated, the executioner caspases cleave hundreds of substrates, resulting in enzymatic degradation of organelles, DNA fragmentation, and membrane blebbing [42]. Activation of caspases instigates mechanisms that actively suppress potentially inflammatory responses. For example, the apoptotic caspase cascade functions to prevent type I interferon production during Bax/Bak dependent apoptosis [48], and caspases-3 and -7 have been shown to inactivate IL-33 [80]. Genomic DNA is cleaved into small fragments [37], and the prototypical DAMP, HMGB1, remains bound to chromatin [14] limiting its inflammatory potential until a late stage of apoptosis following DNA fragmentation [81]. Exposure of phosphatidylserines on the cell surface act as ‘find me’ and ‘eat me’ signals to attract phagocytes and prompt the rapid consumption of the dying cell in a process known as efferocytosis [82]. This, in theory, should limit activation of PRR pathways in surrounding cells, as it prevents the release of cell contents capable of provoking inflammation.
Apoptosis in normal skin function
Apoptosis plays an essential role in maintaining and restoring normal skin function. For example, hair follicles are a complex mini-organ of the skin that host the stem cell microenvironment. Maintenance of hair follicles is fundamental for skin homeostasis and hair growth and for efficient initiation of tissue repair [83,84]. Hair follicle renewal involves cycling through a growth phase (anagen), apoptosis-driven hair growth recession (catagen), a resting phase (telogen), and shedding (exogen) [84]. Defects in the regulation of intrinsic apoptosis can affect the hair follicle cycle, causing poor hair growth, premature greying, and alopecia [50]. Anti-apoptotic Bcl-2 is highly expressed in melanocyte stem cells, specifically protecting them from apoptosis during catagen. Bcl-2 deficiency in mice causes their apoptotic elimination resulting in premature hair greying [50,51]. Conditional knockout of Bcl-2 in mouse epidermis or systemic treatment with the Bcl-2 antagonist ABT-199/venetoclax during catagen causes selective loss of hair follicle-associated stem cells leading to disrupted hair follicle regeneration and delayed hair regrowth [52].
Defective apoptosis also leads to an impairment of negative selection in the thymus and the consequent persistence of autoreactive T-cells and B-cells that drive inflammation throughout the body, contributing to autoimmune disease. FAS polymorphisms that lead to defective apoptosis contribute to systemic lupus erythema (SLE) pathogenesis, a systemic disease that commonly presents with chronic inflammatory cutaneous manifestations [85].
In sunburn, UVB-mediated activation of p53 up-regulates pro-apoptotic proteins in basal keratinocytes leading to cell cycle arrest and epidermal apoptosis. This is critical for eliminating DNA-damaged cells to prevent skin carcinogenesis [86–88].
Apoptosis is also essential in the cutaneous wound repair process. Apoptosis of neutrophils and inflammatory cells is critical for the resolution of acute inflammation and can start as early as 12 h post-injury [89–91]. During tissue recovery, immune cell apoptosis and efferocytosis below the healing wound edge signal inflammation down-regulation and trigger proliferative responses in keratinocytes, prompting wound closure and re-epithelialisation [92,93]. Then during remodelling, myofibroblasts apoptosis is initiated to eliminate granulation tissue and restore pre-wound structure [90,91]. Defects in apoptosis can prolong inflammatory responses and delay tissue regeneration and remodelling, leading to poorly-healing wounds, fibrosis, and excessive scarring [90,92,94].
When apoptosis is not so silent
Millions of cells die by apoptosis in the average human every day. Apoptotic cell death must, therefore, operate in a non-inflammatory manner. The caspase cascade functions to minimise inflammation during apoptosis by preventing activation of inflammatory responses within the cell and reducing the immunogenic potential of cell contents [14,38,48]. However, the assumption that all apoptosis is inherently immunologically silent is an oversimplification. Rapid clearance of apoptotic cells is important for limiting immunogenicity. Efferocytosis typically occurs very early after the initiation of cell death, before membrane permeabilisation, thereby limiting the leakage of intracellular contents. Delayed engulfment allows cells to progress to late apoptosis. Late apoptotic cells will begin to lose membrane integrity resulting in the release of DAMPs and further inflammatory signalling [14,16,95–97].
Apoptosis in tissues can become inflammatory when excessive, as it pushes the limit of local phagocytes to clear the dying cells effectively. Indeed, this may be the function of late apoptotic loss of membrane integrity. Recent work identified NINJ1 as an important mediator of plasma membrane rupture in some forms of PCD. In its absence, BMDMs treated with the chemotherapeutic agents venetoclax or cisplatin to induce apoptosis had reduced indicators of membrane rupture despite cell non-viability, suggesting that the late apoptotic membrane permeabilisation is not simply the passive necrotic breakdown of a dead cell, but can be an active process [98]. The release of cell contents during late apoptosis can indicate at a tissue level the insufficiency of local phagocytes and send the message that systemic reinforcements are needed to clear apoptotic cells, while the previous processing of cellular components by caspases to limit immunogenicity avoids an excessive response. Unfortunately, in some circumstances, that inflammation may also drive further cell death, exacerbating the problem when efferocytosis is already insufficient. In such cases, the inflammatory situation may not be fully resolved without addressing the underlying problem by limiting excessive apoptosis.
Disposal of apoptotic cells in barrier tissues such as the skin can also pose unique problems. Rapid removal of dying cells, without a mechanism to seal the gap, may be more damaging to barrier integrity than retaining a dying cell in place for as long as possible. If the epidermal barrier is breached, commensal and pathogenic bacteria can invade, unleashing an intense inflammatory response. By analogy, a crumbling brick in a wall may function better than no brick at all. Retention of apoptotic keratinocytes can be seen in sunburn, where apoptosis leads to the formation of sun-burn cells (SBCs). These are clearly dead, with pyknotic nuclei and eosinophilic cytoplasm, but are retained in place following the execution of apoptosis, detectable up to 36 h after UVB exposure [99]. While retention of such cells may be useful in the short term, ongoing delays in engulfment will eventually result in late apoptosis and enhanced inflammation.
Excess epidermal apoptosis drives inflammation in mouse models
Despite its generally being an inflammation moderating force, several mouse models highlight the inflammatory potential of poorly regulated or excessive apoptosis in the skin. Differentiating epidermal KCs appear primed to die, and the active suppression of apoptosis is necessary to maintain barrier integrity. Complete loss of apoptosis inhibition is devastating, resulting in widespread loss of the epidermal barrier. Tamoxifen-induced epidermal loss of cFLIP or pharmacological depletion of the IAPs, cIAP1, cIAP2, and XIAP, result in a complete loss of apoptosis inhibition, causing widespread keratinocyte death and severe, acute dermatological disease resembling toxic epidermal necrolysis (TEN) [100,101]. In both the cFlip and IAP depletion models, the disease pathology is excessive apoptotic keratinocyte death, leading to epidermal necrosis.
The body of evidence supports the idea that reduced expression of NF-κB-dependent genes and thus reduced inhibition of apoptosis allow spontaneous KC apoptosis to occur. Genetic disruption of receptor signalling to NF-κB induces inflammatory skin phenotypes in mice. While there is a diversity of manifestations, including the age of onset and localisation, all are to some degree characterised by epidermal hyperplasia, keratinocyte cell death, infiltration by immune cells, and production of pro-inflammatory cytokines. Rapid elimination of apoptotic cells risks compromising barrier integrity, increasing exposure of metabolically active cells to PAMPs, resulting in increased inflammation. Retention, on the other hand, may support the maintenance of the barrier, but as cells progress to late apoptosis, cell lysis and release of DAMPs can also drive tissue inflammation.
There are several genetic models that demonstrate the inflammatory potential of epidermal apoptosis. Epidermal loss of cIAP1 combined with ubiquitous cIAP2 produced mice with severe neonatal skin inflammation and widespread epidermal apoptosis that was lethal by day 10 postpartum. The loss of one allele of Ripk1 limited lesion formation and significantly extended the lifespan of the mice [100].
Traf2 EKO mice develop epidermal hyperplasia and skin inflammation from around 10 weeks of age, which can be delayed by genetic deletion of Tnf [102]. The combined loss of mixed lineage kinase domain-like (MLKL) and caspase-8, but not MLKL alone, prevented TNF-dependent cutaneous disease, demonstrating that apoptotic cell death is a driver of the inflammatory phenotype.
Another genetic model demonstrating the inflammatory potential of epidermal apoptosis is the chronic proliferative dermatitis mutation (cpdm) in the LUBAC component, Sharpin. This spontaneous loss-of-function mutation results in a progressive dermatitis phenotype that shares clinical and histopathological features with chronic eczema and psoriasis [103,104]. The mutation results in reduced activation of NF-κB and AP1-dependent genes from TNF, CD40L, and TLR signalling pathways and sensitises cells to TNF-induced death [65,105,106]. Extensive genetic experiments have shown that SHARPIN deficient keratinocytes are sensitised to TNFR1 induced, caspase-8 mediated apoptosis and that it is this process that drives the dermatitis [104,107]. Recent work identified LCs as a potential cellular source of pathogenic TNF driving apoptotic cell death in the Sharpin mutant mice [108].
Necroptosis
In contrast with apoptosis, where regulatory mechanisms have evolved to limit inflammation, necroptosis occurs independent of caspases [26]. It causes permeabilisation of the plasma membrane and early cell lysis without protective mechanisms to limit immunogenicity. The process is highly inflammatory as it releases intracellular contents that function as DAMPs [9,109]. Necroptosis is triggered by many of the same stimuli as apoptosis, including death receptor ligation, DNA damage, and infection. The latter may be of particular importance as the lack of cell content processing, and thus the greater inflammatory potential of necroptosis, can promote a more robust tissue response. Something that may be required to effectively combat infection. The trade-off is an increased propensity towards excessive inflammation, with disruptions in regulation of necroptosis potentially predisposing towards the development of inflammatory disease.
TNF-mediated necroptosis
Complex II formation downstream of TNFR1 activates caspase-8 and causes apoptotic cell death. Activated caspase-8 also cleaves RIPK1 and potentially RIPK3 [110], preventing their oligomerisation and auto-phosphorylation within a second cytosolic complex called the necrosome. Necroptosis is, therefore, not only caspase-independent, missing the protective mechanisms of the caspase cascade, but also caspase inhibited. Complete disruption of caspase-8 activity or increased levels of RIPK3 and the pseudokinase, MLKL, can promote the formation of the necrosome. Within this complex, RIPK3 binds to the RHIM domain of RIPK1 and phosphorylation events stabilise the association and prompt activation of RIPK3 [111–113]. Uncleaved phospho-RIPK3 phosphorylates MLKL, which then oligomerises and translocates to the cell membrane. MLKL-mediated disruption of the plasma membrane results in the release of cell contents, cellular swelling and rupture and necroptotic cell death [9,114,115] (Figure 4).
Figure 4. Overview of necroptosis.
Downstream of TNFR1, following the formation and dissociation of complex II, activated caspase-8 will cleave RIPK1 and possibly RIPK3, preventing the formation of a second cytolosolic complex, the Necrosome. RIPK1 containing death complexes can also form downstream of FAS and TRAIL and the pattern recognition receptors. Complete inhibition of caspase-8 enables recruitment and phosphorylation of RIPK3 forming the necrosome. Necrosomes can also form independent of RIPK1 via complexes involving other RHIM domain-containing proteins such as ZBP1 upon detecting dsRNA within the cell, or TRIF via endosomal TLR3 or membrane-bound TLR4. Phosphorylated RIPK3 phosphorylates and activates MLKL. Activated MLKL oligomerises and translocates to the cell membrane, generating pores that result in the release of cell contents, cellular swelling and rupture, and the highly inflammatory necroptotic death of the cell.
The precise mechanisms by which oligomerised MLKL causes membrane permeabilisation is a subject of much enquiry but remains somewhat unclear. Two earlier studies suggested that MLKL oligomers could form cation channels facilitating changes in osmotic gradients leading to cellular swelling and eventual membrane rupture [116,117]. However, recent work on Ninj1 showed that osmotic swelling alone is insufficient to prompt membrane rupture at least in the case of the gasdermin D (GSDMD) pore (discussed later in pyroptosis) [98]. Higher order MLKL structures consistent with the formation of a membrane-spanning pore have not been identified. However, it has been reported that oligomerised pMLKL co-accumulates with tight-junction proteins to form plasma membrane ‘hot spots’ during epithelial cell necroptosis [9,118]. Once a threshold density of these spots is reached the membrane ruptures, resulting in necroptotic death. Interestingly, these pMLKL-tight-junction-associated hotspots accelerated necroptosis in neighbouring cells, prior to membrane rupture, suggesting that MLKL-mediated membrane damage can be propagated between cells so long as those cells are also primed to undergo necroptosis [118].
The direct cleavage of RIPK1 by caspase-8 was initially thought to be a mechanism to inhibit necroptosis. However, targeted mutation of RIPK1 to prevent caspase-8 mediated cleavage caused accumulation of RIPK1 and increased stability of the apoptotic death-inducing complex. The result was enhanced apoptosis rather than the formation of the necrosome [119–121]. This supports the idea of a hierarchy of responses in TNFR1 signalling. Signalling to NF-κB activation promotes cell survival and is generally the first response; however, ongoing cellular stress or targeting of complex I signalling components by, for example, infectious agents diverts signalling to initiate cell death [71,122,123]. Apoptosis is the preferred mechanism as it controls the release of cell contents and prevents excessive inflammation. Cleavage of RIPK1 inhibits its kinase activity, preventing necroptosis as hypothesised but does not affect its scaffold function as required for apoptosis. So long as it remains possible, the cell favours apoptosis over necroptosis even when kinase function is restored.
If apoptosis cannot proceed, then necroptosis is activated to ensure cell death occurs. In this case, the death of the cell is more important than the prevention of inflammatory consequences. Indeed, necroptotic cell death, despite being highly inflammatory, can reduce overall tissue inflammation by removing damaged or infected cells that might otherwise continue to produce large amounts of inflammatory mediators [100,124].
Alternative initiation mechanisms for necroptosis
While TNF-mediated necroptosis is the best understood necroptotic pathway, cytosolic complexes involving RIPK1 also occur downstream of TRAIL and FAS [125–127] as well as PRRs [128,129]. Necroptosis can also be initiated independently of RIPK1 via complexes involving other RHIM domain-containing proteins such as Z-DNA-binding protein 1 (ZBP1, also known as DAI) and TIR domain-containing adapter protein inducing IFNβ (TRIF) [130–132]. These RIPK1-independent necroptotic pathways may be cell-type restricted. They have been observed in vitro in endothelial cells and fibroblasts. However, in human immortalised keratinocytes (HaCaT cells) and in macrophages, TLR3-induced necroptosis requires the formation of the RIPK1-RIPK3 necrosome [128,133].
Necroptosis in mouse models of skin inflammation
As disruption of TNFR1 signalling can favour initiation of not only apoptosis but also necroptosis, there are unsurprisingly several genetic mouse models in which TNFR1-mediated necroptosis has been identified as a driving force behind the inflammatory phenotypes.
Epidermal knockouts (EKO) of the IKK subunits, Nemo or Ikk2, develop severe, widespread dermatological phenotypes shortly after birth. Nemo EKOs die between postpartum day (P)7 and P10, while Ikk2 EKOs are lethal by P9 [134–136]. Mice with combined epidermal loss of the NF-κB subunits, RelA and c-Rel, also developed inflammatory skin lesions similar to the Ikk2 EKOs. Tnfr1 KO rescues these inflammatory phenotypes with mice surviving into adulthood [136,137]. Epidermal-specific KO of TNFR1 was sufficient to rescue the IKK2 EKOs showing again that it is TNFR1 signalling in keratinocytes driving disease [134,138]. Interestingly in the Ikk2 EKOs, blocking apoptosis by crossing to FaddEKO worsened dermatitis while crossing to Ripk3−/− or Mlkl−/− to block necroptosis reduced the severity of the early disease. The necroptotic KOs went on to develop progressive skin lesions from 2 months of age. Blocking both apoptosis and necroptosis by crossing the Ikk2EKO to FaddEKO and Ripk3−/− completely prevented the phenotype. These experiments elegantly demonstrated that keratinocyte death by both apoptosis and necroptosis is responsible for the skin inflammation in these mice [134]. Similarly, in the RelaEKOcRelEKO crossing to Mlkl−/− mice prevented the early onset disease with mice developing inflammatory skin lesions in adulthood instead [134]. The greater disease severity when necroptosis is functional reflects the catastrophic inflammatory potential of this lytic form of cell death. The milder but progressive skin disease when necroptosis is blocked but apoptosis remains functional highlights how low-level but constant apoptotic death in the epidermis can drive chronic inflammation and disease.
Epidermal-specific disruption of RIPK1 binding functions by complete ablation or by RHIM domain mutation also promotes necroptosis-mediated skin inflammation via spontaneous ZBP1 sensing of nucleic acids [22,131,132,139]. While Ripk1EKO mice develop skin lesions from 1 week after birth, crossing of these mice to mice with two RHIM domain-specific mutations in Zbp1 (Zbp1Za1a2/Za1a2) or to MlklEKO mice delayed onset of skin pathology to after 12 weeks of age [22]. This demonstrates a role for RIPK1 scaffold functions in the inhibition of necroptosis even when RIPK1 is not an active participant in the necrosome, most likely by sequestering RIPK3 in an inactive state. While the authors did not specifically show that the late-onset skin phenotype was driven by apoptotic death, they did show that primary keratinocytes derived from Ripk1EKO and from Ripk1EKO Zbp1Za1a2/Za1a2 were sensitised to apoptotic cell death upon treatment with TNF plus cycloheximide [22]. It seems likely that, as in the above mouse models, the late-onset skin phenotype may once again be driven by low-level spontaneous apoptosis precipitated by the loss of RIPK1.
It would be interesting to determine if the necroptotic phenotype of the Ikk2EKO, RelaEKOcRelEKO, and NemoEKO is also driven by nucleic acid-sensing. The question then becomes, what is the triggering event? ZBP1 expression, which is normally restricted to what are likely myeloid cells scattered throughout the dermis, is increased in Ripk1EKO epidermis [22,131,132]. However, the cornification programme actively prevents extracellular nucleic acids through the complete degradation of DNA by epidermal-specific DNases [30,35]. One hypothesis could be that aberrant epidermal apoptosis is still the initiating factor. As discussed earlier, the active suppression of apoptosis during cornification supports the idea that apoptosis has greater inflammatory potential in the epidermis, perhaps because it is a barrier tissue making it more prone to the retention of apoptotic cells. This allows them to progress to late apoptosis and the release of inflammatory cell contents, including nucleic acids. This could then trigger an inflammatory amplification loop where sensing of nucleic acids can drive the expression of type I and type II interferon-mediated genes, including Zbp1. Increased ZBP1 expression and the presence of those same nucleic acids may initiate necroptotic cell death, which drives an even stronger response amplifying the inflammatory effects and producing more extracellular DNA. Unfortunately, because blocking TNFR1-mediated apoptosis will drive RIPK1-mediated necroptosis, simply blocking apoptosis in these models does not prevent disease. It would be interesting, though, to determine if there is a difference in the ratio of various necrosomes depending on whether or not apoptosis is blocked.
Necroptosis can also limit tissue inflammation
Enhanced inflammation in Ripk3−/− and Mlkl−/− at early timepoints in the IAP depletion model shows how necroptotic cell death in the skin may limit tissue inflammation [100]. Similarly, Mlkl KOs have increased inflammatory markers, including IL-6, TNF, and IL-1β, upon Staphylococcus aureus infection [124]. In this case, necroptosis contributes to improved infection outcomes not by participating in bacterial death but by limiting the damage caused by excessive inflammation. Interestingly, in the same model, Ripk3−/− had increased bacterial clearance and reduced inflammation. This can be attributed to the involvement of RIPK3 in multiple cell death and inflammatory responses, independent of MLKL. The absence of RIPK3 in this model led to decreased production of IL-1β and activation of apoptosis, which protected the mice from S. aureus-induced inflammatory damage [124].
Pyroptosis
Pyroptosis is thought to have evolved to prevent pathogen replication in cells. Cell death is characterised by pore formation and the release of pro-inflammatory cytokines. Cellular swelling is followed by loss of membrane integrity and the release of cellular contents, including any intracellular pathogens [140–142]. Pyroptosis is typically initiated downstream of inflammasomes. These large multimeric complexes are nucleated by self-oligomerising sensor proteins that form a molecular scaffold to activate inflammatory caspases. Activation and assembly of inflammasome complexes is mediated by DAMPs and PAMPs through specific PRRs [140,142].
Inflammasome-independent (non-canonical) pyroptosis occurs when caspase-4 or caspase-5 (caspase-11 in mice) directly binds lipopolysaccharides (LPS), independent of TLR4 (the classic PRR of this ligand) [143]. This binding triggers self-oligomerisation and activation of the caspases without the need for an upstream molecular scaffold [144–146] (Figure 5). Non-canonical pyroptosis occurs upon intracellular invasion by Gram-negative bacteria representing a clear infective trigger that will almost always result in pyroptotic death of the cell.
Figure 5. Inflammasome formation and pyroptosis.
Pattern recognition receptor (PRRs) signalling via MYD88 can induce activation of MAPKs and NF-κB, which up-regulate the transcription of genes including those encoding pro-IL-1β and NLRP3. A variety of stimuli trigger the formation of the various inflammasome complexes. Inflammasome assembly leads to the autoproteolytic cleavage and activation of caspase-1, which then cleaves a variety of substrates including pro-IL-1β and pro-IL-18, and the pyroptotic effector GSDMD. Cleaved GSDMD binds to lipids in the plasma membrane and forms oligomeric pores, enabling the release of IL-1β and IL-18. Pore formation also results in cellular swelling due to osmotic pressure and to the death of the cell. Plasma membrane rupture will typically follow pore formation however this is not a passive process due to osmotic swelling as long thought but is instead mediated by Ninj1 following osmotic swelling. Membrane rupture results in the release of cell contents too large to fit through the GSDMD pore. Inflammasome-independent pyroptosis occurs when caspase-4 or caspase-5 (caspase-11 in mice) directly binds LPS and triggers self-oligomerisation, activation of the caspases and cleavage of GSDMD. Unlike caspase-1 these caspases do not directly cleave pro-IL1β and pro-IL18.
Regulation of pattern recognition receptors is an essential component in the skin
Pattern recognition is vital to surveillance and response in the skin. Signalling responses to PRR activation will often involve complex interactions that are in many ways poorly understood. A single PRR can recognise multiple stimuli, and simultaneous signalling within the same cell may be complimentary, amplifying, or inhibitory to the PRR signal. Thus, the downstream response upon activation of these pathways can be highly context dependent. PRRs can activate inflammatory transcription via NF-κB and mitogen-activated protein kinase (MAPK) but also promote inflammasome assembly and the formation of intracellular death complexes [147,148]. Additionally, host monitoring of the microbiota occurs via the TLRs [149–151]. Thus, PRRs operate at the nexus of tolerance, inflammation, and cell death. Proper regulation of these pathways is essential in the skin to ensure a prompt and adequate response upon infection or injury while maintaining tolerance to commensal microorganisms.
PRRs trigger inflammasome formation
There are several types of inflammasomes defined by their core sensor molecule. The most common are of the Nod-like receptor (NLR) family, though inflammasome complexes can also initiate and form around non-NLR proteins, such as AIM2 or Pyrin [147,152]. The NLRP3 inflammasome is the most well studied. The expression of NLRP3 is highly enriched in macrophages and can be detected at lower levels in other immune cells, including epidermal resident LCs [153–156].
Pyroptosis from this complex occurs via a two-step process. First is a priming step such as engagement of a TLR by a bacterial ligand. This results in the activation of transcription factors, including NF-κB, leading to up-regulation of pro-IL-1β and NLRP3. A second stimulus is later detected by NLRP3, prompting assembly of the inflammasome complex. Caspase-1 is recruited to the complex via the adapter protein ASC (Figure 3) [147,148]. Other inflammasome nucleating molecules, such as NLRP1, AIM2, and NLRC4 (also known as IPAF), do not need a priming step for inflammasome formation. However, priming is typically still required for pro-IL-1β expression prior to its caspase 1-mediated cleavage (Figure 3) [147,157].
Inflammasome formation can initiate pyroptosis
The NLRs are structurally related to APAF1, the core component of the apoptosome. Similar to APAF1, NLRs promote caspase oligomerisation and activation. Activated caspases cleave various substrates, including nucleases that facilitate DNA fragmentation, one of the hallmark features of pyroptosis. However, unlike in apoptosis, pyroptotic caspases do not limit inflammation [140]. The pyroptotic caspase, caspase-1, cleaves the critical pyroptotic effector molecule, GSDMD [158,159]. Once cleaved, the N-terminal portion of GSDMD binds to lipids in the plasma membrane and forms oligomeric pores. This leads to increased osmotic pressure, cellular swelling, and the death of the cell [158–161] (Figure 3).
IL-1β and IL-18 secretion
In addition to GSDMD, active caspase-1 also cleaves pro-IL-1β and pro-IL-18, which are then released through pyroptotic pores to the extracellular space [162,163]. There has been some debate as to whether the release of these molecules is directly tied to pyroptotic cell death. Caspase-1 activation is necessary and sufficient for the maturation of IL-1β and is thus a prerequisite for secretion [163]. IL-1β and IL-18 also lack signal peptides for secretion, and thus, pore formation has long been thought to represent their means of release [164]. However, some cells, such as neutrophils, can activate inflammasomes and release these cytokines but are resistant to pyroptotic cell death [165]. One explanation for this is that GSDMD activation and pore formation can induce membrane repair mechanisms in various cell lines. Thus it may be that pyroptosis-resistant cells use the process of inflammasome nucleation and pore formation to secrete IL-1β and IL-18 to drive tissue inflammation while implementing membrane repair mechanisms to prevent death [166].
Casting further doubt on the necessity of pyroptosis for the release of IL-1β, cleaved IL-1β can be secreted in the absence of GSDMD-mediated pore formation, indicating alternative release mechanisms [167–169]. Caspase-1 activation does not automatically trigger pore formation in all cell types. Epithelial cells, including keratinocytes, are resistant to caspase-1-mediated pore formation [140,167,170]. While the efficient, early release of IL-1 β from inflammasome-activated macrophages does appear to be tied to GSDMD pores and pyroptotic cell death [163], slow-release of cleaved IL-1β from macrophages can proceed independently of caspase-1/GSDMD once the mature protein has translocated to the plasma membrane [171].
The resistance of epithelial cells to pore formation and pyroptosis may be protective. Dysregulation of PRRs in barrier tissues could lead to loss of barrier integrity was pyroptosis improperly activated. Additionally, unstimulated human keratinocytes in culture were found to express pro-IL-1β, indicating that keratinocytes do not require priming for its production [172]. PRR signalling in barrier tissues is also responsible for regulating tolerance of the microbiota [173]. As this involves complex interactions between multiple PRR pathways, it makes sense for barrier cells to resist pyroptotic cell death and the associated release of DAMPs, even upon PRR activation and inflammasome formation.
GSDMD-mediated pyroptosis occurs prior to and independent of plasma membrane rupture
Interestingly, Ninjurin-1 (NINJ1)-mediated rupture of cell membranes follows GSDMD-mediated cell death but has been genetically separated from the upstream events. NINJ1 deficient macrophages still die following inflammasome activation and GSDMD pore formation. The cells exhibit normal GSDMD-mediated IL-1β and IL-18 secretion and morphological changes, including cell swelling. However, the cells fail to rupture, preventing the release of many DAMPs that cannot pass through the GSDMD pore. This includes the prototypical pro-inflammatory DAMP, HMGB1 [98]. These findings are significant in several ways. They show that plasma membrane rupture during pyroptosis is not a passive osmotic process as long presumed. They also uncouple GSDMD-mediated cell death from cell rupture and the wholesale release of DAMPs during pyroptosis.
The NLRP1 inflammasome modulates UV-induced inflammation but not pyroptosis in human skin
The most highly expressed NLR in human skin is NLRP1, where it appears to be the key inflammasome sensor in the epidermis [174–176]. It is broadly expressed but can be found at particularly high levels in differentiated epithelial cells such as keratinocytes [177]. The NLRP1 inflammasome is triggered by the bacterial ligand muramyl dipeptide (MDP), by the anthrax lethal toxin, and by UV irradiation via intracellular ATP depletion [177]. What is interesting about the latter is that while keratinocytes remain resistant to pyroptosis upon caspase-1 activation, NLRP1 inflammasome formation in human keratinocytes upon UV irradiation can prompt caspase-1 mediated apoptotic cell death instead. However, the inflammasome itself is dispensable for caspase-1-mediated apoptosis to occur [175,178,179]. This supports the idea that barrier tissue resistance to pyroptosis may function to prevent excessive DAMP release into an already highly immunostimulatory environment while still allowing for the appropriate (and largely DAMP-free) death of potentially malignant, replicating keratinocytes upon UV irradiation.
NETosis
NETosis refers to cell death occurring upon extrusion of neutrophil extracellular traps (NETs), a mesh of decondensed chromatin and histones covered in granular and cytoplasmic proteins [26,180,181]. Despite the name, neutrophils are not the only NET-producing cells. Similar extracellular meshes have been associated with eosinophils, basophils and mast cells [182–185]. The primary function of NETosis appears to be infection control. Antimicrobial agents decorate the NETs, and the mesh will physically confine (trap) pathogens at the site of infection [186–188]. NETs are commonly found in healing wounds where their beneficial effects are linked to their antimicrobial activity [189]. However, NETosis is a highly inflammatory process. The NETs themselves contain autoantigens, and NET extrusion involves cell membrane perforation. NETotic cells with ruptured cell membranes release DAMPs and other inflammatory mediators, thereby exacerbating the inflammatory response and causing tissue pathology [190]. Up-regulation of NETosis in diabetic wounds enhances inflammation and delays healing [191].
Molecular events associated with the NET formation
The precise mechanisms involved in NET formation have not been fully established. An NADPH oxidative burst and peptidyl arginine deiminase 4 (PAD4)-mediated histone citrullination and chromatin decondensation appear to be necessary steps [192]. The migration of neutrophil elastase (NE) to the nucleus is thought to promote histone processing and chromatin decondensation [193,194]. NE and an alternative neutrophil serine protease (NSP), cathepsin G, can also induce GSDMD cleavage in neutrophils [195,196], which is necessary for NET extrusion [197,198]. Thus, NE has long been considered a key mediator of NETosis. However, the use of specific NSP inhibitors failed to prevent DNA extrusion in neutrophils suggesting that the catalytic activity of NSPs, including NE, is dispensable for NET formation [199].
Pyroptosis or necroptosis with NETs: is NETosis truly a separate form of PCD?
There is ongoing debate as to whether NETosis constitutes a separate form of cell death [200,201]. Cellular lysis does not occur in all cases of NET extrusion and can involve the selective extrusion of mitochondrial (rather than nuclear) DNA without lytic cell death [185,202]. There is significant evidence of mechanistic overlap between NETosis and pyroptosis. Both processes play a role in infection control and share many similar triggers [140,203,204]. Caspase-11 and GSDMD are required for plasma membrane perforation to enable NET release [197,198], and pyroptosis-related IL-1β has been associated with the triggering of NETosis [205].
NETs have also been observed in neutrophils undergoing necroptosis in both human and mouse cells [206,207]. Treatment of human neutrophils with an MLKL inhibitor (necrosulfonamide (NSA)) reduced NET formation in vitro [206]. NET formation and associated activation of RIPK3/MLKL has been observed in neutrophil-rich tissue samples from patients with cutaneous vasculitis and psoriasis [207]. However, necroptotic signalling does not appear to be necessary for NET formation [208].
The key distinction between pyroptosis and NETosis is whether DNA is retained (pyroptosis) or expelled (NETosis) during lysis [199]. However, given the mechanistic overlap, another way of thinking about this could be that NET formation is a parallel process that can (but does not always) occur during the execution of lytic cell death programmes. Whether or not NET formation occurs will depend on the stimuli. Extrusion of the NETs could then be described as a by-product of MLKL or GSDMD pore formation.
Ferroptosis
Ferroptosis is a biochemically distinct form of regulated cell death. It is triggered when dysregulation of intracellular iron homeostasis leads to an accumulation of iron-dependent reactive oxygen species (ROS). Excess cellular ROS induces lipid peroxidation causing lethal damage to lipids, proteins and nucleic acids, and the caspase and necrosome independent death of the cell [209–211]. The anti-oxidative enzyme Glutathione peroxidase 4 (GPX4) reduces and prevents lipid peroxidation making it a key regulator of ferroptosis. Inhibition or ablation of GPX can be used to induce ferroptosis in vitro and in vivo [212–214].
Morphological features of ferroptosis include reduced cell volume, shrunken mitochondria, reduction in mitochondria crista, and mitochondrial outer membrane rupture [215], though notably without the release of cytochrome-c or activation of caspases [209]. Ferroptosis can be inhibited by iron chelators, lipophilic antioxidants, and lipid peroxidation inhibitors [216,217].
Ferroptosis-associated inflammation
Ferroptotic cells are potent mediators of tissue inflammation. The release of DAMPs, including HMGB1, cell-free (cf)DNA and IL33, makes ferroptotic cell death inflammatory. However, the mechanisms of DAMP release are not entirely clear. Cytoplasmic and organelle swelling, and plasma membrane rupture as seen in necroptosis or pyroptosis are not universally observed during ferroptosis [209,218,219]. One reason for this may be the tug of war between lipid peroxidation-induced membrane damage and membrane repair mechanisms. Both processes can be initiated by the same triggers such as ER stress-mediated calcium influx [219,220]. Some degree of membrane damage even without wholesale membrane rupture may allow for the passive release of DAMPs. However, there is evidence to suggest that the active release of certain DAMPs may also occur. One study has shown that, the prototypical DAMP, HMGB1, is actively released during ferroptosis upon its autophagy-dependent acetylation [221].
Additionally, ferroptotic cells can work as signal transmitters inducing a chain of further ferroptosis in surrounding cells in a paracrine effect. Lipid peroxidation and its aftereffects can propagate from ferroptotic cells to surrounding cells that were not exposed to ferroptosis inducers [222].
The potential role of ferroptosis in the skin
In recent years excessive or defective ferroptosis has been associated with a plethora of disease states, most notably cancer, neurodegeneration, and ischaemic organ injuries [6,223]. Ferroptosis may also contribute to pathogenesis in autoimmune diseases, including SLE [224,225]. Mice with neutrophil-specific Gpx4 haploinsufficiency (Gpx4fl/wt LysMCre+) develop SLE-like symptoms including autoantibodies, neutropenia, skin lesions, and proteinuria [225]. Until very recently the role of ferroptosis in skin pathophysiology has remained largely unexplored. That said, there are emerging indications of its potential relevance.
Cultured primary keratinocytes treated with the ferroptotic inducer erastin had reduced viability and increased expression of a suite of psoriasis-associated cytokines (TNF-α, IL-6, IL-1α, IL-1β, IL-17, IL-22, and IL-23). These effects were reversed when cells were co-treated with Fer-1, a specific inhibitor of ferroptosis. Fer-1 treatment also reduced the severity of imiquimod-induced psoriasis in mice [226].
Ferroptosis appears to contribute significantly to acute inflammation in sunburn. A portion of the UVB radiation-induced death of primary human epithelial keratinocytes (HEKs) in culture could be prevented by treatment with various ferroptotic inhibitors, particularly in the first 6 h following radiation [227]. The death of these cells in the absence of the inhibitors was accompanied by ferropototic death signals, including the accumulation of oxygenated phospholipids, and the release of HMGB1 into the media. HMGB1 release was mitigated by treatment with Fer-1, but not by the apoptotic inhibitor Z-Vad. Mice topically pre-treated with Fer-1 prior to UVB irradiation had reduced immune cell infiltrate and inflammatory cytokines in the skin. This effect was not seen in mice pre-treated with Z-Vad, supporting the idea that a significant portion of UVB-induced inflammation is due to the ferroptotic death of keratinocytes [227].
While ferroptosis is an inflammatory form of cell death, it may in some circumstances have a protective role in the skin. Down-regulation of GPX4, which is considered a hallmark of ferroptosis, appears to contribute to the resolution of skin inflammation and of non-melanoma skin cancer [228]. Additionally, while excessive or prolonged HMGB1-driven inflammation is associated with chronic inflammatory disease [14,229,230], HMGB1 can also stimulate tissue repair processes, including keratinocyte migration and angiogenesis [231–235]. That HMGB1 appears to be actively released during ferroptosis suggests pathophysiological functions for the DAMP in that context. Acute ferroptotic inflammation associated with UVB radiation, for example, may be an important mechanism for promoting epidermal healing in sunburn. Thus, whether its association with any particular skin disease is causative, reactionary, or protective will need to be carefully examined.
Cell death in human skin disease
Histopathological findings of cell death in patient skin samples are typically described as apoptotic or necrotic, but the precise nature of said necrosis usually remains undefined. This simple dichotomy has been the case for decades. In the meantime, the cell death field has expanded substantially. Late apoptosis, necroptosis, pyroptosis, and NETosis and ferroptosis have all been associated with human skin conditions and could all potentially be described as necrotic by the histopathologist.
Apoptosis and necroptosis in human skin disease
Epidermal apoptosis features in inflammatory skin conditions in humans, including Lichen planus (LP) [236], atopic dermatitis (AD) [237], graft versus host disease (GvHD) [238], bullous pemphigoid [239], and chronic diabetic skin ulcers [240]. Apoptotic keratinocytes that release DAMPs have been posited as a potentially initiating event in the pyoderma gangrenosum [241]. Activation of the RIPK3–MLKL axis has been observed in some neutrophilic diseases [207] including psoriasis [242]. Increased epidermal expression of RIPK3 has also been seen in the lesional epidermis in lichenoid tissue reactions [243], and in SJS/TEN [244]. Loss of cFLIP in keratinocytes may play a role in the increased caspase-8 mediated cell death seen in TEN/SJS patients [101,245].
Inflammasomes and pyroptosis in skin-associated disease
Variations in NLRP1 are associated with susceptibility to inflammation and autoimmunity contributing to vitiligo, psoriasis and AD [177,246–248]. Inflammasome activation and IL-1β and IL-18 release are also seen in pyogenic arthritis, pyoderma gangrenosum, and acne (PAPA) syndrome and in hidradenitis suppurativa. In PAPA, mutations in proline–serine–threonine phosphatase-interacting protein 1 (PSTPIP1) lead to hyperphosphorylation of PSTPIP1, increasing its interaction with the autoinhibitory domain of the pyrin inflammasome, promoting inflammasome activation and IL-1β production [249]. Activated caspase-1 and increased expression of NLRP3 and IL-18 were seen in skin biopsies from hidradenitis suppurativa patients [250,251], suggesting the potential involvement of pyroptotic cell death in this disease.
Ferroptosis in inflammatory skin disease
To date, there have been very few studies directly linking ferroptosis with inflammatory skin diseases in humans. However, there is reason to think it may play a role. GPX4 expression was reduced and markers of lipid oxidation were significantly up-regulated in psoriasis patient samples. Lipid oxidation activity in keratinocytes was highly correlated to Th22/Th17 activity, a hallmark of psoriatic inflammation [226]. Elevation of IL-4 and IL-13 are hallmarks of Th2 inflammatory diseases including AD [252]. Increased expression of these molecules suppresses expression of GPX4 [253], which would in turn sensitise cells to ferroptosis. Keratinocyte-derived ROS, increased oxidative stress, and decreased enzymatic and non-enzymatic antioxidants appear to contribute to barrier defect and pathogenesis in AD [254,255]. ROS are also notably elevated in LP [256] and in OLP [257,258] where lipoperoxidation and carbonyl stress are thought to contribute to the progression to carcinogenesis. Thus far these effects have not been identified explicitly as ferroptosis. They do however suggest that further exploration is warranted. The precise role that ferroptosis may play in inflammatory skin disease remains to be seen,
Multiple inflammatory and cell death programmes contribute to skin disease in complex trait disorders
Disrupted innate immune signalling is a common feature in inflammatory skin disease. As a barrier tissue, the skin is exposed continuously to commensals and pathogens, making it vulnerable to inflammatory consequences. Loss of barrier function in epithelial surfaces can result in prolonged exposure to PAMPs, and a more intense inflammatory milieu. Dysbiosis is a common feature of skin diseases, including psoriasis and AD [149,259–261] and is often thought to contribute to the pathology. The combination of genetic susceptibilities, external triggers, and environmental factors highlights the nature of inflammatory skin diseases as complex trait disorders, where multiple processes may coexist and interact to drive disease.
Psoriasis
Psoriasis is a chronic, immune-mediated skin disease. The condition is characterised by clearly defined, dry, red plaques with silvery-white scales, epidermal hyperplasia, and marked parakeratosis [262]. Psoriasis arises due to long-term interactions between keratinocytes and infiltrating, activated immune cells. Defects in the cornification programme, reduced apoptosis, increased PRR signalling and possibly aberrant necroptosis, pyroptosis, ferroptosis and NETosis may all play some role in psoriasis pathogenesis and inflammatory exacerbation.
Twin and family studies have shown an important, though undeniably complex, genetic component to psoriasis. What is striking is that genetic susceptibilities for psoriasis frequently involve genes that encode innate immune components including IL-23 signalling (IL23R, IL12B, TRAF3IP2, IL23A, TYK2), IFN signalling (IL28RA, IFIH1, TYK2, RNF114, SOCS1), NF-κB signalling (REL, TNIP1, TRAF3IP2, TNFAIP3, NFKBIA, FBXL19, CARD14, CARM1), IL-4/IL-13 signalling (IL4, IL13), and bacterial or viral responsiveness (NOS2, IL28RA, DDX58, ELMO1) [263–266]. The effects of these polymorphisms on interacting immune signalling and cell death pathways is no doubt immensely complex and still largely unexplored.
Infiltration by dendritic cells (DCs) expressing TNFα and iNOS is heavily featured in psoriatic lesions (Harden et al. [265]), and the efficacy of anti-TNF in treating psoriasis highlights its importance in the disease. Psoriatic lesions are notably deficient in apoptosis [267]. However, both NET formation and activation of the RIPK3–MLKL axis have been found in neutrophil-rich psoriatic tissue samples [207,268], and mast cell NETosis (METosis) has been identified as an important source of IL-17 in the disease [183]. Increased circulating and lesional HMGB1 is associated with psoriasis pathogenesis, [269] supporting the potential role of these lytic cell death programmes in exacerbating inflammation in psoriasis.
Defective cornification is a feature of psoriasis. Deleting two members of the late cornified envelope (LCE) gene cluster (LCE3B and LCE3C) confers significant risk for the development of psoriasis [270]. While initially, these proteins were assumed to contribute to skin barrier function, further investigation showed this was not the case and instead identified them as having a defensin-like antimicrobial activity [271]. IL-37 expression is decreased in psoriasis [272,273], and mutations of IL36RN can cause generalised pustular psoriasis [274]. Both of these are among the anti-inflammatory proteins that are up-regulated during cornification [41]. NLRP1, NLRP3, AIM2, caspase-1, IL-1, and IL-18 have also all been identified as elevated in psoriatic skin samples compared with healthy controls.
The increased expression of the DNA sensing protein AIM2 in psoriatic keratinocytes is interesting when considered alongside the down-regulation of DNase1L2 observed in parakeratotic psoriasis lesions [35]. While combined loss of DNase1L2 and DNase2 proteins in mice does lead to DNA retention (parakeratosis) in the stratum corneum, it was insufficient to activate inflammatory pathways [30]. However, in healthy skin, AIM2 expression is restricted to LCs and melanocytes [153]. The combination of parakeratosis and up-regulation of AIM2 in psoriatic keratinocytes may contribute to pathogenic inflammation in disease.
Polymorphisms in NLRP3 and CARD8 are associated with increased risk of psoriasis development, while NLRP1 mutations have been associated with diverse cutaneous inflammatory diseases, including psoriasis, highlighting the critical role of NLRP1 in epidermal biology [246]. All of which point towards a role for epidermal host defence, abnormal and chronic activation of inflammasome complexes, and potentially pyroptotic cell death, in the psoriasis pathology.
Eczematous dermatitis
Eczematous disease encompasses several similar disorders, including allergic contact dermatitis (ACD), irritant contact dermatitis (ICD), and AD, which is what is commonly and colloquially known as eczema. These disorders present similar symptoms; a highly pruritic (itchy), often painful, red rash. They are acutely eczematous but can progress to a chronic stage which is dry with thickened skin. [275]. Keratinocyte apoptosis caused by skin-infiltrating T-cells appears to be a key event in the pathogenesis of AD and ACD. Keratinocyte apoptosis was observed in lesional skin affected by AD, ACD, and in patch tests. IFNg-mediated up-regulation of FasR sensitises keratinocytes to T-cell-mediated apoptosis [237].
Defects in cornification cause structural abnormalities of the cornified layer that affect epidermal barrier functions. Loss-of-function mutations in the Filaggrin (FLG) gene are the most significant genetic risk factor for AD development, though they are neither sufficient nor necessary for disease [276–278]. Polymorphisms in the cornification-associated serine protease inhibitor SPINK5 have also been associated with dermatitis [279–281].
AIM2 expression can also be detected in keratinocytes in AD and ACD [282]. Genome-wide association studies identified additional disease risk loci, many of which are involved in immune regulation, including IL4, IL4RA, IL13, RANTES, IL18RAP, TNFSF6B, IL2RA, IL7R, STAT3, NOD1, NOD2, TLR2, and CD14 [283,284]. Immune dysregulation may contribute to an overreactive response to allergens and to the pathogenic skin microbe S. aureus [260,261,285].
Lichen planus
LP is a common, chronic, inflammatory disease that can affect mucosal and cutaneous sites, including the skin (cutaneous lichen planus, CLP) and oral cavity (oral lichen planus, OLP). The disease is mediated by activated CD8+ lymphocyte-induced keratinocyte apoptosis, and the consequent release of pro-inflammatory mediators and chronic lymphocytic infiltrate [236,286]. Increased Fas/FasL expression in basal keratinocytes results in increased epidermal apoptosis [287], while little to no apoptosis in the subepithelial cell infiltrate causes persistence of the inflammatory infiltrate and chronicity of the disease [288].
NF-κB expression is increased in keratinocytes from CLP and OLP compared with healthy tissue; however, more severe manifestations of the disease with erosive lesions have reduced epithelial expression of NF-κB and increased keratinocyte apoptosis, demonstrating how a shift from inflammation to apoptotic cell death can exacerbate disease [236,289]. On the other hand, one theory regarding malignant transformation in OLP is that epithelial cells may develop mechanisms to evade apoptosis and increase their proliferation rate [288]. While this may be an attempt to preserve the epithelial structure in the face of the Fas onslaught, it also, unfortunately, instigates two of the hallmarks of cancer [290].
Expression of the caspase-1 inhibitor, CARD18, which is up-regulated during keratinocyte differentiation and cornification, was strongly reduced in involved skin of CLP patients. CARD18 inhibits caspase-1, preventing pyroptosis and indirectly suppressing the secretion of IL-1β [41,291]. The inflammasome nucleating proteins AIM2 and NLRP1 were also up-regulated in CLP in the epidermis and dermis, respectively [292], again suggesting a potential role for inflammasomes and pyroptosis in enhancing inflammation in LP.
Therapeutic opportunities for targeting cell death pathways
Immunomodulatory drugs are the cornerstone of treatment for ISDs. In mild to moderate disease, this means topical corticosteroids. If the disease is severe or widespread topical treatments are less practical or effective, indicating the use of systemic immunosuppressant medications such as cyclosporin or methotrexate [293,294]. In the last decade, biologic drugs that target mediators of inflammatory and immune responses have had considerable success in treating a range of inflammatory disorders. However, their action is not universal. The nature of ISDs as complex trait disorders means that the success of any particular treatment can vary widely from person to person. Some patients have no initial response, while others may lose the response over time. A subset of patients also experience a paradoxical induction or exacerbation of cutaneous disease in response to biologic treatment [295–301].
There is the additional consideration that both steroids and non-steroidal immunosuppressant drugs have factors that limit their long-term use [293,302]. These treatments are not a cure, and conditions such as AD and psoriasis can be lifelong. Maintaining patients on systemic immune-suppressing medications such as steroids, anti-TNF, or T-cell modulation therapies means maintaining a lifelong state of immunosuppression.
Given the role that DAMPs can play in enhancing inflammatory feedback loops, there is interest in targeting them to short circuit chronic inflammation. However, there are many potential DAMPs and much uncertainty as to their relative, context dependent, inflammatory potentials. Direct targeting of DAMPs to prevent inflammation, is therefore a difficult proposition. If cell death is a source of DAMPs contributing to chronic inflammation, then inhibiting cell death could be an effective anti-inflammatory approach. Targeting cell death signalling pathways provides an additional therapeutic opportunity for treating inflammatory disease. Small molecule inhibitors targeting critical regulators and inducers of cell death (caspases and RIP kinases) as well as the membrane-disrupting proteins (MLKL, GSDMD and NINJ1) are of considerable interest.
Targeting RIPKs
A set of small-molecule inhibitors of necroptosis, referred to as necrostatins, were initially identified using unbiased cell-based screening. They were later shown to function by inhibiting RIPK1 kinase functions [303]. As they did not affect the scaffold functions of RIPK1, which, as previously discussed, are required for the formation of molecular platforms during pro-survival signalling, these necrostatins and other RIPK1 targeting drugs could block RIPK1-dependent cell death (both apoptotic and necroptotic) without impairing survival signalling to NF-κB and MAPK. However, early iterations of these drugs had off-target effects, including it seems inhibition of ferroptosis in a RIPK1-independent manner. More selective RIPK1 inhibitors entered human clinical trials for the treatment of IBD, Crohn's disease, RA, ALS, and Alzheimer's disease [303,304]. A randomised control trial of the RIPK1 inhibitor GSK2982772 found use of the drug to be safe and saw some improvements in clinical disease, though the interpretation of results was hampered by an unusually high placebo effect in one of the dosing cohorts. The researchers have suggested that further trials using a higher drug dose and in patients with more active disease are warranted [305,306].
RIPK3 is involved in necroptotic cell death and, in some circumstances, in pyroptosis, apoptosis, and cytokine production. As RIPK3 knockout mice are viable and fertile, RIPK3 inhibition was assumed to be safe and was thus considered a promising treatment target for chronic inflammatory diseases. However, surprisingly, specific inhibition of the RIPK3 kinase domain can be toxic. Mice with a Ripk3 kinase-dead mutation (Ripk3D161N) are embryonic lethal due to excessive RIPK1-mediated apoptosis [307]. While several small-molecule inhibitors specific for the RIPK3 kinase domain were developed, they were also found to induce RIPK1-mediated apoptotic cell death [308] and have not progressed through therapeutic development [309]. If interest in RIPK3 inhibition were to be reignited, complete inhibition or protein degradation might be required to recapitulate the inflammatory benefits seen in RIPK3 KO mice.
Targeting caspases
Caspases are key inducers of extrinsic apoptosis and pyroptosis, both of which are increasingly implicated in inflammatory disease pathogenesis. While this makes them potentially attractive therapeutic targets, there has not been much success with this strategy so far. Targeting specific caspases is difficult due to highly conserved active sites [310], while pan-caspase inhibition risks inhibiting healthy, physiological apoptosis or negating the anti-inflammatory moderation of intrinsic apoptosis. Caspase 8-specific inhibitors might be useful in conditions with excessive extrinsic apoptosis; however, as complete caspase-8 inhibition can trigger necroptosis [311] inhibition would need to be carefully modulated to avoid exacerbating inflammation. Caspase-1 is a promising target for treating IL-1β-mediated conditions. Unlike caspase-8, knockout of caspase-1 in mice is not lethal, so a selective caspase-1 inhibitor is less likely to cause serious off-target effects.
Targeting membrane permeabilization and cell lysis
MLKL and GSDMD, the membrane-disrupting executioner proteins of necroptosis and pyroptosis, respectively, are also of interest as therapeutic targets [312–314]. As well as their importance in executing those highly inflammatory cell death mechanisms, GSDMD pores are important for the release of IL-1β, and GSDMD and maybe MLKL, are associated with NET extrusion. The MLKL inhibitor NSA blocks MLKL-mediated membrane disruption possibly by affecting oligomerisation, though recent work in epithelial cells showed NSA blocked trafficking of oligomers to the membrane [118]. NSA can also bind to GSDMD, where it appears to affect the formation of the oligomeric pore [312]. NSA, or chemically similar compounds, may be of value in the treatment of IL-1β-mediated conditions, such as PAPA and HS, and in diseases associated with NETosis, including vasculitis, SLE, and psoriasis.
Another potentially attractive target for mitigating inflammation during lytic cell death is NINJ1 [98]. NINJ1 mediates plasma membrane rupture following pyroptotic cell death. It also significantly contributes to membrane rupture in post-apoptotic necrosis and necrosis induced by pore-forming bacterial toxins. A NINJ1 monoclonal antibody prevented plasma membrane rupture in cell culture, indicating that it is effectively targetable [98]. While it would not necessarily be helpful in treating IL-1Β mediated disease as its loss has no impact on GSDMD pore formation and cytokine release, NINJ1 inhibition may have therapeutic benefit in mitigating the wholesale release of DAMPs following pyroptosis or post-apoptotic necrosis.
Concluding remarks
Inflammation caused by lytic forms of cell death such as necroptosis, pyroptosis and NETosis do have important physiological functions, in particular as part of infection control systems. The physiological importance of ferroptotic inflammation is less clear but it may have a role in immune surveillance and tumour suppression systems [6]. However, dysfunctional innate immune signalling can lead to inappropriate activation of these highly inflammatory processes. The immunostimulatory environment of the skin makes it particularly prone to such over-activation. Even apoptotic cell death, which follows a programme specifically meant to limit or prevent inflammation, can instigate and drive inflammation in the skin. That apoptosis is actively suppressed in keratinocytes during terminal differentiation and cornification indicates the inflammatory potential of apoptotic cells in the epithelial barrier. Accumulating evidence in mouse models and some human studies suggests that targeting cell death has therapeutic potential in inflammatory skin disease. A difficulty remains, however, in the accurate identification of specific cell death modalities in skin disease. Development of good histopathological biomarkers that can distinguish the different cell death modalities is needed if we are to develop better, more targeted treatments.
Inflammatory skin diseases are typically complex trait disorders meaning multiple interacting factors, genetic and environmental, will contribute to disease pathogenesis. While this review has outlined several individual, well-described cell death pathways that can play a role in skin inflammation, the reality is that multiple signalling pathways can coexist and interact in the same disease. Effective treatments may require targeting multiple cell death and inflammatory pathways. That said, a better understanding of the molecular mechanisms that regulate cell death and their specific and interacting contributions to inflammation in the skin has the potential to produce better treatments for inflammatory skin diseases.
Abbreviations
- ACD
allergic contact dermatitis
- AD
atopic dermatitis
- cFLIP
cellular FLICE-like inhibitory protein
- CLP
cutaneous lichen planus
- DAMPs
damage-associated molecular patterns
- EKO
epidermal knockouts
- FADD
Fas-associated death domain
- GPX4
glutathione peroxidase 4
- GSDMD
gasdermin D
- HOIL-1
hemeoxidized iron-regulatory protein 2 ubiquitin ligase-1
- IAP
inhibitor of apoptosis protein
- IKK
IκB kinase
- LCs
Langerhans cells
- LP
lichen planus
- LUBAC
linear ubiquitin-chain assembly complex
- MAPK
mitogen-activated protein kinase
- MLKL
mixed lineage kinase domain-like
- MOMP
mitochondrial outer membrane permeabilisation
- NETs
neutrophil extracellular traps
- NF-κB
nuclear factor-kappa B
- NLR
Nod-like receptor
- NSP
neutrophil serine protease
- PAMPs
pathogen-associated molecular patterns
- PAPA
pyoderma gangrenosum, and acne
- PCD
programmed cell death
- PRRs
pattern recognition receptors
- ROS
reactive oxygen species
- SLE
systemic lupus erythema
- TAK1
transforming growth factor-β-activated kinase 1
- TEN
toxic epidermal necrolysis
- TLR
Toll-like receptor
- XIAP
X-linked Inhibitor of apoptosis protein
Competing Interests
The authors declare that there are no competing interests associated with the manuscript.
Open Access
Open access for this article was enabled by the participation of University of Melbourne in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with CAUL.
CRediT Author Contribution
Holly Anderton: Conceptualization, Supervision, Investigation, Writing — original draft, Writing — review and editing. Suhaib Alqudah: Investigation, Writing — original draft, Writing — review and editing.
References
- 1.Eckhart, L., Lippens, S., Tschachler, E. and Declercq, W. (2013) Cell death by cornification. Biochim . Biophys. Acta 1833, 3471–3480 10.1016/j.bbamcr.2013.06.010 [DOI] [PubMed] [Google Scholar]
- 2.Hosakote, Y.M., Brasier, A.R., Casola, A., Garofalo, R.P. and Kurosky, A. (2016) Respiratory syncytial virus infection triggers epithelial HMGB1 release as a damage-associated molecular pattern promoting a monocytic inflammatory response. J. Virol. 90, 9618–9631 10.1128/JVI.01279-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Place, D.E. and Kanneganti, T.D. (2019) Cell death-mediated cytokine release and its therapeutic implications. J. Exp. Med. 216, 1474–1486 10.1084/jem.20181892 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Rodriguez-Rosales, Y.A., Langereis, J.D., Gorris, M.A.J., van den Reek, J.M.P.A., Fasse, E., Netea, M.G.et al. (2021) Immunomodulatory aged neutrophils are augmented in blood and skin of psoriasis patients. J. Allergy Clin. Immunol. 148, 1030–1040 10.1016/j.jaci.2021.02.041 [DOI] [PubMed] [Google Scholar]
- 5.Venereau, E., Ceriotti, C. and Bianchi, M.E. (2015) DAMPs from cell death to new life. Front. Immunol. 6, 422 10.3389/fimmu.2015.00422 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jiang, X., Stockwell, B.R. and Conrad, M. (2021) Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 10.1038/s41580-020-00324-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Garg, A.D., Galluzzi, L., Apetoh, L., Baert, T., Birge, R.B., Bravo-San Pedro, J.M.et al. (2015) Molecular and translational classifications of DAMPs in immunogenic cell death. Front. Immunol. 6, 588 10.3389/fimmu.2015.00588 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Murao, A., Aziz, M., Wang, H., Brenner, M. and Wang, P. (2021) Release mechanisms of major DAMPs. Apoptosis 26, 152–162 10.1007/s10495-021-01663-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Nakano, H., Murai, S. and Moriwaki, K. (2022) Regulation of the release of damage-associated molecular patterns from necroptotic cells. Biochem. J. 479, 677–685 10.1042/BCJ20210604 [DOI] [PubMed] [Google Scholar]
- 10.Bell, C.W., Jiang, W., Reich, C.F. and Pisetsky, D.S. (2006) The extracellular release of HMGB1 during apoptotic cell death. Am. J. Physiol. Cell Physiol. 291, C1318–C1325 10.1152/ajpcell.00616.2005 [DOI] [PubMed] [Google Scholar]
- 11.Hou, L., Yang, Z., Wang, Z., Zhang, X., Zhao, Y., Yang, H.et al. (2018) NLRP3/ASC-mediated alveolar macrophage pyroptosis enhances HMGB1 secretion in acute lung injury induced by cardiopulmonary bypass. Lab. Investig. 98, 1052–1064 10.1038/s41374-018-0073-0 [DOI] [PubMed] [Google Scholar]
- 12.Lamkanfi, M., Sarkar, A., Vande Walle, L., Vitari, A.C., Amer, A.O., Wewers, M.D.et al. (2010) Inflammasome-dependent release of the alarmin HMGB1 in endotoxemia. J. Immunol. 185, 4385–4392 10.4049/jimmunol.1000803 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Negroni, A., Colantoni, E., Pierdomenico, M., Palone, F., Costanzo, M., Oliva, S.et al. (2017) RIP3 AND pMLKL promote necroptosis-induced inflammation and alter membrane permeability in intestinal epithelial cells. Dig. Liver Dis. 49, 1201–1210 10.1016/j.dld.2017.08.017 [DOI] [PubMed] [Google Scholar]
- 14.Scaffidi, P., Misteli, T. and Bianchi, M.E. (2002) Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 418, 191–195 10.1038/nature00858 [DOI] [PubMed] [Google Scholar]
- 15.Simpson, J., Loh, Z., Ullah, M.A., Lynch, J.P., Werder, R.B., Collinson, N.et al. (2020) Respiratory syncytial virus infection promotes necroptosis and HMGB1 release by airway epithelial cells. Am. J. Respir. Crit. Care Med. 201, 1358–1371 10.1164/rccm.201906-1149OC [DOI] [PubMed] [Google Scholar]
- 16.Urbonaviciute, V., Fürnrohr, B.G., Meister, S., Munoz, L., Heyder, P., De Marchis, F.et al. (2008) Induction of inflammatory and immune responses by HMGB1-nucleosome complexes: implications for the pathogenesis of SLE. J. Exp. Med. 205, 3007–3018 10.1084/jem.20081165 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cevikbas, F. and Steinhoff, M. (2012) IL-33: a novel danger signal system in atopic dermatitis. J. Invest. Dermatol. 132, 1326–1329 10.1038/jid.2012.66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Shlomovitz, I., Erlich, Z., Speir, M., Zargarian, S., Baram, N., Engler, M.et al. (2019) Necroptosis directly induces the release of full-length biologically active IL-33 in vitro and in an inflammatory disease model. FEBS J. 286, 507–522 10.1111/febs.14738 [DOI] [PubMed] [Google Scholar]
- 19.Allam, R., Darisipudi, M.N., Tschopp, J. and Anders, H.J. (2013) Histones trigger sterile inflammation by activating the NLRP3 inflammasome. Eur. J. Immunol. 43, 3336–3342 10.1002/eji.201243224 [DOI] [PubMed] [Google Scholar]
- 20.Tydén, H., Lood, C., Gullstrand, B., Jönsen, A., Ivars, F., Leanderson, T.et al. (2017) Pro-inflammatory S100 proteins are associated with glomerulonephritis and anti-dsDNA antibodies in systemic lupus erythematosus. Lupus 26, 139–149 10.1177/0961203316655208 [DOI] [PubMed] [Google Scholar]
- 21.Basu, S., Binder, R.J., Suto, R., Anderson, K.M. and Srivastava, P.K. (2000) Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway. Int. Immunol. 12, 1539–1546 10.1093/intimm/12.11.1539 [DOI] [PubMed] [Google Scholar]
- 22.Devos, M., Tanghe, G., Gilbert, B., Dierick, E., Verheirstraeten, M., Nemegeer, J.et al. (2020) Sensing of endogenous nucleic acids by ZBP1 induces keratinocyte necroptosis and skin inflammation. J. Exp. Med. 217, e20191913 10.1084/jem.20191913 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Toussaint, M., Jackson, D.J., Swieboda, D., Guedán, A., Tsourouktsoglou, T.D., Ching, Y.M.et al. (2017) Host DNA released by NETosis promotes rhinovirus-induced type-2 allergic asthma exacerbation. Nat. Med. 23, 681–691 10.1038/nm.4332 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tsourouktsoglou, T.D., Warnatsch, A., Ioannou, M., Hoving, D., Wang, Q. and Papayannopoulos, V. (2020) Histones, DNA, and citrullination promote neutrophil extracellular trap inflammation by regulating the localization and activation of TLR4. Cell Rep. 31, 107602 10.1016/j.celrep.2020.107602 [DOI] [PubMed] [Google Scholar]
- 25.Tanzer, M.C., Frauenstein, A., Stafford, C.A., Phulphagar, K., Mann, M. and Meissner, F. (2020) Quantitative and dynamic catalogs of proteins released during apoptotic and necroptotic cell death. Cell Rep. 30, 1260–1270.e5 10.1016/j.celrep.2019.12.079 [DOI] [PubMed] [Google Scholar]
- 26.Galluzzi, L., Vitale, I., Aaronson, S.A., Abrams, J.M., Adam, D., Agostinis, P.et al. (2018) Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 25, 486–541 10.1038/s41418-017-0012-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Arda, O., Goksugur, N. and Tuzun, Y. (2014) Basic histological structure and functions of facial skin. Clin. Dermatol. 32, 3–13 10.1016/j.clindermatol.2013.05.021 [DOI] [PubMed] [Google Scholar]
- 28.Lippens, S., Hoste, E., Vandenabeele, P., Agostinis, P. and Declercq, W. (2009) Cell death in the skin. Apoptosis 14, 549–569 10.1007/s10495-009-0324-z [DOI] [PubMed] [Google Scholar]
- 29.Matsui, T., Kadono-Maekubo, N., Suzuki, Y., Furuichi, Y., Shiraga, K., Sasaki, H.et al. (2021) A unique mode of keratinocyte death requires intracellular acidification. Proc. Natl Acad. Sci. U.S.A. 118, e2020722118 10.1073/pnas.2020722118 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Fischer, H., Buchberger, M., Napirei, M., Tschachler, E. and Eckhart, L. (2017) Inactivation of DNase1L2 and DNase2 in keratinocytes suppresses DNA degradation during epidermal cornification and results in constitutive parakeratosis. Sci. Rep. 7, 6433 10.1038/s41598-017-06652-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Candi, E., Schmidt, R. and Melino, G. (2005) The cornified envelope: a model of cell death in the skin. Nat. Rev. Mol. Cell Biol. 6, 328–340 10.1038/nrm1619 [DOI] [PubMed] [Google Scholar]
- 32.Eckhart, L. and Tschachler, E. (2018) Control of cell death-associated danger signals during cornification prevents autoinflammation of the skin. Exp. Dermatol. 27, 884–891 10.1111/exd.13700 [DOI] [PubMed] [Google Scholar]
- 33.Elias, P.M. (2012) Structure and function of the stratum corneum extracellular matrix. J. Invest. Dermatol. 132, 2131–2133 10.1038/jid.2012.246 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sandilands, A., Sutherland, C., Irvine, A.D. and McLean, W.H. (2009) Filaggrin in the frontline: role in skin barrier function and disease. J. Cell Sci. 122(Pt 9), 1285–1294 10.1242/jcs.033969 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fischer, H., Eckhart, L., Mildner, M., Jaeger, K., Buchberger, M., Ghannadan, M.et al. (2007) DNase1l2 degrades nuclear DNA during corneocyte formation. J. Invest. Dermatol. 127, 24–30 10.1038/sj.jid.5700503 [DOI] [PubMed] [Google Scholar]
- 36.He, B., Lu, N. and Zhou, Z. (2009) Cellular and nuclear degradation during apoptosis. Curr. Opin. Cell Biol. 21, 900–912 10.1016/j.ceb.2009.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Nagata, S., Nagase, H., Kawane, K., Mukae, N. and Fukuyama, H. (2003) Degradation of chromosomal DNA during apoptosis. Cell Death Differ. 10, 108–116 10.1038/sj.cdd.4401161 [DOI] [PubMed] [Google Scholar]
- 38.Kawane, K., Motani, K. and Nagata, S. (2014) DNA degradation and its defects. Cold Spring Harb. Perspect. Biol. 6, a016394 10.1101/cshperspect.a016394 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Furio, L. and Hovnanian, A. (2014) Netherton syndrome: defective kallikrein inhibition in the skin leads to skin inflammation and allergy. Biol. Chem. 395, 945–958 10.1515/hsz-2014-0137 [DOI] [PubMed] [Google Scholar]
- 40.Chavanas, S., Bodemer, C., Rochat, A., Hamel-Teillac, D., Ali, M., Irvine, A.D.et al. (2000) Mutations in SPINK5, encoding a serine protease inhibitor, cause Netherton syndrome. Nat. Genet. 25, 141–142 10.1038/75977 [DOI] [PubMed] [Google Scholar]
- 41.Lachner, J., Mlitz, V., Tschachler, E. and Eckhart, L. (2017) Epidermal cornification is preceded by the expression of a keratinocyte-specific set of pyroptosis-related genes. Sci. Rep. 7, 17446 10.1038/s41598-017-17782-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Martin, S.J., Henry, C.M. and Cullen, S.P. (2012) A perspective on mammalian caspases as positive and negative regulators of inflammation. Mol. Cell 46, 387–397 10.1016/j.molcel.2012.04.026 [DOI] [PubMed] [Google Scholar]
- 43.Czabotar, P.E., Lessene, G., Strasser, A. and Adams, J.M. (2014) Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 15, 49–63 10.1038/nrm3722 [DOI] [PubMed] [Google Scholar]
- 44.Delbridge, A.R., Grabow, S., Strasser, A. and Vaux, D.L. (2016) Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies. Nat. Rev. Cancer 16, 99–109 10.1038/nrc.2015.17 [DOI] [PubMed] [Google Scholar]
- 45.Adrain, C., Creagh, E.M. and Martin, S.J. (2001) Apoptosis-associated release of Smac/DIABLO from mitochondria requires active caspases and is blocked by Bcl-2. EMBO J. 20, 6627–6636 10.1093/emboj/20.23.6627 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Du, C., Fang, M., Li, Y., Li, L. and Wang, X. (2000) Smac, a mitochondrial protein that promotes cytochrome c–dependent caspase activation by eliminating IAP inhibition. Cell 102, 33–42 10.1016/S0092-8674(00)00008-8 [DOI] [PubMed] [Google Scholar]
- 47.Parsons, M.J. and Green, D.R. (2010) Mitochondria in cell death. Essays Biochem. 47, 99–114 10.1042/bse0470099 [DOI] [PubMed] [Google Scholar]
- 48.White, M.J., McArthur, K., Metcalf, D., Lane, R.M., Cambier, J.C., Herold, M.J.et al. (2014) Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production. Cell 159, 1549–1562 10.1016/j.cell.2014.11.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.McArthur, K., Whitehead, L.W., Heddleston, J.M., Li, L., Padman, B.S., Oorschot, V.et al. (2018) BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis. Science 359, eaao6047. 10.1126/science.aao6047 [DOI] [PubMed] [Google Scholar]
- 50.Botchkareva, N.V., Ahluwalia, G. and Shander, D. (2006) Apoptosis in the hair follicle. J. Invest. Dermatol. 126, 258–264 10.1038/sj.jid.5700007 [DOI] [PubMed] [Google Scholar]
- 51.Nishimura, E.K., Granter, S.R. and Fisher, D.E. (2005) Mechanisms of hair graying: incomplete melanocyte stem cell maintenance in the niche. Science 307, 720–724 10.1126/science.1099593 [DOI] [PubMed] [Google Scholar]
- 52.Geueke, A., Mantellato, G., Kuester, F., Schettina, P., Nelles, M., Seeger, J.M.et al. (2021) The anti-apoptotic Bcl-2 protein regulates hair follicle stem cell function. EMBO Rep. 22, e52301 10.15252/embr.202052301 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Amarante-Mendes, G.P., Adjemian, S., Branco, L.M., Zanetti, L.C., Weinlich, R. and Bortoluci, K.R. (2018) Pattern recognition receptors and the host cell death molecular machinery. Front. Immunol. 9, 2379 10.3389/fimmu.2018.02379 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Dickens, L.S., Boyd, R.S., Jukes-Jones, R., Hughes, M.A., Robinson, G.L., Fairall, L.et al. (2012) A death effector domain chain DISC model reveals a crucial role for caspase-8 chain assembly in mediating apoptotic cell death. Mol. Cell 47, 291–305 10.1016/j.molcel.2012.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Singh, N., Hassan, A. and Bose, K. (2016) Molecular basis of death effector domain chain assembly and its role in caspase-8 activation. FASEB J. 30, 186–200 10.1096/fj.15-272997 [DOI] [PubMed] [Google Scholar]
- 56.Jang, D.I., Lee, A.H., Shin, H.Y., Song, H.R., Park, J.H., Kang, T.B.et al. (2021) The role of tumor necrosis factor alpha (TNF-α) in autoimmune disease and current TNF-α inhibitors in therapeutics. Int. J. Mol. Sci. 22, 2719 10.3390/ijms22052719 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Jarosz-Griffiths, H.H., Holbrook, J., Lara-Reyna, S. and McDermott, M.F. (2019) TNF receptor signalling in autoinflammatory diseases. Int. Immunol. 31, 369–348 10.1093/intimm/dxz024 [DOI] [PubMed] [Google Scholar]
- 58.Silke, J. and Meier, P. (2013) Inhibitor of apoptosis (IAP) proteins-modulators of cell death and inflammation. Cold Spring Harb. Perspect. Biol. 5, a008730 10.1101/cshperspect.a008730 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Vince, J.E., Pantaki, D., Feltham, R., Mace, P.D., Cordier, S.M., Schmukle, A.C.et al. (2009) TRAF2 must bind to cellular inhibitors of apoptosis for tumor necrosis factor (tnf) to efficiently activate nf-{kappa}b and to prevent tnf-induced apoptosis. J. Biol. Chem. 284, 35906–35915 10.1074/jbc.M109.072256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bertrand, M.J., Milutinovic, S., Dickson, K.M., Ho, W.C., Boudreault, A., Durkin, J.et al. (2008) cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol. Cell 30, 689–700 10.1016/j.molcel.2008.05.014 [DOI] [PubMed] [Google Scholar]
- 61.Dynek, J.N., Goncharov, T., Dueber, E.C., Fedorova, A.V., Izrael-Tomasevic, A., Phu, L.et al. (2010) c-IAP1 and UbcH5 promote K11-linked polyubiquitination of RIP1 in TNF signalling. EMBO J. 29, 4198–4209 10.1038/emboj.2010.300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Varfolomeev, E., Goncharov, T., Fedorova, A.V., Dynek, J.N., Zobel, K., Deshayes, K.et al. (2008) c-IAP1 and c-IAP2 are critical mediators of tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB activation. J. Biol. Chem. 283, 24295–24299 10.1074/jbc.C800128200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ting, A.T. and Bertrand, M.J.M. (2016) More to life than NF-kappaB in TNFR1 signaling. Trends Immunol. 37, 535–545 10.1016/j.it.2016.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Varfolomeev, E. and Vucic, D. (2018) Intracellular regulation of TNF activity in health and disease. Cytokine 101, 26–32 10.1016/j.cyto.2016.08.035 [DOI] [PubMed] [Google Scholar]
- 65.Gerlach, B., Cordier, S.M., Schmukle, A.C., Emmerich, C.H., Rieser, E., Haas, T.L.et al. (2011) Linear ubiquitination prevents inflammation and regulates immune signalling. Nature 471, 591–596 10.1038/nature09816 [DOI] [PubMed] [Google Scholar]
- 66.Haas, T.L., Emmerich, C.H., Gerlach, B., Schmukle, A.C., Cordier, S.M., Rieser, E.et al. (2009) Recruitment of the linear ubiquitin chain assembly complex stabilizes the TNF-R1 signaling complex and is required for TNF-mediated gene induction. Mol. Cell 36, 831–844 10.1016/j.molcel.2009.10.013 [DOI] [PubMed] [Google Scholar]
- 67.Tokunaga, F., Sakata, S., Saeki, Y., Satomi, Y., Kirisako, T., Kamei, K.et al. (2009) Involvement of linear polyubiquitylation of NEMO in NF-kappaB activation. Nat. Cell Biol. 11, 123–132 10.1038/ncb1821 [DOI] [PubMed] [Google Scholar]
- 68.Tokunaga, F., Nakagawa, T., Nakahara, M., Saeki, Y., Taniguchi, M., Sakata, S.et al. (2011) SHARPIN is a component of the NF-kappaB-activating linear ubiquitin chain assembly complex. Nature 471, 633–636 10.1038/nature09815 [DOI] [PubMed] [Google Scholar]
- 69.Tsuchiya, Y., Nakabayashi, O. and Nakano, H. (2015) FLIP the switch: regulation of apoptosis and necroptosis by cFLIP. Int. J. Mol. Sci. 16, 30321–30341 10.3390/ijms161226232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Annibaldi, A., Wicky John, S., Vanden Berghe, T., Swatek, K.N., Ruan, J., Liccardi, G.et al. (2018) Ubiquitin-mediated regulation of RIPK1 kinase activity independent of IKK and MK2. Mol. Cell 69, 566–580.e5 10.1016/j.molcel.2018.01.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dondelinger, Y., Delanghe, T., Priem, D., Wynosky-Dolfi, M.A., Sorobetea, D., Rojas-Rivera, D.et al. (2019) Serine 25 phosphorylation inhibits RIPK1 kinase-dependent cell death in models of infection and inflammation. Nat. Commun. 10, 1729 10.1038/s41467-019-09690-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jaco, I., Annibaldi, A., Lalaoui, N., Wilson, R., Tenev, T., Laurien, L.et al. (2017) MK2 phosphorylates RIPK1 to prevent TNF-induced cell death. Mol. Cell 66, 698–710.e5 10.1016/j.molcel.2017.05.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Dondelinger, Y., Jouan-Lanhouet, S., Divert, T., Theatre, E., Bertin, J., Gough, P.J.et al. (2015) NF-kappaB-independent role of IKKalpha/IKKbeta in preventing RIPK1 kinase-dependent apoptotic and necroptotic cell death during TNF signaling. Mol. Cell 60, 63–76 10.1016/j.molcel.2015.07.032 [DOI] [PubMed] [Google Scholar]
- 74.Geng, J., Ito, Y., Shi, L., Amin, P., Chu, J., Ouchida, A.T.et al. (2017) Regulation of RIPK1 activation by TAK1-mediated phosphorylation dictates apoptosis and necroptosis. Nat. Commun. 8, 359 10.1038/s41467-017-00406-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Deveraux, Q.L., Roy, N., Stennicke, H.R., Van Arsdale, T., Zhou, Q., Srinivasula, S.M.et al. (1998) IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. EMBO J. 17, 2215–2223 10.1093/emboj/17.8.2215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Srinivasula, S.M., Hegde, R., Saleh, A., Datta, P., Shiozaki, E., Chai, J.et al. (2001) A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature 410, 112–116 10.1038/35065125 [DOI] [PubMed] [Google Scholar]
- 77.Scott, F.L., Denault, J.B., Riedl, S.J., Shin, H., Renatus, M. and Salvesen, G.S. (2005) XIAP inhibits caspase-3 and -7 using two binding sites: evolutionarily conserved mechanism of IAPs. EMBO J. 24, 645–655 10.1038/sj.emboj.7600544 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Rehm, M., Düßmann, H. and Prehn, J.H.M. (2003) Real-time single cell analysis of Smac/DIABLO release during apoptosis. J. Cell Biol. 162, 1031–1043 10.1083/jcb.200303123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Hansen, T.M., Smith, D.J. and Nagley, P. (2006) Smac/DIABLO is not released from mitochondria during apoptotic signalling in cells deficient in cytochrome c. Cell Death Differ. 13, 1181–1190 10.1038/sj.cdd.4401795 [DOI] [PubMed] [Google Scholar]
- 80.Luthi, A.U., Cullen, S.P., McNeela, E.A., Duriez, P.J., Afonina, I.S., Sheridan, C.et al. (2009) Suppression of interleukin-33 bioactivity through proteolysis by apoptotic caspases. Immunity 31, 84–98 10.1016/j.immuni.2009.05.007 [DOI] [PubMed] [Google Scholar]
- 81.Yamada, Y., Fujii, T., Ishijima, R., Tachibana, H., Yokoue, N., Takasawa, R.et al. (2011) The release of high mobility group box 1 in apoptosis is triggered by nucleosomal DNA fragmentation. Arch. Biochem. Biophys. 506, 188–193 10.1016/j.abb.2010.11.011 [DOI] [PubMed] [Google Scholar]
- 82.Green, D.R., Oguin, T.H. and Martinez, J. (2016) The clearance of dying cells: table for two. Cell Death Differ. 23, 915–926 10.1038/cdd.2015.172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Haensel, D., Jin, S., Sun, P., Cinco, R., Dragan, M., Nguyen, Q.et al. (2020) Defining epidermal basal cell states during skin homeostasis and wound healing using single-cell transcriptomics. Cell Rep. 30, 3932–3947.e6 10.1016/j.celrep.2020.02.091 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hu, X.M., Li, Z.X., Zhang, D.Y., Yang, Y.C., Fu, S.A., Zhang, Z.Q.et al. (2021) A systematic summary of survival and death signalling during the life of hair follicle stem cells. Stem Cell Res. Ther. 12, 453 10.1186/s13287-021-02527-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mistry, P. and Kaplan, M.J. (2017) Cell death in the pathogenesis of systemic lupus erythematosus and lupus nephritis. Clin. Immunol. 185, 59–73 10.1016/j.clim.2016.08.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Martens, M.C., Seebode, C., Lehmann, J. and Emmert, S. (2018) Photocarcinogenesis and skin cancer prevention strategies: an update. Anticancer Res. 38, 1153–1158 10.21873/anticanres.12334 [DOI] [PubMed] [Google Scholar]
- 87.Wang, H., Zhang, M., Xu, X., Hou, S., Liu, Z., Chen, X.et al. (2021) IKKα mediates UVB-induced cell apoptosis by regulating p53 pathway activation. Ecotoxicol. Environ. Saf. 227, 112892 10.1016/j.ecoenv.2021.112892 [DOI] [PubMed] [Google Scholar]
- 88.Yogosawa, S. and Yoshida, K. (2018) Tumor suppressive role for kinases phosphorylating p53 in DNA damage-induced apoptosis. Cancer Sci. 109, 3376–3382 10.1111/cas.13792 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Brown, D.L., Kao, W.W.Y. and Greenhalgh, D.G. (1997) Apoptosis down-regulates inflammation under the advancing epithelial wound edge: delayed patterns in diabetes and improvement with topical growth factors. Surgery 121, 372–380 10.1016/S0039-6060(97)90306-8 [DOI] [PubMed] [Google Scholar]
- 90.Rodrigues, M., Kosaric, N., Bonham, C.A. and Gurtner, G.C. (2019) Wound healing: a cellular perspective. Physiol. Rev. 99, 665–706 10.1152/physrev.00067.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wu, Y.S. and Chen, S.N. (2014) Apoptotic cell: linkage of inflammation and wound healing. Front. Pharmacol. 5, 1 10.3389/fphar.2014.00001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Ellis, S., Lin, E.J. and Tartar, D. (2018) Immunology of wound healing. Curr. Dermatol. Rep. 7, 350–358 10.1007/s13671-018-0234-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Piipponen, M., Li, D. and Landén, N.X. (2020) The immune functions of keratinocytes in skin wound healing. Int. J. Mol. Sci. 21, 8790 10.3390/ijms21228790 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Cañedo-Dorantes, L. and Cañedo-Ayala, M. (2019) Skin acute wound healing: a comprehensive review. Int. J. Inflamm. 2019, e3706315 10.1155/2019/3706315 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Green, D.R., Ferguson, T., Zitvogel, L. and Kroemer, G. (2009) Immunogenic and tolerogenic cell death. Nat. Rev. Immunol. 9, 353–363 10.1038/nri2545 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Peter, C., Wesselborg, S., Herrmann, M. and Lauber, K. (2010) Dangerous attraction: phagocyte recruitment and danger signals of apoptotic and necrotic cells. Apoptosis 15, 1007–1028 10.1007/s10495-010-0472-1 [DOI] [PubMed] [Google Scholar]
- 97.Silva, M.T. (2010) Secondary necrosis: the natural outcome of the complete apoptotic program. FEBS Lett. 584, 4491–4499 10.1016/j.febslet.2010.10.046 [DOI] [PubMed] [Google Scholar]
- 98.Kayagaki, N., Kornfeld, O.S., Lee, B.L., Stowe, I.B., O'Rourke, K., Li, Q.et al. (2021) NINJ1 mediates plasma membrane rupture during lytic cell death. Nature 591, 131–136 10.1038/s41586-021-03218-7 [DOI] [PubMed] [Google Scholar]
- 99.Maß, P., Hoffmann, K., Gambichler, T., Altmeyer, P. and Mannherz, H.G. (2003) Premature keratinocyte death and expression of marker proteins of apoptosis in human skin after UVB exposure. Arch. Dermatol. Res. 295, 71–79 10.1007/s00403-003-0403-x [DOI] [PubMed] [Google Scholar]
- 100.Anderton, H., Rickard, J.A., Varigos, G.A., Lalaoui, N. and Silke, J. (2017) Inhibitor of apoptosis proteins (IAPs) limit RIPK1-mediated skin inflammation. J. Invest. Dermatol. 137, 2371–2379 10.1016/j.jid.2017.05.031 [DOI] [PubMed] [Google Scholar]
- 101.Panayotova-Dimitrova, D., Feoktistova, M., Ploesser, M., Kellert, B., Hupe, M., Horn, S.et al. (2013) cFLIP regulates skin homeostasis and protects against TNF-induced keratinocyte apoptosis. Cell Rep. 5, 397–408 10.1016/j.celrep.2013.09.035 [DOI] [PubMed] [Google Scholar]
- 102.Etemadi, N., Chopin, M., Anderton, H., Tanzer, M.C., Rickard, J.A., Abeysekera, W.et al. (2015) TRAF2 regulates TNF and NF-kappaB signalling to suppress apoptosis and skin inflammation independently of Sphingosine kinase 1. Elife 4, 1–27 10.7554/eLife.10592 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.HogenEsch, H., Gijbels, M.J., Offerman, E., van Hooft, J., van Bekkum, D.W. and Zurcher, C. (1993) A spontaneous mutation characterized by chronic proliferative dermatitis in C57BL mice. Am. J. Pathol. 143, 972–982 PMID: [PMC free article] [PubMed] [Google Scholar]
- 104.Rickard, J.A., Anderton, H., Etemadi, N., Nachbur, U., Darding, M., Peltzer, N.et al. (2014) TNFR1-dependent cell death drives inflammation in Sharpin-deficient mice. Elife 3, 1–23 10.7554/eLife.03464 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Teh, C.E., Lalaoui, N., Jain, R., Policheni, A.N., Heinlein, M., Alvarez-Diaz, S.et al. (2016) Linear ubiquitin chain assembly complex coordinates late thymic T-cell differentiation and regulatory T-cell homeostasis. Nat. Commun. 7, 13353 10.1038/ncomms13353 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Zak, D.E., Schmitz, F., Gold, E.S., Diercks, A.H., Peschon, J.J., Valvo, J.S.et al. (2011) Systems analysis identifies an essential role for SHANK-associated RH domain-interacting protein (SHARPIN) in macrophage Toll-like receptor 2 (TLR2) responses. Proc. Natl Acad. Sci. U.S.A. 108, 11536–11541 10.1073/pnas.1107577108 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kumari, S., Redouane, Y., Lopez-Mosqueda, J., Shiraishi, R., Romanowska, M., Lutzmayer, S.et al. (2014) Sharpin prevents skin inflammation by inhibiting TNFR1-induced keratinocyte apoptosis. Elife 3, e03422. 10.7554/eLife.03422 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Anderton, H., Chopin, M., Dawson, C.A., Nutt, S.L., Whitehead, L., Silke, N.et al. (2022) Langerhans cells are an essential cellular intermediary in chronic dermatitis. Cell Rep. 39, 110922 10.1016/j.celrep.2022.110922 [DOI] [PubMed] [Google Scholar]
- 109.Kim, E.H., Wong, S.W. and Martinez, J. (2019) Programmed necrosis and disease: we interrupt your regular programming to bring you necroinflammation. Cell Death Differ. 26, 25–40 10.1038/s41418-018-0179-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Feng, S., Yang, Y., Mei, Y., Ma, L., Zhu, D.E., Hoti, N.et al. (2007) Cleavage of RIP3 inactivates its caspase-independent apoptosis pathway by removal of kinase domain. Cell Signal. 19, 2056–2067 10.1016/j.cellsig.2007.05.016 [DOI] [PubMed] [Google Scholar]
- 111.Cho, Y.S., Challa, S., Moquin, D., Genga, R., Ray, T.D., Guildford, M.et al. (2009) Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 137, 1112–1123 10.1016/j.cell.2009.05.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Li, J., McQuade, T., Siemer, A.B., Napetschnig, J., Moriwaki, K., Hsiao, Y.S.et al. (2012) The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 150, 339–350 10.1016/j.cell.2012.06.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.McQuade, T., Cho, Y. and Chan, F.K.M. (2013) Positive and negative phosphorylation regulates RIP1- and RIP3-induced programmed necrosis. Biochem. J. 456, 409–415 10.1042/BJ20130860 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Huang,, D., Zheng,, X., Wang,, Z.A., Chen,, X., He W,, T., Zhang,, Y., et al. (2017) The MLKL channel in necroptosis is an octamer formed by tetramers in a dyadic process. Mol. Cell. Biol. 37, e00497-16 10.1128/MCB.00497-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Murphy, J.M., Czabotar, P.E., Hildebrand, J.M., Lucet, I.S., Zhang, J.G., Alvarez-Diaz, S.et al. (2013) The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 39, 443–453 10.1016/j.immuni.2013.06.018 [DOI] [PubMed] [Google Scholar]
- 116.Xia, B., Fang, S., Chen, X., Hu, H., Chen, P., Wang, H.et al. (2016) MLKL forms cation channels. Cell Res. 26, 517–528 10.1038/cr.2016.26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Cai, Z., Jitkaew, S., Zhao, J., Chiang, H.C., Choksi, S., Liu, J.et al. (2014) Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat. Cell Biol. 16, 55–65 10.1038/ncb2883 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Samson, A.L., Zhang, Y., Geoghegan, N.D., Gavin, X.J., Davies, K.A., Mlodzianoski, M.J.et al. (2020) MLKL trafficking and accumulation at the plasma membrane control the kinetics and threshold for necroptosis. Nat. Commun. 11, 3151 10.1038/s41467-020-16887-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lalaoui, N., Boyden, S.E., Oda, H., Wood, G.M., Stone, D.L., Chau, D.et al. (2020) Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease. Nature 577, 103–108 10.1038/s41586-019-1828-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Newton, K., Wickliffe, K.E., Dugger, D.L., Maltzman, A., Roose-Girma, M., Dohse, M.et al. (2019) Cleavage of RIPK1 by caspase-8 is crucial for limiting apoptosis and necroptosis. Nature 574, 428–431 10.1038/s41586-019-1548-x [DOI] [PubMed] [Google Scholar]
- 121.Zhang, X., Dowling, J.P. and Zhang, J. (2019) RIPK1 can mediate apoptosis in addition to necroptosis during embryonic development. Cell Death Dis. 10, 245 10.1038/s41419-019-1490-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Lin, Y., Devin, A., Rodriguez, Y. and Liu, Z.G. (1999) Cleavage of the death domain kinase RIP by caspase-8 prompts TNF-induced apoptosis. Genes Dev. 13, 2514–2526 10.1101/gad.13.19.2514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Zhang, L., Blackwell, K., Workman, L.M., Chen, S., Pope, M.R., Janz, S.et al. (2015) RIP1 cleavage in the kinase domain regulates TRAIL-induced NF-kappaB activation and lymphoma survival. Mol. Cell. Biol. 35, 3324–3338 10.1128/MCB.00692-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kitur, K., Wachtel, S., Brown, A., Wickersham, M., Paulino, F., Peñaloza, H.F.et al. (2016) Necroptosis promotes Staphylococcus aureus clearance by inhibiting excessive inflammatory signaling. Cell Rep. 16, 2219–2230 10.1016/j.celrep.2016.07.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Dondelinger, Y., Darding, M., Bertrand, M.J. and Walczak, H. (2016) Poly-ubiquitination in TNFR1-mediated necroptosis. Cell. Mol. Life Sci. 73, 2165–2176 10.1007/s00018-016-2191-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Festjens, N., Vanden Berghe, T., Cornelis, S. and Vandenabeele, P. (2007) RIP1, a kinase on the crossroads of a cell's decision to live or die. Cell Death Differ. 14, 400–410 10.1038/sj.cdd.4402085 [DOI] [PubMed] [Google Scholar]
- 127.Pasparakis, M. and Vandenabeele, P. (2015) Necroptosis and its role in inflammation. Nature 517, 311–320 10.1038/nature14191 [DOI] [PubMed] [Google Scholar]
- 128.Feoktistova, M., Geserick, P., Kellert, B., Dimitrova, D.P., Langlais, C., Hupe, M.et al. (2011) cIAPs block Ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol. Cell 43, 449–463 10.1016/j.molcel.2011.06.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Lawlor, K.E., Khan, N., Mildenhall, A., Gerlic, M., Croker, B.A., D'Cruz, A.A.et al. (2015) RIPK3 promotes cell death and NLRP3 inflammasome activation in the absence of MLKL. Nat. Commun. 6, 6282 10.1038/ncomms7282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Grootjans, S., Vanden Berghe, T. and Vandenabeele, P. (2017) Initiation and execution mechanisms of necroptosis: an overview. Cell Death Differ. 24, 1184–1195 10.1038/cdd.2017.65 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lin, J., Kumari, S., Kim, C., Van, T.M., Wachsmuth, L., Polykratis, A.et al. (2016) RIPK1 counteracts ZBP1-mediated necroptosis to inhibit inflammation. Nature 540, 124–128 10.1038/nature20558 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Newton, K., Wickliffe, K.E., Maltzman, A., Dugger, D.L., Strasser, A., Pham, V.C.et al. (2016) RIPK1 inhibits ZBP1-driven necroptosis during development. Nature 540, 129–133 10.1038/nature20559 [DOI] [PubMed] [Google Scholar]
- 133.Kaiser, W.J., Sridharan, H., Huang, C., Mandal, P., Upton, J.W., Gough, P.J.et al. (2013) Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J. Biol. Chem. 288, 31268–31279 10.1074/jbc.M113.462341 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Kumari, S., Van, T.M., Preukschat, D., Schuenke, H., Basic, M., Bleich, A.et al. (2021) NF-κB inhibition in keratinocytes causes RIPK1-mediated necroptosis and skin inflammation. Life Sci. Alliance 4, e202000956 10.26508/lsa.202000956 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Nenci, A., Becker, C., Wullaert, A., Gareus, R., van Loo, G., Danese, S.et al. (2007) Epithelial NEMO links innate immunity to chronic intestinal inflammation. Nature 446, 557–561 10.1038/nature05698 [DOI] [PubMed] [Google Scholar]
- 136.Pasparakis, M., Courtois, G., Hafner, M., Schmidt-Supprian, M., Nenci, A., Toksoy, A.et al. (2002) TNF-mediated inflammatory skin disease in mice with epidermis-specific deletion of IKK2. Nature 417, 861–866 10.1038/nature00820 [DOI] [PubMed] [Google Scholar]
- 137.Grinberg-Bleyer, Y., Dainichi, T., Oh, H., Heise, N., Klein, U., Schmid, R.M.et al. (2015) NF-κB p65 and c-Rel control epidermal development and immune homeostasis in the skin. J. Immunol. 194, 2472–2476 10.4049/jimmunol.1402608 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Kumari, S., Bonnet, M.C., Ulvmar, M.H., Wolk, K., Karagianni, N., Witte, E.et al. (2013) Tumor necrosis factor receptor signaling in keratinocytes triggers interleukin-24-dependent psoriasis-like skin inflammation in mice. Immunity 39, 899–911 10.1016/j.immuni.2013.10.009 [DOI] [PubMed] [Google Scholar]
- 139.Dannappel, M., Vlantis, K., Kumari, S., Polykratis, A., Kim, C., Wachsmuth, L.et al. (2014) RIPK1 maintains epithelial homeostasis by inhibiting apoptosis and necroptosis. Nature 513, 90–94 10.1038/nature13608 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Bergsbaken, T., Fink, S.L. and Cookson, B.T. (2009) Pyroptosis: host cell death and inflammation. Nat. Rev. Microbiol. 7, 99–109 10.1038/nrmicro2070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Miao, E.A., Leaf, I.A., Treuting, P.M., Mao, D.P., Dors, M., Sarkar, A.et al. (2010) Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat. Immunol. 11, 1136–1142 10.1038/ni.1960 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Miao, E.A., Rajan, J.V. and Aderem, A. (2011) Caspase-1-induced pyroptotic cell death. Immunol. Rev. 243, 206–214 10.1111/j.1600-065X.2011.01044.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Downs, K.P., Nguyen, H., Dorfleutner, A. and Stehlik, C. (2020) An overview of the non-canonical inflammasome. Mol. Aspects Med. 76, 100924 10.1016/j.mam.2020.100924 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Huang, X., Feng, Y., Xiong, G., Whyte, S., Duan, J., Yang, Y.et al. (2019) Caspase-11, a specific sensor for intracellular lipopolysaccharide recognition, mediates the non-canonical inflammatory pathway of pyroptosis. Cell Biosci. 9, 31 10.1186/s13578-019-0292-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Kayagaki, N., Stowe, I.B., Lee, B.L., O'Rourke, K., Anderson, K., Warming, S.et al. (2015) Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526, 666–671 10.1038/nature15541 [DOI] [PubMed] [Google Scholar]
- 146.Shi, J., Zhao, Y., Wang, Y., Gao, W., Ding, J., Li, P.et al. (2014) Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 514, 187–192 10.1038/nature13683 [DOI] [PubMed] [Google Scholar]
- 147.Guo, H., Callaway, J.B. and Ting, J.P. (2015) Inflammasomes: mechanism of action, role in disease, and therapeutics. Nat. Med. 21, 677–687 10.1038/nm.3893 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Jo, E.K., Kim, J.K., Shin, D.M. and Sasakawa, C. (2016) Molecular mechanisms regulating NLRP3 inflammasome activation. Cell. Mol. Immunol. 13, 148–159 10.1038/cmi.2015.95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Benhadou, F., Mintoff, D., Schnebert, B. and Thio, H. (2018) Psoriasis and microbiota: a systematic review. Diseases 6, 47 10.3390/diseases6020047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Foster, K.R., Schluter, J., Coyte, K.Z. and Rakoff-Nahoum, S. (2017) The evolution of the host microbiome as an ecosystem on a leash. Nature 548, 43–51 10.1038/nature23292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Tang, C., Makusheva, Y., Sun, H., Han, W. and Iwakura, Y. (2019) Myeloid C-type lectin receptors in skin/mucoepithelial diseases and tumors. J. Leukoc. Biol. 106, 903–917 10.1002/JLB.2RI0119-031R [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Schroder, K. and Tschopp, J. (2010) The inflammasomes. Cell 140, 821–832 10.1016/j.cell.2010.01.040 [DOI] [PubMed] [Google Scholar]
- 153. The Human Protein Atlas. (2008) proteinatlas.org.
- 154.Thul, P.J., et al. (2017) A subcellular map of the human proteome. Science PMID: 10.1126/science.aal3321 [DOI] [PubMed] [Google Scholar]
- 155.Thul, P.J., Åkesson, L., Wiking, M., Mahdessian, D., Geladaki, A., Ait Blal, H.et al. (2017) A subcellular map of the human proteome. Science 356, eaal3321 10.1126/science.aal3321 [DOI] [PubMed] [Google Scholar]
- 156.Uhlén, M., Fagerberg, L., Hallström, B.M., Lindskog, C., Oksvold, P., Mardinoglu, A.et al. (2015) Tissue-based map of the human proteome. Science 347, 1260419 10.1126/science.1260419 [DOI] [PubMed] [Google Scholar]
- 157.Latz, E., Xiao, T.S. and Stutz, A. (2013) Activation and regulation of the inflammasomes. Nat. Rev. Immunol. 13, 397–411 10.1038/nri3452 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.He, W.T., Wan, H., Hu, L., Chen, P., Wang, X., Huang, Z.et al. (2015) Gasdermin D is an executor of pyroptosis and required for interleukin-1beta secretion. Cell Res. 25, 1285–1298 10.1038/cr.2015.139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Sborgi, L., Ruhl, S., Mulvihill, E., Pipercevic, J., Heilig, R., Stahlberg, H.et al. (2016) GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death. EMBO J. 35, 1766–1778 10.15252/embj.201694696 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Frank, D. and Vince, J.E. (2019) Pyroptosis versus necroptosis: similarities, differences, and crosstalk. Cell Death Differ. 26, 99–114 10.1038/s41418-018-0212-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Shi, J., Gao, W. and Shao, F. (2017) Pyroptosis: gasdermin-mediated programmed necrotic cell death. Trends Biochem. Sci. 42, 245–254 10.1016/j.tibs.2016.10.004 [DOI] [PubMed] [Google Scholar]
- 162.Ruan, J., Xia, S., Liu, X., Lieberman, J. and Wu, H. (2018) Cryo-EM structure of the gasdermin A3 membrane pore. Nature 557, 62–67 10.1038/s41586-018-0058-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Tsuchiya, K., Hosojima, S., Hara, H., Kushiyama, H., Mahib, M.R., Kinoshita, T.et al. (2021) Gasdermin D mediates the maturation and release of IL-1α downstream of inflammasomes. Cell Rep. 34, 108887. 10.1016/j.celrep.2021.108887 [DOI] [PubMed] [Google Scholar]
- 164.Lopez-Castejon, G. and Brough, D. (2011) Understanding the mechanism of IL-1β secretion. Cytokine Growth Factor Rev. 22, 189–195 10.1016/j.cytogfr.2011.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Sollberger, G. (2021) Approaching neutrophil pyroptosis. J. Mol. Biol. 434, 167335 10.1016/j.jmb.2021.167335 [DOI] [PubMed] [Google Scholar]
- 166.Rühl, S., Shkarina, K., Demarco, B., Heilig, R., Santos, J.C. and Broz, P. (2018) ESCRT-dependent membrane repair negatively regulates pyroptosis downstream of GSDMD activation. Science 362, 956–960 10.1126/science.aar7607 [DOI] [PubMed] [Google Scholar]
- 167.Conos, S.A., Lawlor, K.E., Vaux, D.L., Vince, J.E. and Lindqvist, L.M. (2016) Cell death is not essential for caspase-1-mediated interleukin-1beta activation and secretion. Cell Death Differ. 23, 1827–1838 10.1038/cdd.2016.69 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Martin-Sanchez, F., Diamond, C., Zeitler, M., Gomez, A.I., Baroja-Mazo, A., Bagnall, J.et al. (2016) Inflammasome-dependent IL-1beta release depends upon membrane permeabilisation. Cell Death Differ. 23, 1219–1231 10.1038/cdd.2015.176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Rashidi, M., Simpson, D.S., Hempel, A., Frank, D., Petrie, E., Vince, A.et al. (2019) The pyroptotic cell death effector gasdermin D is activated by gout-associated uric acid crystals but is dispensable for cell death and IL-1beta release. J. Immunol. 203, 736–748 10.4049/jimmunol.1900228 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Gurcel, L., Abrami, L., Girardin, S., Tschopp, J. and van der Goot, F.G. (2006) Caspase-1 activation of lipid metabolic pathways in response to bacterial pore-forming toxins promotes cell survival. Cell 126, 1135–1145 10.1016/j.cell.2006.07.033 [DOI] [PubMed] [Google Scholar]
- 171.Monteleone, M., Stanley, A.C., Chen, K.W., Brown, D.L., Bezbradica, J.S., von Pein, J.B.et al. (2018) Interleukin-1β maturation triggers its relocation to the plasma membrane for gasdermin-D-dependent and -independent secretion. Cell Rep. 24, 1425–1433 10.1016/j.celrep.2018.07.027 [DOI] [PubMed] [Google Scholar]
- 172.Feldmeyer, L., Keller, M., Niklaus, G., Hohl, D., Werner, S. and Beer, H.D. (2007) The inflammasome mediates UVB-induced activation and secretion of interleukin-1β by keratinocytes. Curr. Biol. 17, 1140–1145 10.1016/j.cub.2007.05.074 [DOI] [PubMed] [Google Scholar]
- 173.Chu, H. and Mazmanian, S.K. (2013) Innate immune recognition of the microbiota promotes host-microbial symbiosis. Nat. Immunol. 14, 668–675 10.1038/ni.2635 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Burian, M. and Yazdi, A.S. (2018) NLRP1 is the key inflammasome in primary human keratinocytes. J. Invest. Dermatol. 138, 2507–2510 10.1016/j.jid.2018.08.004 [DOI] [PubMed] [Google Scholar]
- 175.Fenini, G., Grossi, S., Contassot, E., Biedermann, T., Reichmann, E., French, L.E.et al. (2018) Genome editing of human primary keratinocytes by CRISPR/Cas9 reveals an essential role of the NLRP1 inflammasome in UVB sensing. J. Invest. Dermatol. 138, 2644–2652 10.1016/j.jid.2018.07.016 [DOI] [PubMed] [Google Scholar]
- 176.Zhong, F.L., Mamaï, O., Sborgi, L., Boussofara, L., Hopkins, R., Robinson, K.et al. (2016) Germline NLRP1 mutations cause skin inflammatory and cancer susceptibility syndromes via inflammasome activation. Cell 167, 187–202.e17 10.1016/j.cell.2016.09.001 [DOI] [PubMed] [Google Scholar]
- 177.Fenini, G., Karakaya, T., Hennig, P., Di Filippo, M. and Beer, H.D. (2020) The NLRP1 inflammasome in human skin and beyond. Int. J. Mol. Sci. 21, 4788 10.3390/ijms21134788 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Grossi, S., Fenini, G., Kockmann, T., Hennig, P., Filippo, M.D. and Beer, H.D. (2020) Inactivation of the cytoprotective major vault protein by caspase-1 and -9 in epithelial cells during apoptosis. J. Invest. Dermatol. 140, 1335–1345.e10 10.1016/j.jid.2019.11.015 [DOI] [PubMed] [Google Scholar]
- 179.Sollberger, G., Strittmatter, G.E., Grossi, S., Garstkiewicz, M., auf dem Keller, U., French, L.E.et al. (2015) Caspase-1 activity is required for UVB-induced apoptosis of human keratinocytes. J. Invest. Dermatol. 135, 1395–1404 10.1038/jid.2014.551 [DOI] [PubMed] [Google Scholar]
- 180.Delgado-Rizo, V., Martínez-Guzmán, M., Iñiguez-Gutierrez, L., García-Orozco, A., Alvarado-Navarro, A. and Fafutis-Morris, M. (2017) Neutrophil extracellular traps and its implications in inflammation: an overview. Front. Immunol. 8, 81 10.3389/fimmu.2017.00081 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Lee, K.H., Kronbichler, A., Park, D.D., Park, Y., Moon, H., Kim, H.et al. (2017) Neutrophil extracellular traps (NETs) in autoimmune diseases: a comprehensive review. Autoimmun. Rev. 16, 1160–1173 10.1016/j.autrev.2017.09.012 [DOI] [PubMed] [Google Scholar]
- 182.Goldmann, O. and Medina, E. (2012) The expanding world of extracellular traps: not only neutrophils but much more. Front. Immunol. 3, 420 10.3389/fimmu.2012.00420 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Lin, A.M., Rubin, C.J., Khandpur, R., Wang, J.Y., Riblett, M., Yalavarthi, S.et al. (2011) Mast cells and neutrophils release IL-17 through extracellular trap formation in psoriasis. J. Immunol. 187, 490–500 10.4049/jimmunol.1100123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Morshed, M., Hlushchuk, R., Simon, D., Walls, A.F., Obata-Ninomiya, K., Karasuyama, H.et al. (2014) NADPH oxidase-independent formation of extracellular DNA traps by basophils. J. Immunol. 192, 5314–5323 10.4049/jimmunol.1303418 [DOI] [PubMed] [Google Scholar]
- 185.Yousefi, S., Gold, J.A., Andina, N., Lee, J.J., Kelly, A.M., Kozlowski, E.et al. (2008) Catapult-like release of mitochondrial DNA by eosinophils contributes to antibacterial defense. Nat. Med. 14, 949–953 10.1038/nm.1855 [DOI] [PubMed] [Google Scholar]
- 186.Branzk, N., Lubojemska, A., Hardison, S.E., Wang, Q., Gutierrez, M.G., Brown, G.D.et al. (2014) Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens. Nat. Immunol. 15, 1017–1025 10.1038/ni.2987 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Brinkmann, V., Reichard, U., Goosmann, C., Fauler, B., Uhlemann, Y., Weiss, D.S.et al. (2004) Neutrophil extracellular traps kill bacteria. Science 303, 1532–1535 10.1126/science.1092385 [DOI] [PubMed] [Google Scholar]
- 188.Remijsen, Q., Kuijpers, T.W., Wirawan, E., Lippens, S., Vandenabeele, P. and Vanden Berghe, T. (2011) Dying for a cause: NETosis, mechanisms behind an antimicrobial cell death modality. Cell Death Differ. 18, 581–588 10.1038/cdd.2011.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Sabbatini, M., Magnelli, V. and Renò, F. (2021) NETosis in wound healing: when enough is enough. Cells 10, 494 10.3390/cells10030494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Mutua, V. and Gershwin, L.J. (2020) A review of neutrophil extracellular traps (NETs) in disease: potential anti-NETs therapeutics. Clin. Rev. Allergy Immunol. 61, 194–211 10.1007/s12016-020-08804-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Lee, Y.S., Kang, S.U., Lee, M.H., Kim, H.J., Han, C.H., Won, H.R.et al. (2020) GnRH impairs diabetic wound healing through enhanced NETosis. Cell. Mol. Immunol. 17, 856–864 10.1038/s41423-019-0252-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Leshner, M., Wang, S., Lewis, C., Zheng, H., Chen, X., Santy, L.et al. (2012) PAD4 mediated histone hypercitrullination induces heterochromatin decondensation and chromatin unfolding to form neutrophil extracellular trap-like structures. Front. Immunol. 3, 307 10.3389/fimmu.2012.00307 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Metzler, K.D., Goosmann, C., Lubojemska, A., Zychlinsky, A. and Papayannopoulos, V. (2014) A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis. Cell Rep. 8, 883–896 10.1016/j.celrep.2014.06.044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Papayannopoulos, V., Metzler, K.D., Hakkim, A. and Zychlinsky, A. (2010) Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J. Cell Biol. 191, 677–691 10.1083/jcb.201006052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Burgener, S.S., Leborgne, N.G.F., Snipas, S.J., Salvesen, G.S., Bird, P.I. and Benarafa, C. (2019) Cathepsin G inhibition by Serpinb1 and Serpinb6 prevents programmed necrosis in neutrophils and monocytes and reduces GSDMD-driven inflammation. Cell Rep. 27, 3646–3656.e5 10.1016/j.celrep.2019.05.065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Kambara, H., Liu, F., Zhang, X., Liu, P., Bajrami, B., Teng, Y.et al. (2018) Gasdermin D exerts anti-inflammatory effects by promoting neutrophil death. Cell Rep. 22, 2924–2936 10.1016/j.celrep.2018.02.067 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Chen, K.W., Monteleone, M., Boucher, D., Sollberger, G., Ramnath, D., Condon, N.D.et al. (2018) Noncanonical inflammasome signaling elicits gasdermin D-dependent neutrophil extracellular traps. Sci. Immunol. 3, eaar6676. 10.1126/sciimmunol.aar6676 [DOI] [PubMed] [Google Scholar]
- 198.Sollberger, G., Choidas, A., Burn, G.L., Habenberger, P., Di Lucrezia, R., Kordes, S.et al. (2018) Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci. Immunol. 3, eaar6689. 10.1126/sciimmunol.aar6689 [DOI] [PubMed] [Google Scholar]
- 199.Kasperkiewicz, P., Hempel, A., Janiszewski, T., Kołt, S., Snipas, S.J., Drag, M.et al. (2020) NETosis occurs independently of neutrophil serine proteases. J. Biol. Chem. 295, 17624–17631 10.1074/jbc.RA120.015682 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Desai, J., Mulay, S.R., Nakazawa, D. and Anders, H.J. (2016) Matters of life and death. How neutrophils die or survive along NET release and is ‘NETosis’ = necroptosis? Cell. Mol. Life Sci. 73, 2211–2219 10.1007/s00018-016-2195-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Yousefi, S., Stojkov, D., Germic, N., Simon, D., Wang, X., Benarafa, C.et al. (2019) Untangling “NETosis” from NETs. Eur. J. Immunol. 49, 221–227 10.1002/eji.201747053 [DOI] [PubMed] [Google Scholar]
- 202.Yipp, B.G. and Kubes, P. (2013) NETosis: how vital is it? Blood 122, 2784–2794 10.1182/blood-2013-04-457671 [DOI] [PubMed] [Google Scholar]
- 203.Kaplan, M.J. and Radic, M. (2012) Neutrophil extracellular traps (NETs): double-edged swords of innate immunity. J. Immunol. 189, 2689–2695 10.4049/jimmunol.1201719 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Próchnicki, T., Mangan, M.S. and Latz, E. (2016) Recent insights into the molecular mechanisms of the NLRP3 inflammasome activation. F1000Research 5, F1000 Faculty Rev-1469 10.12688/f1000research.8614.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Mitroulis, I., Kambas, K., Chrysanthopoulou, A., Skendros, P., Apostolidou, E., Kourtzelis, I.et al. (2011) Neutrophil extracellular trap formation is associated with IL-1β and autophagy-related signaling in gout. PLoS One 6, e29318 10.1371/journal.pone.0029318 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Desai, J., Kumar, S.V., Mulay, S.R., Konrad, L., Romoli, S., Schauer, C.et al. (2016) PMA and crystal-induced neutrophil extracellular trap formation involves RIPK1-RIPK3-MLKL signaling. Eur. J. Immunol. 46, 223–229 10.1002/eji.201545605 [DOI] [PubMed] [Google Scholar]
- 207.Wang, X., Yousefi, S. and Simon, H.U. (2018) Necroptosis and neutrophil-associated disorders. Cell Death Dis. 9, 111 10.1038/s41419-017-0058-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Amini, P., Stojkov, D., Wang, X., Wicki, S., Kaufmann, T., Wong, W.W.et al. (2016) NET formation can occur independently of RIPK3 and MLKL signaling. Eur. J. Immunol. 46, 178–184 10.1002/eji.201545615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Dixon, S.J., Lemberg, K.M., Lamprecht, M.R., Skouta, R., Zaitsev, E.M., Gleason, C.E.et al. (2012) Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 10.1016/j.cell.2012.03.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Cao, J.Y. and Dixon, S.J. (2016) Mechanisms of ferroptosis. Cell. Mol. Life Sci. 73, 2195–2209 10.1007/s00018-016-2194-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Stockwell, B.R., Friedmann Angeli, J.P., Bayir, H., Bush, A.I., Conrad, M., Dixon, S.J.et al. (2017) Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 10.1016/j.cell.2017.09.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Yang, W.S., SriRamaratnam, R., Welsch, M.E., Shimada, K., Skouta, R., Viswanathan, V.S.et al. (2014) Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 10.1016/j.cell.2013.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Matsushita, M., Freigang, S., Schneider, C., Conrad, M., Bornkamm, G.W. and Kopf, M. (2015) T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J. Exp. Med. 212, 555–568 10.1084/jem.20140857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Angeli JP, F., Schneider, M., Proneth, B., Tyurina, Y.Y., Tyurin, V.A., Hammond, V.J.et al. (2014) Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol. 16, 1180–1191 10.1038/ncb3064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Xie, Y., Hou, W., Song, X., Yu, Y., Huang, J., Sun, X.et al. (2016) Ferroptosis: process and function. Cell Death Differ. 23, 369–379 10.1038/cdd.2015.158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Dixon, S.J., Patel, D.N., Welsch, M., Skouta, R., Lee, E.D., Hayano, M.et al. (2014) Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife 3, e02523 10.7554/eLife.02523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Yang, W.S. and Stockwell, B.R. (2016) Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 26, 165–176 10.1016/j.tcb.2015.10.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Yu, H., Guo, P., Xie, X., Wang, Y. and Chen, G. (2017) Ferroptosis, a new form of cell death, and its relationships with tumourous diseases. J. Cell. Mol. Med. 21, 648–657 10.1111/jcmm.13008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Tang, D., Chen, X., Kang, R. and Kroemer, G. (2021) Ferroptosis: molecular mechanisms and health implications. Cell Res. 31, 107–125 10.1038/s41422-020-00441-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Dai, E., Meng, L., Kang, R., Wang, X. and Tang, D. (2020) ESCRT-III-dependent membrane repair blocks ferroptosis. Biochem. Biophys. Res. Commun. 522, 415–421 10.1016/j.bbrc.2019.11.110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Wen, Q., Liu, J., Kang, R., Zhou, B. and Tang, D. (2019) The release and activity of HMGB1 in ferroptosis. Biochem. Biophys. Res. Commun. 510, 278–283 10.1016/j.bbrc.2019.01.090 [DOI] [PubMed] [Google Scholar]
- 222.Nishizawa, H., Matsumoto, M., Chen, G., Ishii, Y., Tada, K., Onodera, M.et al. (2021) Lipid peroxidation and the subsequent cell death transmitting from ferroptotic cells to neighboring cells. Cell Death Dis. 12, 332 10.1038/s41419-021-03613-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Yan, H.F., Zou, T., Tuo, Q.Z., Xu, S., Li, H., Belaidi, A.A.et al. (2021) Ferroptosis: mechanisms and links with diseases. Signal Transduct. Target Ther. 6, 49 10.1038/s41392-020-00428-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Hu, C.L., Nydes, M., Shanley, K.L., Pantoja, I.E.M., Howard, T.A. and Bizzozero, O.A. (2019) Reduced expression of the ferroptosis inhibitor GPx4 in multiple sclerosis and experimental autoimmune encephalomyelitis. J. Neurochem. 148, 426–439 10.1111/jnc.14604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Li, P., Jiang, M., Li, K., Li, H., Zhou, Y., Xiao, X.et al. (2021) Glutathione peroxidase 4-regulated neutrophil ferroptosis induces systemic autoimmunity. Nat. Immunol. 22, 1107–1117 10.1038/s41590-021-00993-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Shou, Y., Yang, L., Yang, Y. and Xu, J. (2021) Inhibition of keratinocyte ferroptosis suppresses psoriatic inflammation. Cell Death Dis. 12, 1009 10.1038/s41419-021-04284-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Vats, K., Kruglov, O., Mizes, A., Samovich, S.N., Amoscato, A.A., Tyurin, V.A.et al. (2021) Keratinocyte death by ferroptosis initiates skin inflammation after UVB exposure. Redox Biol. 47, 102143 10.1016/j.redox.2021.102143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Arbiser, J.L., Bonner, M.Y., Ward, N., Elsey, J. and Rao, S. (2018) Selenium unmasks protective iron armor: a possible defense against cutaneous inflammation and cancer. Biochim. Biophys. Acta 1862, 2518–2527 10.1016/j.bbagen.2018.05.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Magna, M. and Pisetsky, D.S. (2014) The role of HMGB1 in the pathogenesis of inflammatory and autoimmune diseases. Mol. Med. 20, 138–146 10.2119/molmed.2013.00164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Yang, H., Wang, H., Chavan, S.S. and Andersson, U. (2015) High mobility group box protein 1 (HMGB1): the prototypical endogenous danger molecule. Mol. Med. 21, S6–S12 10.2119/molmed.2015.00087 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Straino, S., Di Carlo, A., Mangoni, A., De Mori, R., Guerra, L., Maurelli, R.et al. (2008) High-mobility group box 1 protein in human and murine skin: involvement in wound healing. J. Invest. Dermatol. 128, 1545–1553 10.1038/sj.jid.5701212 [DOI] [PubMed] [Google Scholar]
- 232.Ranzato, E., Patrone, M., Pedrazzi, M. and Burlando, B. (2010) Hmgb1 promotes wound healing of 3T3 mouse fibroblasts via rage-dependent ERK1/2 activation. Cell Biochem. Biophys. 57, 9–17 10.1007/s12013-010-9077-0 [DOI] [PubMed] [Google Scholar]
- 233.Wilgus, T.A. (2018) Alerting the body to tissue injury: the role of alarmins and DAMPs in cutaneous wound healing. Curr. Pathobiol. Rep. 6, 55–60 10.1007/s40139-018-0162-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Zhang, Q., O'Hearn, S., Kavalukas, S.L. and Barbul, A. (2012) Role of high mobility group box 1 (HMGB1) in wound healing. J. Surg. Res. 176, 343–347 10.1016/j.jss.2011.06.069 [DOI] [PubMed] [Google Scholar]
- 235.Palumbo, R., Sampaolesi, M., De Marchis, F., Tonlorenzi, R., Colombetti, S., Mondino, A.et al. (2004) Extracellular HMGB1, a signal of tissue damage, induces mesoangioblast migration and proliferation. J. Cell Biol. 164, 441–449 10.1083/jcb.200304135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Santoro, A., Majorana, A., Bardellini, E., Gentili, F., Festa, S., Sapelli, P.et al. (2004) Cytotoxic molecule expression and epithelial cell apoptosis in oral and cutaneous lichen planus. Am. J. Clin. Pathol. 121, 758–764 10.1309/GHY8AL2D45P2R234 [DOI] [PubMed] [Google Scholar]
- 237.Trautmann, A., Akdis, M., Kleemann, D., Altznauer, F., Simon, H.U., Graeve, T.et al. (2000) T cell-mediated Fas-induced keratinocyte apoptosis plays a key pathogenetic role in eczematous dermatitis. J. Clin. Invest. 106, 25–35 10.1172/JCI9199 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Hofmeister, C.C., Quinn, A., Cooke, K.R., Stiff, P., Nickoloff, B. and Ferrara, J.L. (2004) Graft-versus-host disease of the skin: life and death on the epidermal edge. Biol. Blood Marrow Transpl. 10, 366–372 10.1016/j.bbmt.2004.03.003 [DOI] [PubMed] [Google Scholar]
- 239.Liu, Y., Peng, L., Li, L., Liu, C., Hu, X., Xiao, S.et al. (2017) TWEAK/fn14 activation contributes to the pathogenesis of bullous pemphigoid. J. Invest. Dermatol. 137, 1512–1522 10.1016/j.jid.2017.03.019 [DOI] [PubMed] [Google Scholar]
- 240.Arya, A.K., Tripathi, R., Kumar, S. and Tripathi, K. (2014) Recent advances on the association of apoptosis in chronic non healing diabetic wound. World J. Diabetes 5, 756–762 10.4239/wjd.v5.i6.756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Ahn, C., Negus, D. and Huang, W. (2018) Pyoderma gangrenosum: a review of pathogenesis and treatment. Expert. Rev. Clin. Immunol. 14, 225–233 10.1080/1744666X.2018.1438269 [DOI] [PubMed] [Google Scholar]
- 242.Duan, X., Liu, X., Liu, N., Huang, Y., Jin, Z., Zhang, S.et al. (2020) Inhibition of keratinocyte necroptosis mediated by RIPK1/RIPK3/MLKL provides a protective effect against psoriatic inflammation. Cell Death Dis. 11, 134 10.1038/s41419-020-2328-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Lauffer, F., Jargosch, M., Krause, L., Garzorz-Stark, N., Franz, R., Roenneberg, S.et al. (2018) Type I immune response induces keratinocyte necroptosis and is associated with interface dermatitis. J. Invest. Dermatol. 138, 1785–1794 10.1016/j.jid.2018.02.034 [DOI] [PubMed] [Google Scholar]
- 244.Kim, S.K., Kim, W.J., Yoon, J.H., Ji, J.H., Morgan, M.J., Cho, H.et al. (2015) Upregulated RIP3 expression potentiates MLKL phosphorylation-mediated programmed necrosis in toxic epidermal necrolysis. J. Invest. Dermatol. 135, 2021–2030 10.1038/jid.2015.90 [DOI] [PubMed] [Google Scholar]
- 245.Feoktistova, M., Makarov, R., Leverkus, M., Yazdi, A.S. and Panayotova-Dimitrova, D. (2020) TNF is partially required for cell-death-triggered skin inflammation upon acute loss of cFLIP. Int. J. Mol. Sci. 21, E8859 10.3390/ijms21228859 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Ciążyńska, M., Olejniczak-Staruch, I., Sobolewska-Sztychny, D., Narbutt, J., Skibińska, M. and Lesiak, A. (2021) The role of NLRP1, NLRP3, and AIM2 inflammasomes in psoriasis: review. Int. J. Mol. Sci. 22, 5898 10.3390/ijms22115898 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Levandowski, C.B., Mailloux, C.M., Ferrara, T.M., Gowan, K., Ben, S., Jin, Y.et al. (2013) NLRP1 haplotypes associated with vitiligo and autoimmunity increase interleukin-1β processing via the NLRP1 inflammasome. Proc. Natl. Acad. Sci. U.S.A. 110, 2952–2956 10.1073/pnas.1222808110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Tang, L. and Zhou, F. (2020) Inflammasomes in common immune-related skin diseases. Front. Immunol. 11, 882 10.3389/fimmu.2020.00882 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Hoffman, H.M. and Broderick, L. (2016) The role of the inflammasome in patients with autoinflammatory diseases. J. Allergy Clin. Immunol. 138, 3–14 10.1016/j.jaci.2016.05.001 [DOI] [PubMed] [Google Scholar]
- 250.Campbell, L., Raheem, I., Malemud, C.J. and Askari, A.D. (2016) The relationship between NALP3 and autoinflammatory syndromes. Int. J. Mol. Sci. 17, 725. 10.3390/ijms17050725 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Kelly, G., Hughes, R., McGarry, T., van den Born, M., Adamzik, K., Fitzgerald, R.et al. (2015) Dysregulated cytokine expression in lesional and nonlesional skin in hidradenitis suppurativa. Br. J. Dermatol. 173, 1431–1439 10.1111/bjd.14075 [DOI] [PubMed] [Google Scholar]
- 252.Bao, K. and Reinhardt, R.L. (2015) The differential expression of IL-4 and IL-13 and its impact on type-2 immunity. Cytokine 75, 25–37 10.1016/j.cyto.2015.05.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Schnurr, K., Borchert, A. and Kuhn, H. (1999) Inverse regulation of lipid-peroxidizing and hydroperoxyl lipid-reducing enzymes by interleukins 4 and 13. FASEB J. 13, 143–154 10.1096/fasebj.13.1.143 [DOI] [PubMed] [Google Scholar]
- 254.Sivaranjani, N., Rao, S.V. and Rajeev, G. (2013) Role of reactive oxygen species and antioxidants in atopic dermatitis. J. Clin. Diagn. Res. 7, 2683–2685 10.7860/JCDR/2013/6635.3732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Choi, D.I., Park, J.H., Choi, J.Y., Piao, M., Suh, M.S., Lee, J.B.et al. (2021) Keratinocytes-derived reactive oxygen species play an active role to induce type 2 inflammation of the skin: a pathogenic role of reactive oxygen species at the early phase of atopic dermatitis. Ann. Dermatol. 33, 26–36 10.5021/ad.2021.33.1.26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Panchal, F.H., Ray, S., Munshi, R.P., Bhalerao, S.S. and Nayak, C.S. (2015) Alterations in lipid metabolism and antioxidant status in lichen planus. Indian J. Dermatol. 60, 439–444 10.4103/0019-5154.159624 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Upadhyay, R.B., Carnelio, S., Shenoy, R.P., Gyawali, P. and Mukherjee, M. (2010) Oxidative stress and antioxidant defense in oral lichen planus and oral lichenoid reaction. Scand. J. Clin. Lab. Invest. 70, 225–228 10.3109/00365511003602455 [DOI] [PubMed] [Google Scholar]
- 258.Vlková, B., Stanko, P., Minárik, G., Tóthová, L., Szemes, T., Baňasová, L.et al. (2012) Salivary markers of oxidative stress in patients with oral premalignant lesions. Arch. Oral Biol. 57, 1651–1656 10.1016/j.archoralbio.2012.09.003 [DOI] [PubMed] [Google Scholar]
- 259.Fahlen, A., Engstrand, L., Baker, B.S., Powles, A. and Fry, L. (2012) Comparison of bacterial microbiota in skin biopsies from normal and psoriatic skin. Arch. Dermatol. Res. 304, 15–22 10.1007/s00403-011-1189-x [DOI] [PubMed] [Google Scholar]
- 260.Kobayashi, T., Glatz, M., Horiuchi, K., Kawasaki, H., Akiyama, H., Kaplan, D.H.et al. (2015) Dysbiosis and Staphylococcus aureus colonization drives inflammation in atopic dermatitis. Immunity 42, 756–766 10.1016/j.immuni.2015.03.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Yamazaki, Y., Nakamura, Y. and Nunez, G. (2017) Role of the microbiota in skin immunity and atopic dermatitis. Allergol. Int. 66, 539–544 10.1016/j.alit.2017.08.004 [DOI] [PubMed] [Google Scholar]
- 262.Rendon, A. and Schäkel, K. (2019) Psoriasis pathogenesis and treatment. Int. J. Mol. Sci. 20, 1475 10.3390/ijms20061475 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Capon, F., Burden, A.D., Trembath, R.C. and Barker, J.N. (2012) Psoriasis and other complex trait dermatoses: from loci to functional pathways. J. Invest. Dermatol. 132, 915–922 10.1038/jid.2011.395 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Das, S., Stuart, P.E., Ding, J., Tejasvi, T., Li, Y., Tsoi, L.C.et al. (2015) Fine mapping of eight psoriasis susceptibility loci. Eur. J. Hum. Genet. 23, 844–853 10.1038/ejhg.2014.172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Harden, J.L., Krueger, J.G. and Bowcock, A.M. (2015) The immunogenetics of psoriasis: a comprehensive review. J. Autoimmun. 64, 66–73 10.1016/j.jaut.2015.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Tsoi, L.C., Spain, S.L., Knight, J., Ellinghaus, E., Stuart, P.E., Capon, F.et al. (2012) Identification of 15 new psoriasis susceptibility loci highlights the role of innate immunity. Nat. Genet. 44, 1341–1348 10.1038/ng.2467 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Raj, D., Brash, D.E. and Grossman, D. (2006) Keratinocyte apoptosis in epidermal development and disease. J. Invest. Dermatol. 126, 243–257 10.1038/sj.jid.5700008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Hu, S.C.S., Yu, H.S., Yen, F.L., Lin, C.L., Chen, G.S. and Lan, C.C.E. (2016) Neutrophil extracellular trap formation is increased in psoriasis and induces human β-defensin-2 production in epidermal keratinocytes. Sci. Rep. 6, 31119 10.1038/srep31119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Casciaro, M., Di Salvo, E. and Gangemi, S. (2021) HMGB-1 in psoriasis. Biomolecules 12, 60 10.3390/biom12010060 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.de Cid, R., Riveira-Munoz, E., Zeeuwen, P.L.J.M., Robarge, J., Liao, W., Dannhauser, E.N.et al. (2009) Deletion of the late cornified envelope (LCE) 3B and 3C genes as a susceptibility factor for psoriasis. Nat. Genet. 41, 211–215 10.1038/ng.313 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Niehues, H., Tsoi, L.C., van der Krieken, D.A., Jansen, P.A.M., Oortveld, M.A.W., Rodijk-Olthuis, D.et al. (2017) Psoriasis-associated late cornified envelope (LCE) proteins have antibacterial activity. J. Invest. Dermatol. 137, 2380–2388 10.1016/j.jid.2017.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Rønholt, K., Nielsen, A.L.L., Johansen, C., Vestergaard, C., Fauerbye, A., López-Vales, R.et al. (2020) IL-37 expression is downregulated in lesional psoriasis skin. ImmunoHorizons 4, 754–761 10.4049/immunohorizons.2000083 [DOI] [PubMed] [Google Scholar]
- 273.Teng, X., Hu, Z., Wei, X., Wang, Z., Guan, T., Liu, N.et al. (2014) IL-37 ameliorates the inflammatory process in psoriasis by suppressing proinflammatory cytokine production. J. Immunol. 192, 1815–1823 10.4049/jimmunol.1300047 [DOI] [PubMed] [Google Scholar]
- 274.Marrakchi, S., Guigue, P., Renshaw, B.R., Puel, A., Pei, X.Y., Fraitag, S.et al. (2011) Interleukin-36–receptor antagonist deficiency and generalized pustular psoriasis. N. Engl. J. Med. 365, 620–628 10.1056/NEJMoa1013068 [DOI] [PubMed] [Google Scholar]
- 275.Guttman-Yassky, E., Nograles, K.E. and Krueger, J.G. (2011) Contrasting pathogenesis of atopic dermatitis and psoriasis–part I: clinical and pathologic concepts. J. Allergy Clin. Immunol. 127, 1110–1118 10.1016/j.jaci.2011.01.053 [DOI] [PubMed] [Google Scholar]
- 276.Clausen, M.L., Agner, T., Lilje, B., Edslev, S.M., Johannesen, T.B. and Andersen, P.S. (2018) Association of disease severity with skin microbiome and filaggrin gene mutations in adult atopic dermatitis. JAMA Dermatol. 154, 293–300 10.1001/jamadermatol.2017.5440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Drislane, C. and Irvine, A.D. (2020) The role of filaggrin in atopic dermatitis and allergic disease. Ann. Allergy Asthma Immunol. 124, 36–43 10.1016/j.anai.2019.10.008 [DOI] [PubMed] [Google Scholar]
- 278.Weidinger, S., Beck, L.A., Bieber, T., Kabashima, K. and Irvine, A.D. (2018) Atopic dermatitis. Nat. Rev. Primers 4, 1 10.1038/s41572-018-0001-z [DOI] [PubMed] [Google Scholar]
- 279.Dežman, K., Korošec, P., Rupnik, H. and Rijavec, M. (2017) SPINK5 is associated with early-onset and CHI3L1 with late-onset atopic dermatitis. Int. J. Immunogenet. 44, 212–218 10.1111/iji.12327 [DOI] [PubMed] [Google Scholar]
- 280.Kato, A., Fukai, K., Oiso, N., Hosomi, N., Murakami, T. and Ishii, M. (2003) Association of SPINK5 gene polymorphisms with atopic dermatitis in the Japanese population. Br. J. Dermatol. 148, 665–669 10.1046/j.1365-2133.2003.05243.x [DOI] [PubMed] [Google Scholar]
- 281.Zhao, L.P., Di, Z., Zhang, L., Wang, L., Ma, L., Lv, Y.et al. (2012) Association of SPINK5 gene polymorphisms with atopic dermatitis in Northeast China. J. Eur. Acad. Dermatol. Venereol. 26, 572–577 10.1111/j.1468-3083.2011.04120.x [DOI] [PubMed] [Google Scholar]
- 282.Sharma, B.R., Karki, R. and Kanneganti, T.D. (2019) Role of AIM2 inflammasome in inflammatory diseases, cancer and infection. Eur. J. Immunol. 49, 1998–2011 10.1002/eji.201848070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Paternoster, L., Standl, M., Waage, J., Baurecht, H., Hotze, M., Strachan, D.P.et al. (2015) Multi-ancestry genome-wide association study of 21,000 cases and 95,000 controls identifies new risk loci for atopic dermatitis. Nat. Genet. 47, 1449–1456 10.1038/ng.3424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Salpietro, C., Rigoli, L., Miraglia Del Giudice, M., Cuppari, C., Di Bella, C., Salpietro, A.et al. (2011) TLR2 and TLR4 gene polymorphisms and atopic dermatitis in Italian children: a multicenter study. Int. J. Immunopathol. Pharmacol. 24, 33–40 10.1177/03946320110240S408 [DOI] [PubMed] [Google Scholar]
- 285.Belkaid, Y. and Tamoutounour, S. (2016) The influence of skin microorganisms on cutaneous immunity. Nat. Rev. Immunol. 16, 353–366 10.1038/nri.2016.48 [DOI] [PubMed] [Google Scholar]
- 286.Parihar, A., Sharma, S., Bhattacharya, S.N. and Singh, U.R. (2015) A clinicopathological study of cutaneous lichen planus. J. Dermatol. Dermatol. Surg. 19, 21–26 10.1016/j.jssdds.2013.12.003 [DOI] [Google Scholar]
- 287.Neppelberg, E., Johannessen, A.C. and Jonsson, R. (2001) Apoptosis in oral lichen planus. Eur. J. Oral Sci. 109, 361–364 10.1034/j.1600-0722.2001.00081.x [DOI] [PubMed] [Google Scholar]
- 288.Bascones-Ilundain, C., Gonzalez-Moles, M.A., Esparza-Gómez, G., Gil-Montoya, J.A. and Bascones-Martínez, A. (2006) Importance of apoptotic mechanisms in inflammatory infiltrate of oral lichen planus lesions. Anticancer Res. 26, 357–362 PMID: [PubMed] [Google Scholar]
- 289.Santoro, A., Majorana, A., Bardellini, E., Festa, S., Sapelli, P. and Facchetti, F. (2003) NF-κB expression in oral and cutaneous lichen planus. J. Pathol. 201, 466–472 10.1002/path.1423 [DOI] [PubMed] [Google Scholar]
- 290.Hanahan, D. and Weinberg, R.A. (2011) Hallmarks of cancer: the next generation. Cell 144, 646–674 10.1016/j.cell.2011.02.013 [DOI] [PubMed] [Google Scholar]
- 291.Qin, H., Jin, J., Fischer, H., Mildner, M., Gschwandtner, M., Mlitz, V.et al. (2017) The caspase-1 inhibitor CARD18 is specifically expressed during late differentiation of keratinocytes and its expression is lost in lichen planus. J. Dermatol. Sci. 87, 176–182 10.1016/j.jdermsci.2017.04.015 [DOI] [PubMed] [Google Scholar]
- 292.Domingues, R., Pietrobon, A.J., Carvalho, G.C., Pereira, N.Z., Pereira, N.V., Sotto, M.N.et al. (2019) Lichen planus: altered AIM2 and NLRP1 expression in skin lesions and defective activation in peripheral blood mononuclear cells. Clin. Exp. Dermatol. 44, e89–e95 10.1111/ced.13859 [DOI] [PubMed] [Google Scholar]
- 293.Leung, D.Y. and Guttman-Yassky, E. (2014) Deciphering the complexities of atopic dermatitis: shifting paradigms in treatment approaches. J. Allergy Clin. Immunol. 134, 769–779 10.1016/j.jaci.2014.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Rapalli, V.K., Singhvi, G., Dubey, S.K., Gupta, G., Chellappan, D.K. and Dua, K. (2018) Emerging landscape in psoriasis management: from topical application to targeting biomolecules. Biomed. Pharmacother. 106, 707–713 10.1016/j.biopha.2018.06.136 [DOI] [PubMed] [Google Scholar]
- 295.Maglie, R., Di Cesare, A., Lazzeri, L., Pescitelli, L., Ricceri, F., Vannucchi, M.et al. (2018) Lichen planus triggered by CT-P13 and recurrence during secukinumab treatment. Br. J. Dermatol. 178, 303–304 10.1111/bjd.16003 [DOI] [PubMed] [Google Scholar]
- 296.Nakamura, M., Lee, K., Singh, R., Zhu, T.H., Farahnik, B., Abrouk, M.et al. (2017) Eczema as an adverse effect of anti-TNFalpha therapy in psoriasis and other Th1-mediated diseases: a review. J. Dermatol. Treat. 28, 237–241 10.1080/09546634.2016.1230173 [DOI] [PubMed] [Google Scholar]
- 297.Pugliese, D., Guidi, L., Ferraro, P.M., Marzo, M., Felice, C., Celleno, L.et al. (2015) Paradoxical psoriasis in a large cohort of patients with inflammatory bowel disease receiving treatment with anti-TNF alpha: 5-year follow-up study. Aliment. Pharmacol. Ther. 42, 880–888 10.1111/apt.13352 [DOI] [PubMed] [Google Scholar]
- 298.Rallis, E., Korfitis, C., Stavropoulou, E. and Papaconstantis, M. (2010) Onset of palmoplantar pustular psoriasis while on Adalimumab for psoriatic arthritis: a ‘class effect’ of TNF-alpha antagonists or simply an anti-psoriatic treatment adverse reaction? J. Dermatol. Treat. 21, 3–5 10.3109/09546630902882089 [DOI] [PubMed] [Google Scholar]
- 299.Thompson, J.M., Cohen, L.M., Yang, C.S. and Kroumpouzos, G. (2016) Severe, ulcerative, lichenoid mucositis associated with secukinumab. JAAD Case Rep. 2, 384–386 10.1016/j.jdcr.2016.07.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Vasconcellos, J.B., Pereira, D.D., Vargas, T.J., Levy, R.A., Pinheiro, G.D. and Cursi, I.B. (2016) Paradoxical psoriasis after the use of anti-TNF in a patient with rheumatoid arthritis. An. Bras. Dermatol. 91, 137–139 10.1590/abd1806-4841.20164456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Yamauchi, P.S., Bissonnette, R., Teixeira, H.D. and Valdecantos, W.C. (2016) Systematic review of efficacy of anti-tumor necrosis factor (TNF) therapy in patients with psoriasis previously treated with a different anti-TNF agent. J. Am. Acad. Dermatol. 75, 612–618.e6 10.1016/j.jaad.2016.02.1221 [DOI] [PubMed] [Google Scholar]
- 302.Radtke, M.A., Reich, K., Spehr, C. and Augustin, M. (2015) Treatment goals in psoriasis routine care. Arch. Dermatol. Res. 307, 445–449 10.1007/s00403-014-1534-y [DOI] [PubMed] [Google Scholar]
- 303.Degterev, A., Ofengeim, D. and Yuan, J. (2019) Targeting RIPK1 for the treatment of human diseases. Proc. Natl Acad. Sci. U.S.A. 116, 9714–9722 10.1073/pnas.1901179116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Harris, P.A., Berger, S.B., Jeong, J.U., Nagilla, R., Bandyopadhyay, D., Campobasso, N.et al. (2017) Discovery of a first-in-class receptor interacting protein 1 (RIP1) kinase specific clinical candidate (GSK2982772) for the treatment of inflammatory diseases. J. Med. Chem. 60, 1247–1261 10.1021/acs.jmedchem.6b01751 [DOI] [PubMed] [Google Scholar]
- 305.Weisel, K., Berger, S., Papp, K., Maari, C., Krueger, J.G., Scott, N.et al. (2020) Response to inhibition of receptor-interacting protein kinase 1 (RIPK1) in active plaque psoriasis: a randomized placebo-controlled study. Clin. Pharmacol. Ther. 108, 808–816 10.1002/cpt.1852 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Weisel, K., Scott, N.E., Tompson, D.J., Votta, B.J., Madhavan, S., Povey, K.et al. (2017) Randomized clinical study of safety, pharmacokinetics, and pharmacodynamics of RIPK1 inhibitor GSK2982772 in healthy volunteers. Pharmacol. Res. Perspect. 5, e00365 10.1002/prp2.365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Newton, K., Dugger, D.L., Wickliffe, K.E., Kapoor, N., de Almagro, M.C., Vucic, D.et al. (2014) Activity of protein kinase RIPK3 determines whether cells die by necroptosis or apoptosis. Science 343, 1357–1360 10.1126/science.1249361 [DOI] [PubMed] [Google Scholar]
- 308.Mandal, P., Berger, S.B., Pillay, S., Moriwaki, K., Huang, C., Guo, H.et al. (2014) RIP3 induces apoptosis independent of pro-necrotic kinase activity. Mol. Cell 56, 481–495 10.1016/j.molcel.2014.10.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309.Martens, S., Hofmans, S., Declercq, W., Augustyns, K. and Vandenabeele, P. (2020) Inhibitors targeting RIPK1/RIPK3: old and new drugs. Trends Pharmacol. Sci. 41, 209–224 10.1016/j.tips.2020.01.002 [DOI] [PubMed] [Google Scholar]
- 310.Poreba, M., Kasperkiewicz, P., Snipas, S.J., Fasci, D., Salvesen, G.S. and Drag, M. (2014) Unnatural amino acids increase sensitivity and provide for the design of highly selective caspase substrates. Cell Death Differ. 21, 1482–1492 10.1038/cdd.2014.64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Brumatti, G., Ma, C., Lalaoui, N., Nguyen, N.Y., Navarro, M., Tanzer, M.C.et al. (2016) The caspase-8 inhibitor emricasan combines with the SMAC mimetic birinapant to induce necroptosis and treat acute myeloid leukemia. Sci. Transl. Med. 8, 339ra69 10.1126/scitranslmed.aad3099 [DOI] [PubMed] [Google Scholar]
- 312.Rathkey, J.K., Zhao, J., Liu, Z., Chen, Y., Yang, J., Kondolf, H.C.et al. (2018) Chemical disruption of the pyroptotic pore-forming protein gasdermin D inhibits inflammatory cell death and sepsis. Sci. Immunol. 3, eaat2738 10.1126/sciimmunol.aat2738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313.Sun, L., Wang, H., Wang, Z., He, S., Chen, S., Liao, D.et al. (2012) Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 10.1016/j.cell.2011.11.031 [DOI] [PubMed] [Google Scholar]
- 314.Yan, B., Liu, L., Huang, S., Ren, Y., Wang, H., Yao, Z.et al. (2017) Discovery of a new class of highly potent necroptosis inhibitors targeting the mixed lineage kinase domain-like protein. Chem. Commun. Camb. 53, 3637–3640 10.1039/C7CC00667E [DOI] [PubMed] [Google Scholar]