Abstract
Skin is a tissue vulnerable to radiation exposure, which causes acute tissue reactions, including erythema, edema, desquamation, ulceration and late effects, such as skin cancers. As the effects of radiation exposure on the skin tissue are easily evaluated by visual examination, much information on radiation-induced skin reactions has been available from the clinical observation of people exposed to ionizing radiation, such as cancer patients receiving radiotherapy, although the mechanisms underlying skin reactions have not yet been fully understood. Recent advances in tissue biology at the molecular level have provided insights into the mechanisms of skin tissue reactions from the stem cell points of view. For example, our understanding of epidermal regeneration by epidermal stem cells as well as cells from the bulge in humans and the sebaceous gland in mouse, descriptions of the role of skin immune cells on inflammatory response and maintenance of genome integrity by epidermal stem cell competition, have greatly improved in the last decade with the identification of several key molecules. Thus, this review will provide an overview of the current status toward the comprehensive understanding of the mechanisms of adverse skin tissue reactions, with respect to mitigation of acute skin injuries as well as late carcinogenesis in response to ionizing radiation. In particular, the pleiotropic features of various types of cells consisting of skin tissue and their roles in securing skin functional homeostasis will be discussed.
Keywords: skin, tissue reaction, skin care, radiation
INTRODUCTION
As the largest organ of the body, the skin plays a crucial role as a physical barrier against pathogens, thermoregulation, water transpiration, immune surveillance and perception. The skin is also essential for ensuring smooth movement of the body and exerts endocrine and exocrine activities [1–4]. The skin is composed of three major layers, which are epidermis, dermis and subcutaneous tissue. Epidermis has four functional layers, including basal, spinous, granular and cornified layers. Dermis connects epidermis and subcutaneous tissue, and skin appendages, such as hair follicles, sebaceous glands and sweat glands, are embedded in the epidermis [3–8].
The skin, as the outermost organ, is exposed to various types of physical, chemical and biological stresses, so that there are sophisticated systems to maintain organizational homeostasis and integrity [2–4, 7, 8]. Skin tissue response so far has been extensively studied after exposure to ionizing radiation, which is one of such stressors. Radiation causes acute skin tissue reactions as well as late skin cancers [9]. As these are mostly an epidermal origin, previous studies have primarily focused on the responses of the epidermis. Since the discovery of X-rays, skin complications were the first medical symptom to be recorded. Therefore, historically, it has been described that the skin is one of the most sensitive organs to radiation [9, 10]. From a radiation protection point of view, the threshold doses were assigned. For example, the threshold dose for early transient erythema, which is due to vascular dilation, is estimated to be 2 Gy. A main erythema reaction, which is caused by the death of basal epithelial cells followed by inflammation, will be manifested with ~6 Gy or more. In contrast to such lower doses, higher dose exposure may cause serious outcomes, such as dry and moist desquamation and ulceration, although severer effects need longer time of onset. The matrix cells at the bottom of hair follicle are also sensitive to radiation exposure, whose death gives rise to epilation and alopecia [9, 11–16].
The late effect of radiation exposure is characterized by the induction of skin cancer, most of which is also of epidermal origin. Non-melanoma skin cancers are the predominant cancer type in human skin, and basal cell carcinoma (BCC) is more frequent than squamous cell carcinoma (SCC) in response to radiation exposure. A dose-dependent and age-at-exposure-dependent risk was described in detail among populations receiving ionizing radiation [17–19].
While previous information described above seems to be sufficient for understanding the effects of radiation on skin tissue, most of them were descriptive and obtained from a clinical point of view. Thus, the knowledge is still insufficient in terms of mechanistic insights in view of recent advances in the skin biology, which is essential for management and mitigation of the adverse effects of skin tissue reactions caused by radiation exposure. Therefore, the present review will provide an overview of the pleiotropic aspects of skin tissue reaction to ionizing radiation obtained recently, with an emphasis on the mechanistic perspective. In this review, unless otherwise stated, all descriptions relate to human skin.
STRUCTURE OF THE SKIN
Epidermis
The uppermost layer of the skin is the epidermis, which forms the waterproof barrier (Fig. 1). It consists of four layers, the stratum corneum (SC), stratum granulosum, stratum spinosum and the basal layer, in which the bottom basal layer contains clonogenic cells. The epidermis is largely composed of keratinocytes, together with melanocytes, Langerhans cells and other immune cells. The renewal of the epidermis is dependent upon growth of the keratinocytes at the basal layer, and the turnover of epidermis takes ~39–56 days in human [20–23]. The turnover of the skin has been demonstrated to decrease as skin ages. In studies, it was reported that 30–50% decrease between third and eighth decades [24–29]. Accordingly, the increase of transepidermal water loss was observed age-dependently [24].
Fig. 1.
Structure of the epidermis. The epidermis consists of four layers. The bottommost layer named basal cell layer consists of keratinocytes with epidermal stem cells. The differentiating cells leave the basal layer, and migrate to the epidermal surface, through stratum spinosum, stratum granulosum, and finally, they are shed periodically from SC. The epidermis also contains other types of cells, including melanocytes and Langerhans cells.
Epidermal stem cells are located in the basal layer, and it has been reported that about 7% of cells in the basal layer could be stem and its progenitor cells. In another study, using stem- and progenitor-specific membrane markers demonstrated that less than 1% and 10% of total cell population are stem and progenitor cells, respectively [30, 31]. More recently, a technique defining long-term repopulating cells was introduced [32], and the frequency of stem cells was estimated to be 0.003% of total epidermal cells, which corresponded to 0.01% of basal cells [32].
While the concept named the epidermal proliferating unit (EPU) was proposed by Potten over 50 years ago [33, 34], the stem cell population in the interfollicular epidermis and its hierarchy within the EPU have not been sufficiently determined yet. Recent studies using lineage-tracing technique and single cell transcriptomics have suggested that stochastic asymmetric division of stem cells within the EPU serves a progenitor cell, and they are doomed to give rise to cornified layers with multiple cell divisions [35–37]. Other studies pointed out that stem cell-derived progenitors from multiple units likely contribute to form EPUs [38]. Furthermore, a single-cell RNA sequencing has uncovered heterogeneity of interfollicular stem cells and demonstrated that four spatially distinct stem cells were identified [39]. Thus, further studies are needed to draw an entire view of the stem cells in epidermis.
Hair follicle and other epidermal appendages
Hair follicles play indispensable roles in the function and regeneration of the skin. The outermost region of the hair follicle is called the connective tissue sheath including inner root sheath and outer root sheath, whose components are provided by the dermis. The lower half of hair follicle contains dermal papilla at the bottom, surrounded by the hair matrix and melanocytes (Fig. 2). The dermal papilla is rich in capillaries and has stem cells. The hair matrix cells form medulla of the shaft and inner root sheath with the aid of dermal papilla. The outer root sheath is provided by epidermis. The upper half contains the bulge, sebaceous gland and arrector pili muscle [40–42]. The hair cycle is divided into three stages, such as anagen, catagen and telogen. The anagen stage is the growing stage of hair cycle, which lasts for 2–8 years, followed by the catagen phase of ~2 weeks. During catagen, matrix cells terminate their growth, and lower follicles regress to move condensed dermal papilla upward toward the bulge region. When regression completes, the follicle enters to telogen, which is the resting phase of hair cycle that lasts for 2–3 months [41–43].
Fig. 2.
Structure of the skin appendages. The skin appendages include hairs, sweat glands and sebaceous glands. The hair structure divides into the hair shaft and hair follicle, the latter contains the hair bulb. The bottom part of the hair follicle is called the dermal papilla, which is an invagination of vascularized connective tissues. Around the papilla is the hair matrix, containing melanocytes. The upper part of hair bulb continues to a root sheath composed of an outer and inner root sheath, and the inner root sheath forms cuticle that constitutes the outermost layer of the hair shaft. Cutaneous microvasculature is organized in the dermis, and there are two layers of horizontal plexuses.
In addition to hair follicles, the epidermal appendages include sebaceous glands and sweat glands, in which the latter has two different types called eccrine and apocrine glands [44, 45] (Fig. 2).
Dermis
The dermis is located beneath the epidermis, and is composed of fibrous proteins, including collagen and elastic fibers, which supports the epidermis. The dermis also plays essential roles in thermoregulation, water transpiration, immune surveillance and perception, as the vasculature, nerves and glands are located within the dermis [46, 47]. The primary cells in the dermis are fibroblasts, and several other types of cells, like immune cells, also exist. The dermis is also a space where cutaneous microvasculature is organized. There are two layers of horizontal plexuses, in which one is beneath the epidermis and the other is above the dermal-subcutaneous junction [48–50].
Subcutaneous tissue
The subcutaneous tissue is the lowermost layer of the skin, which plays a major storage site of fat. The subcutaneous tissue also contains fibroblasts and macrophages, as well as large blood and lymphatic vessels [51–54]. Beneath the subcutaneous layer is the ‘panniculus carnosus’, which is a striated muscular layer with fascia [55–57]. In mouse back skin, the panniculus carnosus is accompanied by the perifascial areolar tissue, which is rich in macrophages and fibroblasts [58].
FUNCTION OF THE SKIN
Barrier
The defensive function is the indispensable role of skin tissue, which serves as the physical barrier against pathogens, limits passive water transpiration and reduces absorption of environmental toxic materials. The outermost physical barrier is secured by SC, which is formed by terminal differentiation of keratinocytes named keratinization. The second biological barrier is ensured by cell-to-cell connections through tight junctions (TJ), adherence junctions (AJ), gap junctions (GJ) and desmosomes. TJ, which constitute the second barrier after SC, function as a water flux regulator. It also plays a crucial role in the epithelial cell polarization. AJ and desmosome are involved in strong cell-to-cell adhesion, while GJ forms a cell-to-cell communication mediating the exchange of ions and second messengers through connexin channels [5–8, 59].
In addition to the physical barrier, chemical barrier, which serves acidic pH condition, and biological barrier through the release of antimicrobial peptides (AMPs), such as defensins, cathelicidins and dermcidin exist [60–64]. The maintenance of acidic pH is managed by several mechanisms, including weakly acidic natural-moisturizing factors derived from epidermal filaggrin, and secreted factors from the sebaceous gland, such as triglycerides, fatty acids, wax esters and squalene [65–67]. For example, triglycerides are hydrolyzed to free fatty acids by epidermal microorganisms, such as bacteria and yeasts, contributing the acidification of the skin [60–64].
Immunity
Cutaneous immune surveillance is an active defence mechanism, by which the host is securely protected from the insults of microbial pathogens. Immune response is highly sophisticated mechanism orchestrated by the interaction of innate and adaptive immune systems [68]. Invasion of microorganisms is attentively watched by the sentinel cells resident in the epidermis and dermis, such as keratinocytes, Langerhans cells (LCs), dendritic cells (DCs), macrophages and mast cells [68–70] (Table 1). Keratinocytes play the primary roles in sensing the pathogens and mediating immune responses. They present receptors recognizing pathogen-associated molecular patterns, among which the Toll-like receptors (TLRs) are the best studied [69]. Keratinocytes express several TLRs, and upon activation of TLRs, they produce AMPs as well as a variety of cytokines, including tumor necrosis factor (TNF), and the IL-33 and IL-1 families, which stimulate innate immune system. AMPs production from keratinocytes is potentiated by IL-17 and IL-22 secreted from TH17 cells, which is activated by IL-1. IL-1 also stimulates DCs and macrophages to execute local inflammation. Keratinocytes also secrete several chemokines, such as CCL20, CXCL9, CXCL10 and CXCL11, thereby activating skin-resident immune cells and recruiting immune cells to the skin [71].
Table 1.
Immune cells in the skin and their radiation sensitivity
| Human | Mouse | Radiation sensitivity | |
|---|---|---|---|
| Epidermis | Langerhans | Langerhans | Resistant |
| CD8+ TRM | CD8+ TRM | Slightly resistant | |
| DETC | Sensitive | ||
| Dermis | cDC1 | cDC1 | Resistant |
| cDC2 | cDC2 | Resistant | |
| Macrophage | Macrophage | Resistant | |
| Treg | Treg | Slightly resistant | |
| CD4+ TRM | CD4+ TRM | Slightly resistant | |
| γδT cells | γδT cells | Sensitive/resistant | |
| ILC2 | ILC2 | Sensitive | |
| NKT | NKT | Slightly resistant |
For more information, see Ref. [16].
Once epidermal LCs and dermal DCs are activated as professional antigen presenting cells (APCs), they present antigens and promote maturation of naive T cells [72–77]. Together with CD8+ T cells in the epidermis, functionally activated T cells constitute one branch of adaptive immune systems [78]. In the dermis, B cells also constitute another branch of adaptive immune systems. Like T cells, B cells are migrated to the site of inflammation, which is mediated by selectins, chemokines receptors and integrins [79, 80].
Accumulating evidences have demonstrated that other types of immune cells exist in the skin [81, 82]. Those include γδT (dendritic epidermal T cells: DETCs in mice) cells and CD8+ resident memory cells in the epidermis, and CD4+ and CD8+ T cells, NK cells, NKT cells and innate lymphoid cells in the dermis [83–93] (Table 1). Several reports have indicated that they also play a role in orchestrating the skin immunity, however, further studies will be needed to understand complex and sophisticated crosstalk between these cells.
MECHANISMS OF SKIN TISSUE REACTION TO RADIATION
Radiation exposure induces various types of DNA damage, among which DNA double-strand breaks (DSBs) are the most harmful [10]. Mammalian cells have multiple sophisticated DSB repair systems, such as non-homologous end-joining and homologous recombination, however, as the dose increases the fraction of DSBs that are unable to repair increase. Such irreparable DSBs trigger cell death, giving rise to tissue reaction [9–11].
So far, it has been described that radiation exposure executes several cell death pathways including apoptosis, necrosis, autophagy and premature senescence. Manifestation of cell death modes is dependent on tissue types. For example, hematopoietic tissues such as bone marrow, thymus and spleen prefer to induce apoptosis, while many epithelial tissues, such as lung, liver, thyroid gland and mammary gland, primarily exhibit premature senescence. Cells comprising connective tissues, such as fibroblasts, endothelial cells and macrophages also induce premature senescence. If the microvasculature is disabled by radiation exposure, necrosis is finally triggered [94].
In the skin, epidermal keratinocytes mostly induce premature senescence in response to ionizing radiation, while they induce apoptosis after ultraviolet light irradiation [95–97]. Apoptosis could be the major mode of cell death in hair follicles [15, 98, 99], sebaceous glands and sweat glands, although the final conclusion still awaits further confirmation [100–104]. Whereas keratinocytes and epithelial cells of sebaceous and sweat glands are radiation sensitive, dermal fibroblasts are more resistant. While T cells in epidermis are radiation sensitive, LC in the epidermis, DC and macrophages in the dermis are resistant [16] (Table 1).
Acute radiation syndrome
The skin is one of the most responsive tissues to radiation exposure. In general, as the dose increases, the number of dead cells also increases, giving rise to severe symptoms (Table 2). In other words, even if cell death is induced, functional failure may not be visible, if the amount of dead cells is marginal. Thus, even if the skin tissue is exposed to radiation, it may not always manifest tissue reaction. This is the fundamental mechanism defining the threshold dose. Therefore, over the threshold dose, several adverse reactions are induced. For example, several waves of erythema are well documented [9, 10]. Epilation, dry and moist desquamation and ulceration become detectable depending on the dose [9, 10] (Table 2).
Table 2.
Approximate threshold single dose for the reaction of human skin tissue
| Effect | Approximate threshold dose (Gy) | Time of onset |
|---|---|---|
| Early transient erythema | 2 | 2–24 h |
| Main erythema reaction | 6 | ~1.5 weeks |
| Temporary epilation | 3 | ~3 weeks |
| Permanent epilation | 7 | ~3 weeks |
| Dry desquamation | 14 | ~4–6 weeks |
| Moist desquamation | 18 | ~4 weeks |
| Secondary ulceration | 24 | >6 weeks |
| Late erythema | 15 | 8–10 weeks |
| Ischaemic dermal necrosis | 18 | >10 weeks |
| Dermal atrophy (first phase) | 10 | >52 weeks |
| Telangiectasia | 10 | >52 weeks |
| Dermal necrosis (late phase) | >15? | >52 weeks |
From ICRP publication 18, 2012 (Ref. [10]).
Since acute radiation dermatitis is one critical adverse effect of radiotherapy, particularly in the patients with the radiotherapy for breast, and head and neck cancers, scoring systems for grading acute radiation dermatitis caused by radiotherapy have been established by several organizations (Table 3), which are useful for preventing and management of acute radiation dermatitis.
Table 3.
Scoring criteria used for grading acute radiation dermatitis
| Grade/Score 1 | Grade/Score 2 | Grade/Score 3 | Grade/Score 4 | |||
|---|---|---|---|---|---|---|
| CTCAE (5.0) | Faint erythema or dry desquamation | Moderate to brisk erythema patchy moist desquamation mostly confined to skin folds and creases; moderate edema | Moist desquamation in areas other than skin folds and creases; bleeding induced by minor trauma or abrasion | Life-threatening consequences; skin necrosis or ulceration of full thickness dermis; spontaneous bleeding from involved site; skin graft indicated | ||
| RTOG | Erythema, epilation dry desquamation | Bright erythema, edema moist desquamation, | Confluent moist desquamation, pitting edema | Ulceration, hemorrhage necrosis | ||
| Score 1.0 | Score 1.5 | Score 2.0 | Score 2.5 | Score 3.0 | Score 3.5 | |
| ONS | Faint or dull erythema | Bright erythema | Dry desquamation with or without erythema | Small to moderate amount of moist desquamation | Confluent moist desquamation | Ulceration, hemorrhage or necrosis |
CTCAE = Common terminology criteria for adverse events, RTOG = Radiation Therapy Oncology Group, ONS = Oncology Nursing Society.
Ryan et al. [105].
The uniqueness of the skin tissue reaction to radiation is that the manifestation of radiation effect shows an area-size dependency. For example, small area of skin is more resistant to higher doses than large area, indicating efficient repair of epidermal injuries, which is confirmed by the evidence that epidermis at the edge of an irradiated area is highly proliferative and mobile [9, 16, 106–108].
Another uniqueness is radiation quality dependence. As radiation dermatitis is well documented in relation to radiotherapy, X-rays or γ-rays, which can penetrate deep into the skin tissue, are the subject of consideration, and therefore, radiation dermatitis is commonly observed. In contrast, α-rays are unable to penetrate even the outermost keratin layer of skin, so that the skin injury, called alpha burns, is only limited to mild reddening of the outmost layers [109, 110]. β-rays are able to penetrate into the skin layer, although their penetrability is limited to ~1 cm, and so-called beta burns, which are superficial injuries like sunburn, are caused [109, 110]. Accordingly, different threshold dose for the acute effects from β-rays was documented [111]. For example, the threshold dose for early erythema was ~6 Gy for β-rays, which is much higher than that reported after radiotherapy (Table 2).
Multiple mechanisms are involved in the manifestation of radiation-induced dermatitis [16]. Upon irradiation, keratinocytes halt cell cycle progression, and if DNA damage is irreparable, permanent cell cycle arrest gives rise to the induction of premature senescence [112]. Senesced keratinocytes secrete soluble factors that regulate inflammatory response as described below. They also result in dysregulation of proteins involved in TJ formation [113], thereby, eradicates permeability barrier. These are the mechanisms underlying the manifestation of erythema, desquamation and ulcer. Epilation is another deleterious outcome caused by radiation exposure. Cells consisting hair follicles, including epithelial cells and melanocytes are sensitive to radiation, which die by apoptosis [100, 114]. Proliferating hair matrix keratinocytes are the most sensitive, while quiescent hair follicle stem cells in bulge and melanocyte stem cells are rather resistant [115]. Cells in dermal papilla seem to be the most resistant [100]. Since matrix keratinocytes are essential for regulating hair cycle, their death is directly related to the manifestation of alopecia [104, 115]. Also, the crosstalk between matrix cells and melanocytes in hair bulb is critical for hair pigmentation, so matrix cell death causes gray hair as well [116, 117].
In order to mitigate the manifestation of acute radiation syndrome, single-cell RNA sequencing as well as multi-omics analysis were subjected to identify molecular mechanisms contributing to radiation dermatitis [97, 118, 119]. For example, a study using C57BL/6 mice exposed to 20 Gy of X-rays demonstrated that cell cluster-dependent changes in gene expression in response to radiation exposure. The gene-set enrichment analysis revealed that the BMP signaling pathway, which could activate the WNT and SMAD pathways, was enriched in proliferating keratinocytes. Papillary fibroblasts and lymphatic endothelial cells activated pathways attracting immune cells and migration of leukocytes [118]. Another study described alterations in fatty acid metabolism involved in radiation-induced skin damage [97]. Although the datasets are insufficient at present, integrated analyses in the future are expected to extract critical pathway(s) regulating radiation dermatitis.
Inflammatory response
Excess radiation damage to keratinocytes in the epidermal basal layer triggers premature senescence induction, which gives rise to the activation of inflammatory response. Senescent cells have been proven to show secretory phenotype, known as senescence-associated secretory phenotype (SASP) [120–125], and secrete soluble factors including IL-1β, IL-6, IL-8, CCL20, CCL5 and TNF, all of which attract immune cells, stimulating their invasion to the damaged sites as well as their activity (Table 4). Senesced fibroblasts also coordinate with this process by secreting IL-6 and TNF. Cytokines and chemokines secreted by damaged keratinocytes and activated DCs in the dermis promote differentiation of naive T cells to helper T cells, such as TH17 cells. Then, TH17 cells secrete IL-17 and IL-22 to stimulate keratinocyte growth [126–131]. Fibroblasts in the dermis, which are exposed to TNF, IL-1β and TGF-β from senesced keratinocytes, result in release of KGF, EGF and TGF-β, and this crosstalk stimulates proliferation of keratinocytes, contributing to the recovery of the epidermis (Table 4) [108, 132, 133].
Table 4.
Soluble factors secreted by various types of cells in epidermis of the skin
| Cell source | Factors | Functions |
|---|---|---|
| Keratinocytes | IL-1α/β | Initiate rapid immune response. Stimulate IL-6, IL-8 and TNF-α expression |
| IL-6 | Promote keratinocyte proliferation | |
| IL-7 | Stimulate T cell growth | |
| IL-8 | Attracts neutrophils | |
| IL-15 | Stimulate T cell growth | |
| IL-18 | Potentiates inflammation | |
| CCL5 | Stimulate LC immigration | |
| CCL20/MIP-3α | Attracts immune cells. Stimulates LC immigration | |
| TGF-β | Controls keratinocyte proliferation and differentiation | |
| TNF-α | Regulate various effects, including proinflammatory cytokines, and chemokines | |
| TSLP | Promote Th2 cell differentiation | |
| Langerhans cells | IL-6 | Promote keratinocyte proliferation |
| IL-15 | Stimulates Th17 and Th22 to secrete IL-17 and IL-22 | |
| IL-23 | Stimulates Th17 and Th22 to secrete IL-17 and IL-22 | |
| CXCL9 | Recruitment of T cells | |
| CXCL10 | Recruitment of T cells | |
| CCL20/MIP-3α | Attracts immune cells | |
| DETC | IL-2 | Facilitate pre-DETC expansion and survival |
| IL-13 | Regulate homeostatic proliferation of keratinocyte | |
| IL-15 | Stimulates Th17 and Th22 to secrete IL-17 and IL-22 | |
| IL-17A | Stimulate keratinocyte proliferation and potentiate anti-microviral barrier | |
| CCL3/MIP-1α | Promotes immune cell migration | |
| CCL4/MIP-1β | Promotes immune cell migration | |
| CCL5 | Attracts and promotes immune cell migration | |
| CCL20/MIP-3α | Attracts immune cells | |
| MCP-1 | Attracts immune cells | |
| XCL1 | Attracts immune cells | |
| KGF-1/2 | Promotes keratinocyte proliferation and maturation | |
| IGF-1 | Stimulate keratinocyte growth | |
| TNF-α | Regulate various effects, including proinflammatory cytokines and chemokines | |
| GM-CSF | Promotes differentiation and activation of LC |
Since senescent cells express NKG2D, which is a ligand recognized by NKG2D receptor on NK cells, they are hypothesized to induce apoptosis in senesced keratinocytes. This promotes the clearance of the dead cells, which facilitates the regeneration of the epidermis [92, 93, 134–138].
Apart from the cell death in keratinocytes, the inflammatory response is also activated by pathogen invasion caused by the breakdown of the epidermal barrier [69, 70]. Although γδT cells are disappeared soon after irradiation, LCs remain in the epidermis functionally as the sentinels for securing immune homeostasis [139]. In the dermis, DCs and macrophages are also resistant to radiation exposure and assist in tissue clearance and the presentation of pathological antigens if any [140, 141]. Epidermal γδT cells are more abundant in mice, which consist of more than 90% of T cells in the epidermis, and are named dendritic epidermal T cells [142].
Both LC and dendritic epidermal T cells (DETC), resident in the basal layer of the epidermis, elongate dendrites upwards at the TJs formed between differentiated keratinocytes. DETC secrete several soluble factors that promote keratinocytes survival and proliferation (Table 4), while keratinocytes secrete IL-15 that promote DETC survival and renewal. Once the epidermis is injured, keratinocytes present DETC-specific antigen, which activates DETC and promotes secretion of soluble factors, such as IL-2. IL-13, IGF-1 and KGFs, resulting in the modulation of the functions of neighboring cells (Table 4) [143, 144].
We have discovered that coordinated pattern of DETC is disrupted even 3 weeks after the completion of 40 Gy of X-rays (unpublished data), indicating that they are quite immobile. While both LCs and DETCs are the epidermis-resident APCs, their roles are somewhat different. LCs are the positive regulator of the immune response, whereas DETC seem to negatively regulate inflammation, suggesting that disappearance of DETC after radiation exposure stimulates an inflammatory response that facilitates epidermal regeneration.
Besides γδT cells, other T cell compartments in the epidermis include regulatory T cell (Treg) and tissue-resident memory CD8+ T cell (TRM). Treg is known to be radiation sensitive, and reduction of Treg is reported to cause unwanted enhancement of skin immune function. Upon recognition of cognate antigens, TRM is reactivated and differentiated into cytotoxic effector cells, which secrete Interferon-γ (IFN-γ), potentiating anti-viral activity. Recently, it has been shown that reactivation of TRM requires cross presentation of keratinocyte-expressed antigens on LCs. As such, not only LCs and DETCs, but also keratinocytes are members coordinating the immune response [85, 145, 146].
Regeneration of the skin
Regeneration of the epidermis is carried out by proliferation of epidermal stem cells, which reside in the basal layer of the epidermis and in the bulge region of the hair follicle. The epidermal stem cells provide progenitor cells, which divide symmetrically to supply cells to be differentiated. Recently, it was reported that skin homeostasis is regulated through cell competition between epidermal stem cells in the basal layer [147]. Differential expression of collagen XVII (COL17A1) in basal keratinocytes, which is a component of hemidesmosome, was involved in cell competition. In fact, damage-induced proteolysis of COL17A1 resulted in low expression of COL17A1, and stem cells with low level of COL17A1 will be eliminated by the COL17A1-high, undamaged stem cells through cell competition. Cell competition was also reported to be the fundamental mechanism by which stem cells with DSBs were selectively eliminated. It was shown that selective elimination was coupled with clonal expansion of intact stem cells, keeping the genomic integrity of the epidermis [148].
In conclusion, skin tissue reactions to radiation exposure are manifested by diverse cell types, originally resident or infiltrating, which orchestrate inflammation. Faithful recovery of the epidermis is critical to maintain the homeostasis of the skin. It should be mentioned that understanding of the process of injury and regeneration of skin appendages in response to radiation exposure is less examined compared with the regeneration of the epidermis. As skin appendages are indispensable for the function of the skin, further studies are needed [149–151].
Skin cancer
A proven link between exposure to ionizing radiation and the development of skin cancer was documented in a number of studies, including epidemiological studies among the atomic bomb (A-bomb) survivors, patients receiving radiotherapy and radiologists [9, 17–19, 152–155]. The two major types of skin cancers include BCC and SCC, in which the former accounts for ~90% of all skin cancers, so that BCC is the more frequent skin cancer induced by radiation exposure [9, 152, 156]. An excess relative risk (ERR) for BCC showed dose dependency in the LSS study among A-bomb survivors, and for doses less than 1 Gy, ERR per Gy is 0.48, while it is 2.64 per Gy for doses greater than 1 Gy [18, 19]. Age at exposure is another factor modifying risk, and the estimated ERR per Gy for age at exposure of 0–9, 10–19, 20–39, and over 40 years were 15, 5.7, 1.3, and 0.9, respectively [18, 19]. The ERR for SCC was estimated to be 0.71 per Gy. The dose−response of radiation risk for BCC has been reported to best fit a linear model with a threshold at 0.63 Gy, whose dose–response was not reported in other solid cancer cases [18]. A similar nonlinear dose–response was observed experimentally in an ICR/CRJ mice skin cancer model with fractionated beta-ray exposure, and three-times-weekly exposure through the life span of the mice to 0.5 Gy of beta-rays per fraction (the maximum total surface dose was 195 Gy) from a 90Sr-90Y source did not develop skin cancer [157–159], confirming the presence of a threshold dose.
As skin tumors related to radiation exposure are very similar clinically to sporadic skin tumors [9], IR may result in an earlier spontaneous onset of BCC and SCC, a possibility supported by the molecular mechanisms underlying BCC induction. It is well-described that mutations in the Ptch1 and the Smoothened genes, which result in dysregulated signaling of SHH pathway, leading to aberrant activation of the GLI transcription factor, are driver mutations associated with BCC, causing an inappropriate growth of basal cells [160–162]. Since mutations in those genes were caused predominantly by C > T transition, they seem not be induced by radiation exposure.
Since the mutational signatures in skin cancers induced by ionizing radiation have not yet been determined, we should be cautious to make a final conclusion. But, it is possible to hypothesize that radiation exposure gives rise to a circumstance, where basal cells with spontaneous mutations initiate clonal growth; otherwise, it will be eliminated by cell competition with neighboring normal cells. In fact, recent papers have suggested that radiation exposure may bring early onset of aging [163]. As skin cancers are apparently associated with skin aging, this could be the underlying mechanism, by which skin cancers become manifested among people receiving ionizing radiation.
It should be emphasized that epidemiological studies among A-bomb survivors [164, 165] and skin cancer patients who experienced radiation therapy [17] demonstrated people in an earlier age showed more susceptibility to skin cancer induction by radiation exposure. Also, it was shown that a linear decrease in the relative risk of head and neck skin cancer with increasing age at exposure to therapeutic radiation [165]. Thus, aging may not be a cause of the initiation of skin cancer but plays a role in the manifestation of the initiated cells that do develop into cancers.
SKIN CARE FOR MITIGATION OF RADIATION-INDUCED SKIN COMPLICATION
Acute radiation dermatitis, the representative adverse effect of radiotherapy, has been well documented in people with breast, and head and neck cancers [16]. Although there are several trials for preventing and management of acute radiation dermatitis, it is still a highly prevalent issue, causing impaired health-related quality of life, discomfort, pain, and reduction of treatment efficacy and poorer outcomes. This is due to a lack of a ‘gold standard’ for the prevention and management of radiation dermatitis, and we are still urged to establish particularly an international standard remedy for acute radiation dermatitis prevention and management.
Clinical practice guidelines on acute radiation dermatitis have been published by several organizations [166–171], such as Oncology Nursing Society, the Multinational Association of Supportive Care in Cancer (MASCC), British Columbia Cancer Agency, Cancer Care Manitoba, Society and College of Radiographers, and International Society of Nurses in Cancer Care, however, there are diverse differences in the recommendations, most likely due to the different approaches used in developing recommendations.
Recently, a unique attempt has been made and published by MASCC, which conducts a Delphi consensus-based method with compiling opinions and establishing a possible consensus based upon the evidence from the existing literatures [170]. Different therapeutic interventions have been discussed extensively, and six interventions, including photobiomodulation therapy and Mepitel® film for the patients with breast cancer treatment, Hydrofilm, mometasone, betamethasone and olive oil, are advocated as evidence-based skin-care recommendations.
Photobiomodulation therapy, also known as low-level laser therapy, uses low-power light sources in the visible and infra-red spectrum. Although the mechanism has not been fully understood yet, it is able to promote wound healing and alleviate inflammation [172]. To protect vulnerable skin, Mepitel® Film, a soft silicone transparent film dressing, is used for superficial lacerations. Radiotherapy for breast cancer tends to be more prone to radiation dermatitis than any other form of radiotherapy, but the use of Mepitel® Film has been reported to reduce the incidence of Grade 2 and Grade 3 radiation dermatitis. In addition, it has been shown to significantly reduce total Radiation-Induced Skin Reaction Assessment Scale (RISRAS) scores, which assess skin symptoms by both patients and healthcare professionals [173, 174]. A lower incidence of Grade 2 and Grade 3 radiodermatitis and lower mean RISRAS scores have also been reported in the case of head and neck cancer radiotherapy, but this differs from breast cancer radiotherapy, where poor adherence to medication and scratching have been identified as problems [175, 176].
Hydrofilm is a transparent film dressing, which has a semi-permeable hypoallergenic polyurethane polyacrylate top layer and a hypoallergenic, polyacrylate adhesive layer [177]. As shown with Mepitel® film, it also efficiently reduces the severity of radiation dermatitis. The use of topical corticosteroids is quite common for the treatment of radiation dermatitis, and it now turns out to be clear that mometasone furoate as well as betamethasone are effective for the prevention of severe radiation dermatitis, among which the latter is more effective [178]. Other types of intervention include the usage of topical Hirudoid®, a heparinoid-containing product, and wash with soap, both of which were reported to alleviate some of the symptoms. It is worth noticing that a recent paper comparing radiation dermatitis clinical practice guidelines demonstrated that the use of topical corticosteroids, and washing with water and soap, were consistently supported by all recommendations [179]. Thus, although there is still discordance among guideline recommendations, further comprehensive researches are expected to establish ideal and optimal recommendations for patients and frontline healthcare professionals.
SUMMARY AND FUTURE STUDIES
The skin cancer is one of the malignant adverse effects caused by radiation exposure. Ionizing radiation executes cell death in not only basal cells that are required for epidermis renewal, but also dermal and subcutaneous cells that support the epidermal homeostasis. It is quite unique to ionizing radiation that cell death mode manifested after exposure is primarily premature senescence, triggering a unique inflammatory immune response. Hence, cell death-initiating tissue reaction gives rise to visible clinical symptoms, which was extensively discussed in the previous studies. Understanding such tissue reactions more in detail, particularly the dynamics of stem cells and their competition in the basal layer, should be critical for avoiding unwanted adverse effects and assuring the quality of life of the patients receiving radiotherapy. It is also essential for proper estimation of the risk of skin cancer attributable to radiation exposure.
Contributor Information
Keiko Iwashita, Division of Adult Nursing, Department of Specialized Nursing, Oita University of Nursing and Health Sciences, 2944-9 Megusuno, Oita 840-1201, Japan.
Keiji Suzuki, Department of Radiation Medical Sciences, Atomic Bomb Disease Institute, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
Mitsuaki Ojima, Division of Adult Nursing, Department of Specialized Nursing, Oita University of Nursing and Health Sciences, 2944-9 Megusuno, Oita 840-1201, Japan.
CONFLICT OF INTEREST
All authors declare that there are no conflict of interest.
REFERENCES
- 1. Yin H, Hu M, Li D. Regulation of epidermal stratification and development by basal keratinocytes. J Cell Physiol 2023;238:742–8. 10.1002/jcp.30978. [DOI] [PubMed] [Google Scholar]
- 2. Swaney MH, Kalan LR. Living in your skin: microbes, molecules, and mechanisms. Infect Immun 2021;89:e00695–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Eyerich S, Eyerich K, Traidl-Hoffmann C, Biedermann T. Cutaneous barriers and skin immunity: differentiating a connected network. Trend Immunol 2018;39:315–27. 10.1016/j.it.2018.02.004. [DOI] [PubMed] [Google Scholar]
- 4. Proksch E, Brandner JM, Jensen J-M. The skin: an indispensable barrier. Exp Dermatol 2008;17:1063–72. 10.1111/j.1600-0625.2008.00786.x. [DOI] [PubMed] [Google Scholar]
- 5. Candi E, Schmidt R, Melino G. The cornified envelop: a model of cell death in the skin. Nat Rev Mol Cell Biol 2005;6:328–40. 10.1038/nrm1619. [DOI] [PubMed] [Google Scholar]
- 6. Wickett RR, Visscher MO. Structure and function of the epidermal barrier. Am J Infect Control 2006;34:S98–110. 10.1016/j.ajic.2006.05.295. [DOI] [Google Scholar]
- 7. Lefevre-Utile A, Braun C, Haftek M et al. Five functional aspects of the epidermal barrier. Int J Mol Sci 2012;22:11676. 10.3390/ijms222111676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Lim K-M. Skin epidermis and barrier function. Int J Mol Sci 2021;22:3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Martin MT, Vulin A, Hendry JH. Human epidermal stem cells: role in adverse skin reactions and carcinogenesis from radiation. Mutat Res 2016;770:349–68. 10.1016/j.mrrev.2016.08.004. [DOI] [PubMed] [Google Scholar]
- 10. ICRP . ICRP statement on tissue reactions/early and late effects of radiation in normal tissues and organs – threshold doses for tissue reactions in a radiation protection context. ICRP publication 118. Ann ICRP 2012;41:1–322. [DOI] [PubMed] [Google Scholar]
- 11. Barnett GC, West CML, Dunning AM et al. Normal tissue reactions to radiotherapy: towards tailoring treatment dose by genotype. Nat Rev Cancer 2009;9:134–42. 10.1038/nrc2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Wei J, Meng L, Hou X et al. Radiation-induced skin reactions: mechanism and treatment. Cancer Manag Res 2019;11:167–77. 10.2147/CMAR.S188655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Dicarlo AL, Bandremer AC, Hokkingsworth BA et al. Cutaneous radiation injuries: models, assessment and treatment. Radiat Res 2020;194:315–44. 10.1667/RADE-20-00120.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. McBride WH, Schaue D. Radiation-induced tissue damage and response. J Pathol 2020;250:647–55. 10.1002/path.5389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Yang X, Ren H, Guo X et al. Radiation-induced skin injury: pathogenesis, treatment, and management. Aging 2020; 12:23379–93. 10.18632/aging.103932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Rube CE, Freyter BM, Tewary G et al. Radiation dermatitis: radiation-induced effects on the structural and immunological barrier function of the epidermis. Int J Mol Sci 2024;25:3320. 10.3390/ijms25063320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Karagas MR, McDonald JA, Greenberg ER et al. Risk of basal cell and squamous cell skin cancers after ionizing radiation therapy. J Natl Cancer Inst 1996;88:1848–53. [DOI] [PubMed] [Google Scholar]
- 18. Sugiyama H, Misumi M, Kishikawa M et al. Skin cancer incidence among atomic bomb survivors from 1958 to 1996. Radiat Res 2014;181:531–9. 10.1667/RR13494.1. [DOI] [PubMed] [Google Scholar]
- 19. Ozasa K. Epidermiological research on radiation-induced cancer in atomic bomb survivors. J Radiat Res 2016;57:i112–7. 10.1093/jrr/rrw005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Weinstein GD, McCullough JL, Ross PR. Cell proliferation in normal epidermis. J Invest Dermatol 1984;82:623–8. 10.1111/1523-1747.ep12261462. [DOI] [PubMed] [Google Scholar]
- 21. Bergstresser PR, Taylor JR. Epidermal 'turnover time': a new examination. Br J Dermatol 1977;96:503–6. 10.1111/j.1365-2133.1977.tb07152.x. [DOI] [PubMed] [Google Scholar]
- 22. Koster MI. Making an epidermis. Ann N Y Acad Sci 2009; 1170:7–10. 10.1111/j.1749-6632.2009.04363.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Maeda K. New method of measurement of epidermal turnover in humans. Cosmetics 2017;4:47. 10.3390/cosmetics4040047. [DOI] [Google Scholar]
- 24. Kligman AM. Perspectives and problems in cutaneous gerontology. J Invest Dermatol 1979;73:39–46. 10.1111/1523-1747.ep12532758. [DOI] [PubMed] [Google Scholar]
- 25. Lavker RM. Structural alterations in exposed and unexposed aged skin. J Invest Dermatol 1979;73:59–66. 10.1111/1523-1747.ep12532763. [DOI] [PubMed] [Google Scholar]
- 26. Baumann L. Skin ageing and its treatment. J Pathol 2007; 211:241–51. 10.1002/path.2098. [DOI] [PubMed] [Google Scholar]
- 27. Lee H, Hong Y, Kim M. Structural and functional changes and possible molecular mechanisms in aged skin. Int J Mol Sci 2021;22:12489. 10.3390/ijms222212489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Liang Y, Su W, Wang F. Skin ageing: a progressive, multifactorial condition demanding an integrated, multilayer-targeted remedy. Clin Comset Investig Dermatol 2023;16:1215–29. 10.2147/CCID.S408765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Wyes SP, Carruthers JD, Dashti P et al. Cellular senescence in human skin aging: leveraging senotherapeutics. Gerontol 2024;70:7–14. 10.1159/000534756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Webb A, Li A, Kaur P. Location and phenotype of human adult keratinocyte stem cells of the skin. Different Res Biol Divers 2004; 72:387–95. 10.1111/j.1432-0436.2004.07208005.x. [DOI] [PubMed] [Google Scholar]
- 31. Rachidi W, Harfourche G, Kemaitre G et al. Sensing radiosensitivity of human epidermal stem cells. Radiother Oncol J Eur Soc Ther Radiol Oncol 2007;83:267–76. 10.1016/j.radonc.2007.05.007. [DOI] [PubMed] [Google Scholar]
- 32. Schneider TE, Barland C, Alex AM et al. Measuring stem cell frequency in epidermis: a quantitative in vivo functional assay for long-term repopulating cells. Proc Natl Acad Sci USA 2003; 100:11412–7. 10.1073/pnas.2034935100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Potten CS. The epidermal proliferative unit: the possible role of the central basal cell. Cell Tissue Kinet 1974;7:77–88. 10.1111/j.1365-2184.1974.tb00401.x. [DOI] [PubMed] [Google Scholar]
- 34. Potten CS. Keratinocyte stem cells, label-retaining cells and possible genome protection mechanisms. J Investig Dermatol Symp Proc 2004;9:183–95. 10.1111/j.1087-0024.2004.09305.x. [DOI] [PubMed] [Google Scholar]
- 35. Sada A, Jacob F, Leung E et al. Defining the cellular lineage hierarchy in the interfollicular epidermis of adult skin. Nat Cell Biol 2016;18:619–31. 10.1038/ncb3359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Gonzalez-Celeiro M, Zhang B, Hsu Y-C. Fate by chance, not by choice: epidermal stem cells go live. Cell Stem Cell 2016;19:8–10. 10.1016/j.stem.2016.06.010. [DOI] [PubMed] [Google Scholar]
- 37. Rompolas P, Mesa KR, Kawaguchi K et al. Spatiotemporal coordination of stem cell commitment during epidermal homeostasis. Science 2016;352:1471–4. 10.1126/science.aaf7012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Piedrafita G, Kostiou V, Wabik A et al. A single-progenitor model as the unifying paradigm of epidermal and esophageal epithelial maintenance in mice. Nat Commun 2020;11:1429. 10.1038/s41467-020-15258-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Wang S, Drummond ML, Guerrero-Jurarez C et al. Single cell transcriptomics of human epidermis identifies basal stem cell transition states. Nat Commun 2020;11:4239. 10.1038/s41467-020-18075-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Lee JH, Choi S. Deciphering the molecular mechanisms of stem cell dynamics in hair follicle regeneration. Exp Mol Med 2014;56:110–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Oh JW, Kloepper J, Langan EA et al. A guide to studying human hair follicle cycling in vivo. J Invest Dermatol 2016;136:34–44. 10.1038/JID.2015.354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Lin X, Zhu L, He J. Morphogenesis, growth cycle and molecular regulation of hair follicles. Front Cell Dev Biol 2022;10:3389. 10.3389/fcell.2022.899095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Natarelli N, Gahoonia N, Sivamani RK. Integrative and mechanistic approach to the hair growth cycle and hair loss. J Clin Med 2023;12:893. 10.3390/jcm12030893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Kretzschmar K, Watt FM. Markers of epidermal stem cell subpopulations in adult mammalian skin. Cold Spring Harb Perspect Med 2014;4:a013631. 10.1101/cshperspect.a013631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Rice G, Rompolas P. Advances in resolving the heterogeneity and dynamics of keratinocyte differentiation. Curr Opin Cell Biol 2020;67:92–8. 10.1016/j.ceb.2020.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Shi Z, Liu Z, Wei Y et al. The role of dermal fibroblasts in autoimmune skin diseases. Front Immunol 2014;15:1379490. 10.3389/fimmu.2024.1379490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Rippa AL, Kalbusheva EP, Vorotelyak EA. Regeneration of dermis: scaring and cells involved. Cells 2019;8:607. 10.3390/cells8060607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Braverman IM. The cutaneous microcirculation. J Invest Dermatol Symp Proc 2000;5:3–9. 10.1046/j.1087-0024.2000.00010.x. [DOI] [PubMed] [Google Scholar]
- 49. Dancik Y, Bigliardi PL, Bingliardi-Qi M. What happens in the skin? Integrative skin permeation kinetics into studies of developmental and reproductive toxicity following topical exposure. Reproduct Toxicol 2015;58:252–81. 10.1016/j.reprotox.2015.10.001. [DOI] [PubMed] [Google Scholar]
- 50. Deegan AJ, Wang RK. Microvasculature imaging of the skin. Phys Med Biol 2021;64:07TR01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Rivera-Gonzalez G, Shook B, Horsley V. Adipocytes in skin health and disease. Cold Spring Harb Perspect Med 2014; 4:a015271. 10.1101/cshperspect.a015271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Kruglikov IL, Scherer PE. Skin aging: are adipocytes the next target? Aging 2016;8:1457–69. 10.18632/aging.100999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Guerrero-Jurarez CF, Plikus MV. Emerging nonmetabolic functions of the skin fat. Nat Rev Endocrinol 2018;14:163–73. 10.1038/nrendo.2017.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Kruglikov I, Zhng Z, Scherer PE. The role of immature and mature adipocytes in hair cycling. Trend Endocrinol Metab 2019;30:93–105. 10.1016/j.tem.2018.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Jiang D, Rinkevich Y. Furnishing wound repair by the subcutaneous fascia. Int J Mol Sci 2012;22:9006. 10.3390/ijms22169006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Corres-Gallegos D, Jiang D, Christ S et al. Patch repair of deep wound by mobilized fascia. Nature 2019;576:287–92. 10.1038/s41586-019-1794-y. [DOI] [PubMed] [Google Scholar]
- 57. Ye H, Rinkevich Y. Fascia layer - a novel target for the application of biomaterials in skin wound healing. Int J Mol Sci 2023;24:2936. 10.3390/ijms24032936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Naldaiz-Gastesi N, Bahri OA, de Munain AL et al. The panniculus carnosus muscle: an evolutionary enigma at the intersection of distinct research field. J Anat 2018;233:275–88. 10.1111/joa.12840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Niessen CM. Tight junctions/adherens junctions: basic structure and function. Dermatol 2007;127:2525–32. [DOI] [PubMed] [Google Scholar]
- 60. Yamasaki K, Gallo LR. Antimicrobial peptides in human skin disease. Eur J Dermatol 2008;18:11–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Byrd AL, Belkaid Y, Srgre JA. The human skin microbiome. Nat Rev Microbiol 2018;16:143–55. [DOI] [PubMed] [Google Scholar]
- 62. Wertz PW. Lipids and the permeability and antimicrobial barriers of the skin. J Lipids 2018;2018:1–7. 10.1155/2018/5954034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Yang YY, Qu L, Mijakovic I et al. Advances in the skin microbiota and its roles in cutaneous diseases. Microb Cell Factories 2022;21:176. 10.1186/s12934-022-01901-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Zhu Y, Yu X, Cheng G. Human skin bacterial microbiota homeostasis: a delicate balance between health and disease. mLife 2023;2:107–20. 10.1002/mlf2.12064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Rawlings AV, Harding CR. Moisturization and skin barrier function. Dermatologic Ther 2004;17:43–8. 10.1111/j.1396-0296.2004.04S1005.x. [DOI] [PubMed] [Google Scholar]
- 66. Kezic SK, Calkoen KF, Fluhr JW et al. Natural moisturizing factor components in the stratum corneum as biomarkers of filaggrin genotype: evaluation of minimally invasive methods. Br J Dermatol 2009;161:1098–104. 10.1111/j.1365-2133.2009.09342.x. [DOI] [PubMed] [Google Scholar]
- 67. Voegeli R, Rawlings AV. Moisturizing at a molecular level - the basis of corneocare. Int J Cosmet Sci 2023;45:133–54. 10.1111/ics.12832. [DOI] [PubMed] [Google Scholar]
- 68. Kabashima K, Honda T, Ginhoux F et al. The immunological anatomy of the skin. Nat Rev Immunol 2019;19:19–30. 10.1038/s41577-018-0084-5. [DOI] [PubMed] [Google Scholar]
- 69. Nestle FO, Di Meglio P, Qin J-Z et al. Skin immune sentinels in health and disease. Nat Rev Immunol 2009;9:679–91. 10.1038/nri2622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Pasparakis M, Haase I, Nestle FO. Mechanisms regulating skin immunity and inflammation. Nat Rev Immunol 2014; 14:289–301. 10.1038/nri3646. [DOI] [PubMed] [Google Scholar]
- 71. Jiang Y, Tsoi LC, Billi AC et al. Cytokinocytes: the diverse contribution of keratinocytes to immune responses in skin. JCI Insight 2020;5:e142067. 10.1172/jci.insight.142067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Merad M, Ginhoux F, Collin M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat Rev Immunol 2008;8:935–47. 10.1038/nri2455. [DOI] [PubMed] [Google Scholar]
- 73. Doebel T, Voisin B, Nagao K. Langerhans cells - the macrophage in dendritic cell clothing. Trend Immunol 2017;38:817–28. 10.1016/j.it.2017.06.008. [DOI] [PubMed] [Google Scholar]
- 74. Kaplan DH. Ontogeny and function of murine epidermal Langerhans cells. Nat Immunol 2017;18:1068–75. 10.1038/ni.3815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Deckers J, Hammad H, Hoste E. Langerhans cells: sensing the environment in health and disease. Front Immunol 2018;9:93. 10.3389/fimmu.2018.00093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. West HC, Bennet CL. Redefining of the role of Langerhans cells as immune regulators within the skin. Front Immunol 2018;8:1941. 10.3389/fimmu.2017.01941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Thelen F, Witherden DA. Get in touch with dendritic epithelial T cells! Front Immunol 2020;11:1656. 10.3389/fimmu.2020.01656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Kamenjarin N, Hodapp K, Melchior F et al. Cross-presenting Langerhans cells are required for the early reactivation of resident CD8+ memory T cells in the epidermis. Proc Natl Acad Sci USA 2023;120:e2219932120. 10.1073/pnas.2219932120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Egbuniwe IU, Karagiannis SN, Nestle FO et al. Revisiting the role of B cells in skin immune surveillance. Trend Immunol 2015;36:102–11. 10.1016/j.it.2014.12.006. [DOI] [PubMed] [Google Scholar]
- 80. Lerman I, Mitchell DC, Richardson CT. Human cutaneous B cells: what do we know? Ann Transl Med 2021;9:440. 10.21037/atm-20-5185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Heath W, Carbone FR. The skin-resident and migratory immune system in steady state and memory: innate lymphocytes, dendritic cells and T cells. Nat Immunol 2013;14:978–85. 10.1038/ni.2680. [DOI] [PubMed] [Google Scholar]
- 82. Lim CH, Ito M. Tracking skin and immune cell interactions. Nat Cell Biol 2021;23:439–40. 10.1038/s41556-021-00680-3. [DOI] [PubMed] [Google Scholar]
- 83. Mestas J, Hughes CCW. Of mice and not men: difference between mouse and human immunology. J Immunol 2004; 172:2731–8. 10.4049/jimmunol.172.5.2731. [DOI] [PubMed] [Google Scholar]
- 84. MacLeod AS, Havran WL. Functions of skin-resident gdT cells. Cell Mol Life Sci 2011;68:2399–408. 10.1007/s00018-011-0702-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Ariotti S, Beltman JB, Chodaczek G et al. Tissue-resident memory CD8+ T cells continuously patrol skin epithelia to quickly recognize local antigen. Proc Natl Acad Sci USA 2012; 109:19739–44. 10.1073/pnas.1208927109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Nielsen MM, Witherden DA, Havran WL. gdT cells in homeostasis and host defence of epithelial barrier tissues. Nat Rev Immunol 2017;17:733–45. 10.1038/nri.2017.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Tikoo S, Jain R, Kurz ARM et al. The lymphoid cell network in the skin. Immunol Cell Biol 2018;96:485–96. 10.1111/imcb.12026. [DOI] [PubMed] [Google Scholar]
- 88. Kobayashi T, Ricardo-Gonzalez RR, Moro K. Skin-resident lymphoid cells - cutaneous innate guardians and regulators. Trend Immunol 2020;41:100–12. 10.1016/j.it.2019.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Mraz V, Geisler C, Bonegeld CM. Dendritic epidermal T cells in allergic contact dermatitis. Front Immunol 2020;11:874. 10.3389/fimmu.2020.00874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Hirai T, Whitley SK, Kaplan DH. Migration and function of memory CD8+ T cells in skin. J Invest Dermatol 2020; 140:748–55. 10.1016/j.jid.2019.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Wagner M, Koyasu S. Innate lymphoid cells in skin homeostasis and malignancy. Front Immunol 2021;12:758522. 10.3389/fimmu.2021.758522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Ramirez-Labrada A, Pesini C, Santiago L et al. All about (NK cell-mediated) death in two acts and an unexpected encore: initiation, execution and activation of adaptive immunity. Front Immunol 2022;13:896228. 10.3389/fimmu.2022.896228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Cavalcante-Silva J, Koh TJ. Role of NK cells in skin wound healing of mice. J Immunol 2023;210:981–90. 10.4049/jimmunol.2200637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Suzuki K, Amrenova A, Mitsutake N. Recent advances in radiobiology with respect to pleiotropic aspects of tissue reaction. J Radiat Res 2021;62:i30–5. 10.1093/jrr/rraa086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Lippens S, Denecker G, Ovaere P et al. Death penalty for keratinocytes: apoptosis versus cornification. Cell Death Differ 2005;12:1497–508. 10.1038/sj.cdd.4401722. [DOI] [PubMed] [Google Scholar]
- 96. Kim JH, Jenrow KA, Brown SL. Mechanisms of radiation-induced normal tissue toxicity and implications for future clinical trials. Radiat Oncol J 2014;32:103–15. 10.3857/roj.2014.32.3.103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Tu W, Tang S, Yan T et al. Integrative multi-omic analysis of radiation-induced skin injury reveals the alteration of fatty acid metabolism in early response of ionizing radiation. J Dermatol Sci 2022;108:178–86. 10.1016/j.jdermsci.2023.01.001. [DOI] [PubMed] [Google Scholar]
- 98. Sugaya K, Ishihara Y, Inoue S et al. The effects of gamma rays on the regeneration of hair follicles are carried over to late hair cycles. Int J Radiat Biol 2015;91:957–63. 10.3109/09553002.2015.1101647. [DOI] [PubMed] [Google Scholar]
- 99. Duran C, Barcenas M, Wang Q. Modeling of ionizing radiation induced hair follicle regenerative dynamics. J Theoret Biol 2022;555:111283. 10.1016/j.jtbi.2022.111283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Botchkareva N, Ahluwalia G, Shander D. Apoptosis in the hair follicle. J Invest Dermatol 2006;126:258–64. 10.1038/sj.jid.5700007. [DOI] [PubMed] [Google Scholar]
- 101. Fischer H, Fumicz J, Rossiter H et al. Holocrine secretion of sebum is a unique DNase2-dependent mode of programmed cell death. J Invest Dermatol 2017;137:587–94. 10.1016/j.jid.2016.10.017. [DOI] [PubMed] [Google Scholar]
- 102. Jang H, Myung H, Lee J et al. Impaired skin barrier due to sebaceous gland atrophy in the later stage of radiation-induced skin injury: application of non-invasive diagnostic methods. Int J Mol Sci 2018;19:185. 10.3390/ijms19010185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Veniaminova NA, Jia YY, Hartigan AM et al. Distinct mechanisms for sebaceous gland self-renewal and regeneration provide durability in response to injury. Cell Rep 2023;42:113121. 10.1016/j.celrep.2023.113121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Wang W, Wang H, Long Y et al. Controlling hair loss by regulating apoptosis in hair follicles: a comprehensive overview. Biomolecules 2023;14:20. 10.3390/biom14010020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Ryan JL. Ionizing radiation: the good, the bad, and the ugly. J Clinical Dermatol 2012;132:985–93. 10.1038/jid.2011.411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. ICRP . The biological basis for dose limitation in the skin. A report of a task group committee 1 of the international commission on radiological protection. Ann ICRP 1991;22:A1–104. 10.1016/0146-6453(91)90019-D. [DOI] [PubMed] [Google Scholar]
- 107. Blanpain C, Fuchs E. Epidermal homeostasis: a balancing act of stem cells in the skin. Nat Rev Mol Cell Biol 2009;10:207–17. 10.1038/nrm2636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Pena OA, Martin P. Cellular and molecular mechanisms of skin wound healing. Nat Rev Mol Cell Biol 2024;25:599–616. 10.1038/s41580-024-00715-1. [DOI] [PubMed] [Google Scholar]
- 109. Zwierello W, Piorun K, Skorka-Majewicz et al. Burns: classification, pathophysiology, and treatments: a review. Int J Mol Sci 2023;24:3749. 10.3390/ijms24043749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Bhattacharya S. Radiation injury. Indian J Plast Surg Supplement 2010;43:S91–3. 10.1055/s-0039-1699465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. DOA . Nuclear handbook for medical service personnel. In: Department of the Army Technical Manual. Vol. 215, 1–93, Washington: Department of the Army, 1969. [Google Scholar]
- 112. Chin T, Lee XE, Ng PY et al. The role of cellular senescence in skin aging and age-related skin pathologies. Front Physiol 2023; 14:1297637. 10.3389/fphys.2023.1297637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Tewary G, Freyter B, Al-razaq MA et al. Immunomodulatory effects of histone variant H2A.J in ionizing radiation dermatitis. Int J Radiat Oncol Biol Phys 2024;118:801–16. 10.1016/j.ijrobp.2023.09.022. [DOI] [PubMed] [Google Scholar]
- 114. Lecomte K, toniolo A, Hoste E. Cell death as an architect of adult skin stem cell niches. Cell Death Differ 2024;31:957–69. 10.1038/s41418-024-01297-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Huang WY, Lai SF, Chiu HY et al. Mobilizing transit-amplifying cell-derived ectopic progenitors prevents hair loss from chemotherapy and radiation therapy. Cancer Res 2017;77:6083–96. 10.1158/0008-5472.CAN-17-0667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. O'Sullivan JD, Nicu C, Picard M et al. The biology of human hair greying. Biol Rev Camb Philos Soc 2021;96:107–28. 10.1111/brv.12648. [DOI] [PubMed] [Google Scholar]
- 117. Aoki H, Hara A, Motohashi T et al. Keratinocyte stem cells but not melanocyte stem cells are the primary target for radiation-induced hair greying. J Invest Dermatol 2013;133:2143–51. 10.1038/jid.2013.155. [DOI] [PubMed] [Google Scholar]
- 118. Yu H, Zhong T, Xu Y et al. Molecular profiling of skin cells identifies distinct cellular signatures in radiation-induced skin injury across various stages in the murine dataset. Exp Hematol Oncol 2025;14:18. 10.1186/s40164-025-00596-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Yan T, Yang P, Bai H et al. Single-cell RNA-seq analysis of molecular changes during radiation-induced skin injury: the involvement of Nur77. Theranostics 2024;14:5809–25. 10.7150/thno.100417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Coppe JP, Desprez PY, Krtolica A et al. The senescence-associated secretory phenotype: the dark side of tumor suppression. Ann Rev Pathol Mech Dis 2010;5:99–118. 10.1146/annurev-pathol-121808-102144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Munoz-Espin D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol 2014;15:482–96. 10.1038/nrm3823. [DOI] [PubMed] [Google Scholar]
- 122. Watanabe S, Kawamoto S, Ohtani N et al. Impact of senescence-associated secretory phenotype and its potential as a therapeutic target for senescence-associated diseases. Cancer Sci 2017;108:563–9. 10.1111/cas.13184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer 2019; 19:439–53. 10.1038/s41568-019-0156-2. [DOI] [PubMed] [Google Scholar]
- 124. Gurkar AU, Generencser AA, Mora AL et al. Spatial mapping of cellular senescence: emerging challenges and opportunities. Nat Aging 2023;3:776–90. 10.1038/s43587-023-00446-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Kennedy-Crispin M, Billick E, Mitsui H et al. Human keratinocytes’ response to injury upregulates CCL20 and other genes linking innate and adaptive immunity. J Invest Dermatol 2012;132:105–13. 10.1038/jid.2011.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Liao W, Hei TK, Cheng SK. Radiation-induced dermatitis is mediated by IL17-expressing gd T cells. Radiat Res 2017; 187:464–74. 10.1667/RR007CC.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Furue K, Ito T, Tsuji G et al. The CCL20 and CCR6 axis in psoriasis. Immunol 2019;91:e12846. [DOI] [PubMed] [Google Scholar]
- 128. Liu T, Li S, Ying S et al. The IL-23/IL-17 pathway in inflammatory skin diseases: from bench to bedside. Front Immunol 2020; 11:594735. 10.3389/fimmu.2020.594735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Shin JW, Kwon M, Hwang J et al. Keratinocyte transglutaminase 2 promotes CCR6+ γδT-cell recruitment by upregulating CCL20 in psoriatic inflammation. Cell Death Disease 2020;11:301. 10.1038/s41419-020-2495-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Martin P, Goldstein JD, Mermoud L et al. IL-1 family antagonists in mouse and human skin inflammation. Front Immunol 2021; 12:652846. 10.3389/fimmu.2021.652846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Paldor M, Levkovitch-Siany O, Eidelshtein D et al. Single-cell transcriptomics reveals a senescence-associated IL-6/CCR6 axis driving radiodermatitis. EMBO Mol Med 2022;14:e15653. 10.15252/emmm.202115653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Chen Y, Ma L, Cheng Z et al. Senescent fibroblast facilitates re-epithelization and collagen deposition in radiation-induced skin injury through IL-33-mediated macrophage polarization. J Transl Med 2024;22:176. 10.1186/s12967-024-04972-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Wang Y, Chen S, Bao S et al. Deciphering the fibrotic process: mechanism of chronic radiation skin injury fibrosis. Front Immunol 2024;15:1338922. 10.3389/fimmu.2024.1338922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Sagiv A, Burton DGA, Moshayev Z et al. NKG2D ligands mediate immunosurveillance of senescent cells. Aging 2016;8:328–44. 10.18632/aging.100897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Wensveen FM, Jelencic V, Plic B. NKG2D: a master regulator of immune cell responsiveness. Front Immunol 2018;9:441. 10.3389/fimmu.2018.00441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Antonangeli F, Zingoni A, Soriani A et al. Senescence cells: living or dying is a matter of NK cells. J Leukoc Biol 2019; 105:1275–83. 10.1002/JLB.MR0718-299R. [DOI] [PubMed] [Google Scholar]
- 137. Letafati A, Ardekani OS, Naderisemiromi M et al. Unraveling the dynamic mechanisms of natural killer cells in viral infections: insights and implications. Virol J 2024;21:18. 10.1186/s12985-024-02287-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Torcellan T, Friedrich C, Doucet-Ladeveze RD et al. Circulating NK cells establish tissue residency upon acute infection of skin and mediate accelerated effector responses to secondary infection. Immunity 2024;57:124–140.e7. 10.1016/j.immuni.2023.11.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Kawase Y, Naito S, Ito M et al. The effect of ionizing radiation on epidermal Langerhans cells. J Radiat Res 1990;31:246–55. [DOI] [PubMed] [Google Scholar]
- 140. Berzaghi R, Tomaas S, Lode K et al. Ionizing radiation curtails immunosuppressive effects from cancer-associated fibroblasts on dendritic cells. Front Immunol 2021;12:662594. 10.3389/fimmu.2021.662594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Pinto AT, Machado AB, Osorio H et al. Macrophage resistance to ionizing radiation exposure is accompanied by decreased cathepsin D and increased transferrin receptor an expression. Cancers 2023;15:270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Hu W, Shang R, Yang J et al. Skin γδT cells and their function in wound healing. Front Immunol 2022;13:875076. 10.3389/fimmu.2022.875076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Fan L, Busser BW, Lifsted TQ et al. Antigen presentation by keratinocytes directs autoimmune skin disease. Proc Natl Acad Sci USA 2003;100:3386–91. 10.1073/pnas.0437899100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Kitashima DY, Kobayashi T, Woodring T et al. Langerhans cells prevent autoimmune via expansion of keratinocyte antigen-specific regulatory T cells. EBioMed 2018;27:293–303. 10.1016/j.ebiom.2017.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Mueller SN, Zaid A, Carbone FR. Tissue-resident T cells: dynamic players in skin immunity. Front Immunol 2014;5:332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Zhou Y, Ni H, Balint K et al. Ionizing radiation selectively reduces skin regulatory T cells and alters immune function. PLoS One 2014;9:e100800. 10.1371/journal.pone.0100800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Liu N, Matsumura H, Kato T et al. Stem cell competition orchestrates skin homeostasis and ageing. Nature 2019;568:344–50. 10.1038/s41586-019-1085-7. [DOI] [PubMed] [Google Scholar]
- 148. Kato T, Liu N, Morinaga H et al. Dynamic stem cells selection safeguards the genomic integrity of the epidermis. Dev Cell 2021;56:3309–3320.e5. 10.1016/j.devcel.2021.11.018. [DOI] [PubMed] [Google Scholar]
- 149. Ogita M, Sekiguchi K, Akahane K et al. Damage to sebaceous gland and the efficacy of moisturizer after whole breast radiotherapy: a randomized controlled trial. BMC Cancer 2019;19:125. 10.1186/s12885-019-5334-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Lu C, Fuchs E. Sweat gland progenitors in development, homeostasis, and wound repair. Cold Spring Harb Perspect Med 2014;4:a015222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Weng T, Wu P, Zhang W et al. Regeneration of skin appendages and nerves: current status and further challenges. J Transl Med 2020;18:53. 10.1186/s12967-020-02248-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Li C, Athar M. Ionizing radiation exposure and basal cell carcinoma pathogenesis. Radiat Res 2016;185:217–28. 10.1667/RR4284.S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Watt TC, Inskip PD, Stratton K et al. Radiation-related risk of basal cell carcinoma: A report from the childhood cancer survivors study. J Natl Cancer Inst 2012;104:1240–50. 10.1093/jnci/djs298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Lee T, Sigurdson AJ, Preston DL et al. Occupational ionising radiation and risk of basal cell carcinoma in US radiogenic technologists (1983-2005). Occup Environ Med 2015;72:862–9. 10.1136/oemed-2015-102880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Azizova T, Bannikova MV, Grigoryeva ES et al. Risk of malignant skin neoplasm in a cohort of workers occupationally exposed to ionizing radiation at low dose rates. PLoS One 2018;13:e0205060. 10.1371/journal.pone.0205060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. ICRP . Stem cell biology with respect to carcinogenesis aspects of radiological protection. ICRP publication 131. Ann ICRP 2015;44:1-357. [DOI] [PubMed] [Google Scholar]
- 157. Ootsuyama A, Tanooka H. One hundred percent tumor induction in mouse skin after repeated b irradiation in a limited dose range. Radiat Res 1988;115:488–94. 10.2307/3577298. [DOI] [PubMed] [Google Scholar]
- 158. Ootsuyama A, Tanooka H. Threshold-like dose of local b irradiation repeated throughout the life span of mice for induction of skin and bone tumors. Radiat Res 1991;125:98–101. 10.2307/3577987. [DOI] [PubMed] [Google Scholar]
- 159. Ootsuyama A, Tanooka H. Zero tumor incidence in mice after repeated lifetime exposures to 0.5 Gy of beta radiation. Radiat Res 1993;134:244–6. 10.2307/3578465. [DOI] [PubMed] [Google Scholar]
- 160. Ng JMY, Curran T. The Hedgehog’s tale: developing strategies for targeting cancer. Nat Rev Cancer 2011;11:493–501. 10.1038/nrc3079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Pietrobono S, Gagliardi S, Stecca B. Non-canonical hedgehog signaling pathway in cancer: activation of GLI transcription factors beyond smoothened. Front Genet 2019;10:556. 10.3389/fgene.2019.00556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Pellegrini C, Maturo MG, Nardo LD et al. Understanding the molecular genetics of basal cell carcinoma. Int J Mol Sci 2017;18:2485. 10.3390/ijms18112485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Nakamura N. An examination of the dose rate effect in mice assuming that the carcinogenic effect of radiation is life shortening resulting from a tissue reaction. Int J Radiat Biol 2025; 101:225–31. 10.1080/09553002.2024.2442690. [DOI] [PubMed] [Google Scholar]
- 164. Thompson DE, Mabuchi K, Ron E et al. Cancer incidence in atomic bomb survivors. Part II solid tumors, 1958-1987. Radiat Res 1994;137:S17–67. 10.2307/3578892. [DOI] [PubMed] [Google Scholar]
- 165. Ron E, Modan B, Preston D et al. Radiation-induced skin carcinomas of the head and neck. Radiat Res 1991;125:318–25. 10.2307/3578117. [DOI] [PubMed] [Google Scholar]
- 166. Gosselin T, Ginex PK, Backler C et al. ONS guide lines for cancer treatment-related radiodermatitis. Oncol Nurs Forum 2020;47:654–70. 10.1188/20.ONF.654-670. [DOI] [PubMed] [Google Scholar]
- 167. Finkelstein S, Kanee L, Behroozian T et al. Comparison of clinical guidelines on radiation dermatitis: a narrative review. Support Care Cancer 2022;30:4663–74. 10.1007/s00520-022-06829-6. [DOI] [PubMed] [Google Scholar]
- 168. Bonomo P, Wolf JR. Shedding light on the management of acute radiation dermatitis: insight from the MASCC oncodermatology study group. Support Care Cancer 2023;31:568. 10.1007/s00520-023-08011-y. [DOI] [PubMed] [Google Scholar]
- 169. Behroozian T, Goldstein D, Wolf JR et al. MASCC clinical practice guidelines for the prevention and management of acute radiation dermatitis. Part 1: systematic review. eClinicalMed 2023;58:101886. 10.1016/j.eclinm.2023.101886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Behroozian T, Bonomo P, Patel P et al. Multinational association of supportive care in cancer (MASCC) clinical practice guidelines for the prevention and management of acute radiation dermatitis: international Delphi consensus-based recommendations. Lancet Oncol 2023;24:e172–85. 10.1016/S1470-2045(23)00067-0. [DOI] [PubMed] [Google Scholar]
- 171. Behroozian T, Caini S, van den Hurk C et al. Systematic review and meta-analysis on interventions for radiation dermatitis prevention and management: an overview of the method. Support Care Cancer 2023;31:261. 10.1007/s00520-023-07707-5. [DOI] [PubMed] [Google Scholar]
- 172. Gobbo M, Rico V, Marta GN et al. Photobiomodulation therapy for the prevention of acute radiation dermatitis: a systematic review and meta-analysis. Support Care Cancer 2023;31:227. 10.1007/s00520-023-07673-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Herst PM, Bennett NC, Sutherland AE et al. Prophylactic use of Mepitel film prevents radiation-induced moist desquamation in an intra-patient randomised controlled clinical trial of 78 breast cancer patients. Radiother Oncol 2014;110:137–43. 10.1016/j.radonc.2014.01.005. [DOI] [PubMed] [Google Scholar]
- 174. Behroozian T, Milton L, Karam I et al. Mepitel film for the prevention of acute radiation dermatitis in breast cancer: a randomized multicenter open-label phase III trial. J Clin Oncol 2022;41:1250–64. 10.1200/JCO.22.01873. [DOI] [PubMed] [Google Scholar]
- 175. Wooding H, Yan J, Yuan L et al. The effect of Mepitel film on acute radiation-induced skin reactions in head and neck cancer patients: a feasibility study. Br J Radiol 2018;91:20170298. 10.1259/bjr.20170298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Yan J, Yuan L, Wang J et al. Mepitel film is superior to Biafine cream in managing acute radiation-induced skin reactions in head and neck cancer patients: a randomised intra-patient controlled clinical trial. J Med Radiat Sci 2020;67:208–16. 10.1002/jmrs.397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Robijns J, Aquilano M, Banerjee S et al. Barrier films and dressings for the prevention of acute radiation dermatitis: a systemic review and meta-analysis. Support Care Cancer 2023;31:219. [DOI] [PubMed] [Google Scholar]
- 178. Tam S, Zhou G, Trombetta M et al. Topical corticosteroids for the prevention of severe radiation dermatitis: a systematic review and meta-analysis. Support Care Cancer 2023;31:382. 10.1007/s00520-023-07820-5. [DOI] [PubMed] [Google Scholar]
- 179. Roy I, Fortin A, Larochelle M. The impact of skin washing with water and soap during breast irradiation: a randomized study. Radiother Oncol 2001;58:333–9. 10.1016/S0167-8140(00)00322-4. [DOI] [PubMed] [Google Scholar]


