Figure 3 - uploaded by Tadamichi Shimizu
Content may be subject to copyright.
Epiderm, a human three-dimensional epidermal model, was exposed to 50 mJ per cm 2 UVB irradiation. The culture supernatants were obtained before and 1, 6, 12, 24, and 48 h after UVB irradiation and their MIF contents were measured by ELISA as described in Materials and Methods . MIF contents with ( d ) and without ( s ) UVB irradiation. Results are expressed as mean Ϯ SD of three different experiments. 

Epiderm, a human three-dimensional epidermal model, was exposed to 50 mJ per cm 2 UVB irradiation. The culture supernatants were obtained before and 1, 6, 12, 24, and 48 h after UVB irradiation and their MIF contents were measured by ELISA as described in Materials and Methods . MIF contents with ( d ) and without ( s ) UVB irradiation. Results are expressed as mean Ϯ SD of three different experiments. 

Source publication
Article
Full-text available
Human epidermal cells are capable of secreting various cytokines with immunologic, inflammatory, and proliferative properties. In a previous study, by reverse transcription-polymerase chain reaction and immunohistochemical analysis, we have shown that human epidermal keratinocytes express macrophage migration inhibitory factor and identified its pr...

Contexts in source publication

Context 1
... culture media of lower chambers were obtained before and after UVB irradiation at 1, 6, 12, 24, and 48 h. The results showed that MIF was readily released into the medium from 1 h after the irradiation, and formed biphasic maximal peaks at 6 and 48 h after the exposure (Fig 3). ...
Context 2
... skin is an important physical barrier that protects the body from the damage caused by the outside environment, including trauma, bacterial infection, and ultraviolet (UV) irradiation. As for the biologic mechanism, the skin is known to secrete a number of cytokines that act as immunologic and inflammatory mediators for self-defense. In particular, keratinocytes, which constitute the majority of the cell mass of the epidermis, are a major source for a wide array of these cytokines ( Sauder et al , 1990). Indeed, there is emerging evidence that keratinocytes participate in cutaneous inflammatory reactions and immune responses by producing a variety of cytokines. UV irradiation may trigger cutaneous inflammatory responses by stimulating epidermal keratinocytes to produce biologically potent cytokines such as interleukin (IL)-1 (Kupper et al , 1987; Ansel et al , 1988), IL-6 (Kirnbauer et al , 1991), and tumour necrosis factor (TNF)- α (K ̈ ck et al , 1990). These cytokines are involved not only in the mediation of local inflammatory reactions within the epidermis, but also exert systemic effects through entrance into the circulation. In a previous study, we have shown by reverse transcriptase- polymerase chain reaction (RT-PCR) and immunoblot analysis that human epidermal keratinocytes express macrophage migration inhibitory factor (MIF). Immunohistochemical study revealed that MIF exists in human epidermis, especially in the basal layer (Shimizu et al , 1996). This finding suggested the possibility that MIF regulates epidermal immunity, inflammation, and cellular differentiation. Human MIF cDNA was first cloned from activated T cells, demonstrating that this protein consists of 114 amino acid residues (Weiser et al , 1989). Following this finding various novel properties of MIF have been elucidated. It was reported that MIF is mainly expressed in T lymphocytes and macrophages; however, recent studies have revealed that this protein is ubiquitously expressed in various cells (Lanahan et al , 1992; Wistow et al , 1993; Onodera et al , 1995; Imamura et al , 1996; Suzuki et al , 1996). To date, MIF is known to respond to stimuli such as wounds and infection and is considered to contribute to regulation of inflammatory and immunologic responses to such tissue damage (Bernhagen et al , 1993; Calandra et al , 1994). Recently, it was reported that endotoxin administration induced expression of MIF protein and mRNA in various tissues of the rat (Bacher et al , 1997). As for the environmental effects on the skin, the most relevant physical injury to human skin is UVB radiation. MIF produced by keratinocytes may be profoundly involved in cutaneous inflammatory responses; however, the precise pathophysiologic function of MIF in the skin still remains unclear. UVB radiation constitutes one of the most important physiologic modulators of inflammatory and immunologic processes in human skin. We, therefore, examined the production of MIF induced in keratinocytes by UVB in vivo and in vitro to evaluate its pathophysiologic role. RESULTS In vitro UVB exposure At 24 and 48 h after UVB irradiation, a significant decrease of viability was observed only after more than 100 mJ per cm 2 UVB irradiation (data not shown). To examine the release of MIF caused by UVB exposure, we irradiated the cultured keratinocytes with various amounts of UVB (0–125 mJ per cm 2 ). A significant increase of MIF secretion in the supernatants of cultured keratinocytes was detected at 48 h after UVB radiation in a concentration-dependent manner up to 100 mJ per cm 2 ( Fig 1 A ). Similarly, a significant increase of TNF- α release was demonstrated after UVB radiation until 75 mJ per cm 2 ( Fig 1 B ). Extracellular secretion of MIF was observed at 1 h after UVB irradiation with 50 mJ per cm 2 , and increased in a time-dependent manner up to 48 h ( Fig 2 A ). On the other hand, the release of TNF- α after UVB irradiation reached its maximum at 24 h ( Fig 2 B ). To examine in more detail whether UVB exposure could induce MIF release from the multilayered epidermis, EpiDerm, a three-dimensional epidermis with highly differentiated charac- teristics, was exposed to 50 mJ per cm 2 UVB irradiation through the air–medium interface. The culture media of lower chambers were obtained before and after UVB irradiation at 1, 6, 12, 24, and 48 h. The results showed that MIF was readily released into the medium from 1 h after the irradiation, and formed biphasic maximal peaks at 6 and 48 h after the exposure ( Fig 3 ). MIF content in human epidermal sheet culture supernatants was assessed before and after UVB irradiation. Significant amounts of MIF were detected 12–24 h after 50 mJ per cm 2 UVB irradiation, 44.2 Ϯ 6.0 and 108.5 Ϯ 12.5 ng per ml, respectively ( Fig 4 ). These results were confirmed by immunohistochemical analysis at 12 h after UVB irradiation. This revealed extensive positive staining of all the layers in the epidermis with the anti-MIF antibody ( Fig 5 b ). On the other hand, predominant staining was observed mostly in the basal layer for sham-irradiated skin ( Fig 5 a ) as described previously (Shimizu et al , 1996). No specific positive staining was observed when the tissue was reacted with nonimmune rabbit IgG (data not shown). To examine whether the induction of MIF by human epidermis is regulated at the transcriptional level, the expression of MIF mRNA in cultured keratinocytes was examined by northern blot analysis after 50 mJ per cm 2 UVB exposure. Keratinocytes constitutively expressed MIF mRNA without exposure, and the expression level was upregulated during 1–6 h by 1.4–2.1-fold in comparison with the basal level (0 h) after the UVB exposure and slightly decreased at 12 h ( μ 0.8-fold) ( Fig 6 ). Following this, the expression level was restored to the basal level during 24–48 h. In vivo UVB exposure When serum MIF content was investi- gated in human volunteers before and after UVB exposure (four MED), minimal levels of MIF content could be detected before irradiation (10.8 Ϯ 1.5 ng per ml). After UVB exposure, significantly elevated serum MIF was detected at 48 h (58.1 Ϯ 20.6 ng per ml) ( Fig 7 ). MIF levels were biphasic after the UVB exposure, even though there were some individual differences ( Fig 8 a – e ). The first peak was observed between 1 and 12 h after irradiation and ranged from 15.1 to 82.8 ng per ml (early response). The second peak of MIF content was observed at 12–48 h after UV irradiation and ranged from 24.1 and 132.6 ng per ml (late response) when clinical signs of sunburn were clearly observed. DISCUSSION It has been reported that various cytokines play important parts in the homeostasis of normal skin and in its pathologic states (Barker et al , 1991). Release of cytokines by keratinocytes can be induced by a number of environmental factors, including UV light, trauma, and bacterial toxins. It is well recognized that excessive exposure to sunlight results in a number of deleterious consequences such as erythema and inflammation of the skin. Moreover, UV radiation has a direct carcinogenic effect, which indicates that exposure to sunlight can result in profound immunologic alterations (Scotto et al , 1974). There is increasing evidence that keratinocytes participate in cutaneous immunologic and inflammatory reactions through production of a variety of cytokines. To date, keratinocytes have been shown to secrete several UVB-induced cytokines such as IL-1, IL-3, IL-6, IL-10, IL-15, and TNF- α (Kupper et al , 1987; Ansel et al , 1988; Oxholm et al , 1988; K ̈ ck et al , 1990; Gallo et al , 1991; Kirnbauer et al , 1991; Enk et al , 1993, 1995; Mohamadzadeh et al , 1995). For example, TNF- α is an immunologically potent cytokine regulating immunologic and inflammatory responses of the skin (Oxholm et al , 1988; K ̈ ck et al , 1990), and TNF- α and IL-10 released from keratinocytes subsequent to UVB exposure may play a part in the induction of immunosuppression and tolerance (Yoshikawa and Streilein, 1990; Enk et al , 1993; Shimizu and Streilein, 1994). This study describes enhancement of MIF production in the skin by UVB irradiation in vivo and in vitro . It is of interest that two peaks of serum MIF content were observed after UVB exposure in vivo . The first peak was observed at 1–12 h (early response) after the exposure and the second peak at 12–48 h (late response). In vitro , we carried out northern blot analysis to examine the effect of UVB on the expression of MIF mRNA using cultured keratinocytes, and observed upregulation of the MIF mRNA level during 1–6 h after UVB exposure, but a slight decrease at 12 h. Thereafter, the expression level recovered to the basal level during 24–48 h. It was reported that positive MIF immunostaining of cultured pituitary cells disappeared after 16 h of incubation with lipopolysaccharide (LPS) and, in contrast, MIF mRNA expression was increased (Bernhagen et al , 1993). Similarly, intracellular MIF of macrophages disappeared concomitant with the increase of MIF in the culture medium in response to LPS (Calandra et al , 1994). Consistent with these findings, we found an increase of MIF content in the culture medium after UVB exposure. Based on these facts, it is speculated that the major part of the increased serum MIF content in the early response after UVB exposure in vivo may correspond to the release of MIF protein from keratinocytes. On the other hand, the source of increased serum content MIF in the late response has ...

Citations

... The most important environmental factor leading to skin cancer is UV radiation, which is estimated to be responsible for almost 90% of NMSCs 38,39 . UV exposure triggers acute inflammation in the skin by stimulating the production of proinflammatory cytokines, including MIF and its homolog D-DT 31,40,41 . The release of such cytokines (as well as neurotransmitters, endocrine factors, and neuropeptides) is a local effect after UVB exposure that can cause systemic effects leading to inflammatory diseases and malignancies 42,43 . ...
Article
Full-text available
Non-melanoma skin cancer (NMSC) is the most common cancer in Caucasians worldwide. We investigated the pathophysiological role of MIF and its homolog D-DT in UVB- and chemically induced NMSC using Mif−/−, D-dt−/− and Mif−/−/D-dt−/− mice on a hairless SKH1 background. Knockout of both cytokines showed similar attenuating effects on inflammation after acute UVB irradiation and tumor formation during chronic UVB irradiation, without additive protective effects noted in double knockout mice, indicating that both cytokines activate a similar signaling threshold. In contrast, genetic deletion of Mif and D-dt had no major effects on chemically induced skin tumors. To get insight into the contributing mechanisms, we used an in vitro 3D skin model with incorporated macrophages. Application of recombinant MIF and D-DT led to an accumulation of macrophages within the epidermal part that could be reversed by selective inhibitors of MIF and D-DT pathways. In summary, our data indicate that MIF and D-DT contribute to the development and progression of UVB- but not chemically induced NMSC, a role at least partially accounted by effects of both cytokines on epidermal macrophage accumulation. These data highlight that MIF and D-DT are both potential therapeutic targets for the prevention of photocarcinogenesis but not chemical carcinogenesis.
... Further immunofluorescence analysis suggested MIF was saliently overabundant in vitiligo lesional skin compared with that in healthy skin ( Figure 1D). Considering UVB irradiation might induce MIF expression in the skin, we also stained for MIF in normal skin from different locations and analyzed MIF content [26]. As a result, no difference was observed in epidermal MIF content between sunexposed sites and sun-shielded sites in vitiligo lesional skin or normal skin (supplementary material, Figure S2A,B). ...
... In the present study, we found rising MIF levels in the early stages of vitiligo mouse model development, which indicated that MIF might begin to participate in vitiligo pathogenesis in the very early stages. In addition, a previous study reported that UVB irradiation might induce keratinocytes to overexpress MIF, meaning the epidermal MIF contents might be different between sun-exposed and sun-shielded skin [26]. But our results showed that there was no difference in either vitiliginous skin or normal skin. ...
Article
In vitiligo, autoreactive CD8+ T cell is established as the main culprit considering its pathogenic role in mediating epidermal melanocyte-specific destruction. Macrophage migration inhibitory factor (MIF) is a pleiotropic molecular that plays a central role in various immune processes including the activation and proliferation of T cells; but whether MIF is intertwined in vitiligo development and progression and its involvement in aberrantly activated CD8+ T cells remain ill-defined. In the study, we found that MIF was overabundant in vitiligo patients and a mouse model for human vitiligo. Additionally, inhibiting MIF might ameliorate the disease progression in vitiligo mice, which manifested as less infiltration of CD8+ T cells and more retention of epidermal melanocytes in the tail skin. More importantly, in vitro experiments indicated that MIF-inhibition suppressed the activation and proliferation of CD8+ T cells from lymph nodes of vitiligo mice, and the effect extended to CD8+ T cells in peripheral blood mononuclear cells of vitiligo patients. Finally, CD8+ T cells derived from MIF-inhibited vitiligo mice also exhibited an impaired capacity for activation and proliferation. Taken together, our results show that MIF is clinically targetable in vitiligo treatment, and the inhibition of which might ameliorate vitiligo progression through suppressing autoreactive CD8+ T cell activation and proliferation. This article is protected by copyright. All rights reserved.
... MIF plays a critical role in the pathogenesis of UVB-induced nonmelanoma skin cancer [15]. MIF overexpression has been implicated in chronic inflammatory diseases and malignancies [16]. MIF deficiency significantly reduces acute inflammatory responses in the skin following UVB exposure [17]. ...
Article
Full-text available
Background Chronic exposure to ultraviolet (UV) rays causes severe skin damage by inducing oxidative stress and inflammation. Identifying a safe and natural substance for skin protection is a crucial research goal. Objective The aim of this study was to clarify the effects of genistein on skin inflammation and photoaging by using 3 models (humans: skin parameters; animals: wrinkle formation; and cells: anti-inflammatory effects). Methods Food frequency questionnaire data and serum and skin parameter data from 120 volunteers (a group with a genistein-rich diet [RG group] and a control group). Human keratinocytes were pretreated with genistein before ultraviolet B (UVB) irradiation. Genistein was topically applied to the dorsal skin of rats. Results The blood samples of the RG group had lower serum uric acid levels and blood urea nitrogen levels. The dynamic elasticity level in the RG group was higher than that in the controls. Genistein pretreatment suppressed the expression of proinflammatory cytokines (CXCL1, IL-1, MIF, and PLANH1) and the proteins released by UVB-treated keratinocytes. Topical application of genistein to the dorsal skin of rats reduced the severity of UVB-induced wrinkling. Both intake and topical application of genistein combated UVB-induced inflammation and aging. Conclusions Genistein could be used as a safe and natural compound for use in novel anti-inflammatory agents for topical application. Graphical abstract The experimental design procedure, including the skin parameter and blood serum measurements of 137 participants. Genistein-rich compounds provide protection against UVB-induced inflammation, as determined using in vitro and in vivo animal model experiments.
... MIF is a pro-inflammatory cytokine and immunomodulator that is rapidly released from both immune and non-immune cells in response to various stimuli. Lipopolysaccharides (LPS) of Gram-negative bacteria (Roger, Glauser, & Calandra, 2001), exotoxins of Gram-positive bacteria (Calandra et al., 1998), tumor necrosis factor-α (TNF-α) (Lan et al., 1997), GC (Leech et al., 1999), angiotensin II (Rice et al., 2003), glucose (Toso, Emamaullee, Merani, & Shapiro, 2008), insulin (Waeber et al., 1997), phorbol myristate acetate (Rossi et al., 1998), and ultraviolet B irradiation (Shimizu, Abe, Ohkawara, & Nishihira, 1999) have all been reported to augment the MIF expression and secretion. Among these inducers LPS is a potent pro-inflammatory stimulus that upregulates MIF in a time and dose-dependent manner in various cell types. ...
... MIF was reported to be expressed in the basal layer of skin epidermis (Shimizu, Ohkawara, Nishihira, & Sakamoto, 1996) and keratinocytes (a source of cytokines under stress) and during inflammatory skin diseases (Yoshihisa, Rehman, Kondo, & Shimizu, 2016). MIF was observed to be expressed in the cytoplasm of human keratinocytes and epidermis and was shown to be secreted during immune reactions and inflammatory processes involving cutaneous immunity (Shimizu et al., 1999). In vivo studies by Shimizu et al. proved that MIF production was induced by various levels of ultraviolet B irradiation (0-125mJ per cm 2 ) in keratinocytes. ...
Article
Macrophage migration inhibitory factor (MIF) is a proinflammatory cytokine encoded within a functionally polymorphic genetic locus. MIF was initially recognized as a cytokine generated by activated T cells, but in recent days it has been identified as a multipotent key cytokine secreted by many other cell types involved in immune response and physiological processes. MIF is a highly conserved 12.5 kDa secretory protein that is involved in numerous biological processes. The expression and secretion profile of MIF suggests that MIF to be ubiquitously and constitutively expressed in almost all mammalian cells and is vital for numerous physiological processes. MIF is a critical upstream mediator of host innate and adaptive immunity and survival pathways resulting in the clearance of pathogens thus playing a protective role during infectious diseases. On the other hand, MIF being an immune modulator accelerates detrimental inflammation, promotes cancer metastasis and progression, thus worsening disease conditions. Several reports demonstrated that genetic and physiological factors, including MIF gene polymorphisms, posttranslational regulations, and receptor binding control the functional activities of MIF. Taking into consideration the multi-faceted role of MIF both in physiology and pathology, we thought it is timely to review and summarize the expressional and functional regulation of MIF, its functional mechanisms associated with its beneficial and pathological roles, and MIF-targeting therapies. Thus, our review will provide an overview on how MIF is regulated, its response, and the potency of the therapies that target MIF.
... Uncropped full-length blots are presented in Figure S9 following UVB exposure is similar to the pattern already reported for D-DT and MIF in human skin and highlighted the correlation of D-DT and UVB. 23,32 In the present study, D-DT Tg mice showed an earlier and higher incidence of tumors than WT mice with UVB exposure. D-DT Tg enhances UVB-induced DNA damage and induces the infiltration of inflammatory cells in the skin. ...
... The keratinocytes abundant in the outer layer of skin function as the initial responders to the UV-induced cutaneous inflammatory response by releasing a variety of cytokines, including MIF, IL-1, IL-6, and TNF-α. 23,[36][37][38] These cytokines not only mediate local inflammatory reactions but are known to induce skin carcinogenesis. TNFα-deficient models showed that TNF-α is required for the early stages of skin carcinogenesis and development of SCC. ...
Article
Full-text available
Ultraviolet irradiation (UV) exposure is the leading factor underlying the development of skin malignancies. D‐dopachrome tautomerase (D‐DT), a functional homolog of macrophage migration inhibitory factor (MIF), has functional similarities to MIF. However, its role, unlike the role of MIF in photocarcinogenesis, is unknown. We therefore explored the role of D‐DT in photocarcinogenesis by developing D‐DT transgenic (D‐DT Tg) mice and provided a research model for future studies targeting D‐DT. Chronic UVB exposure accelerated tumor development in D‐DT Tg mice compared with wild‐type (WT) mice, with a higher incidence of tumors observed in D‐DT Tg mice than in WT mice. In D‐DT Tg irradiated mouse keratinocytes, the p53, PUMA, and Bax expression was lower than that in WT mice. These results indicate that D‐DT Tg overexpression confers prevention against UVB‐induced apoptosis in keratinocytes. Taken together, these findings support D‐DT as a functionally important cytokine in photocarcinogenesis and potential therapeutic target for the prevention of photocarcinogenesis.
... DDT shares with MIF some catalytic and immune functions, such as counter-regulating glucocorticoid anti-inflammatory activity and triggering proinflammatory cascades by activating ERK-1/2 MAP kinases. DDT and MIF levels are increased in keratinocytes when exposed to UV rays, and at least the latter regulates melanogenesis [91][92][93]. DDT is also expressed constitutively, it is found at similar plasma levels to MIF in diseases such as ovarian cancer (~15 ng/mL) [9] and multiple sclerosis (~2 ng/mL) [94], and it is higher in critically ill patients (MIF = 133 vs. DDT = 292 ng/mL) [95]. This is remarkable considering that macrophages release 20 times more MIF than DDT upon LPS stimulation [9]. ...
Article
Human macrophage migration inhibition factor (MIF) is a protein with cytokine and chemokine properties that regulates a diverse range of physiological functions related to innate immunity and inflammation. Most research has focused on the role of MIF in different inflammatory diseases. D-dopachrome tautomerase (DDT), a different molecule with structural similarities to MIF, which shares receptors and biological functions, has recently been reported, but little is known about its roles and mechanisms. In this review, we sought to understand the similarities and differences between these molecules by summarizing what is known about their different structures, receptors and mechanisms regulating their expression and biological activities with an emphasis on immunological aspects.
... 12 MIF also regulates delayed-type hypersensitivity (IV), and when released from eosinophils can exacerbate human allergic and inflammatory disease such as asthma and AD. 13,14 It has been reported, 15 that MIF contributes to pollen-induced Accepted allergic conjunctivitis and dermatitis in mice by inducing eosinophil infiltration. Further evidence for a role of MIF in the pathogenesis of AD comes from murine models. ...
Article
Full-text available
Background Atopic dermatitis (AD) is a common inflammatory skin disease of dogs. Macrophage migration inhibitory factor (MIF) initiates pro‐inflammatory cytokine release in human AD and serum concentrations are correlated with disease severity. Hypothesis Canine serum MIF concentrations are increased in dogs with AD and correlate with clinical lesion and pruritus scores. Animals Client owned dogs (n = 49) diagnosed with AD and 17 healthy, unaffected control dogs were used for the study. Methods and materials A commercially available MIF ELISA was optimized for the dog and serum from clinical cases used. Information regarding treatment, Canine Atopic Dermatitis Extent and Severity Index, (CADESI‐4) and pruritus Visual Analog Scale (pVAS) were recorded for each dog at the time of serum collection. Results Dogs with AD which had not received steroid therapy and those treated with oclacitinib had significantly elevated serum MIF concentrations compared to controls. Concentrations of MIF were not significantly different in AD dogs receiving steroids compared to controls. There was no significant correlation between MIF concentrations and clinical scores (CADESI‐4 or pVAS). Conclusions and clinical importance Serum MIF concentrations are increased in dogs with AD and MIF might be a target for therapy.
... The epidermis is a known source of MIF production (30)(31)(32). Immunohistochemical detection of MIF in normal murine and human skin samples revealed that keratinocytes of the epidermis strongly express MIF in a constitutive fashion. MIF expression in both mice and humans was most prominent in the stratum basale and granulosum and became weaker with increasing keratinization of the cells in the stratum spinosum and corneum (Fig. 1A). ...
Article
The response of the skin to harmful environmental agents is shaped decisively by the status of the immune system. Keratinocytes constitutively express and secrete the chemokine-like mediator, macrophage migration inhibitory factor (MIF), more strongly than dermal fibroblasts, thereby creating a MIF gradient in skin. By using global and epidermis-restricted Mif knockout mice (Mif(-/-) and K14-Cre(+/tg); Mif(fl/fl)), we found that MIF both recruits and maintains antigen-presenting cells in the dermis/epidermis. The reduced presence of antigen-presenting cells in the absence of MIF was associated with accelerated and increased formation of nonmelanoma skin tumors during chemical carcinogenesis. Our results demonstrate that MIF is essential for maintaining innate immunity in skin. Loss of keratinocyte-derived MIF leads to a loss of control of epithelial skin tumor formation in chemical skin carcinogenesis, which highlights an unexpected tumor-suppressive activity of MIF in murine skin.-Brocks, T., Fedorchenko, O., Schliermann, N., Stein, A., Moll, U. M., Seegobin, S., Dewor, M., Hallek, M., Marquardt, Y., Fietkau, K., Heise, R., Huth, S., Pfister, H., Bernhagen, J., Bucala, R., Baron, J. M., Fingerle-Rowson, G. Macrophage migration inhibitory factor protects from nonmelanoma epidermal tumors by regulating the number of antigen-presenting cells in skin.
... MIF is expressed in the epidermis of the skin, particularly in the basal layer (24), and has a critical role in numerous inflammatory skin diseases (25,26). Skin keratinocytes are capable of producing a variety of cytokines and are thought to be the principal source of cytokines derived from the epidermis under stress conditions (27). Effects of MIF on UV-induced apoptosis have been well studied (28,29); however, its effects on HT-induced changes in the normal human skin cell have not yet been determined. ...
Article
In human skin, keratinocytes are constantly challenged by adverse influences, such as hot and cold temperatures; however, the effects of heat on apoptosis induction in keratinocytes are not well understood. Macrophage migration inhibitory factor (MIF) is a potent cytokine that overcomes p53 function by suppressing its transcriptional activity. Here, we evaluated the effects of MIF on hyperthermia (HT)-induced apoptosis in MIF-deficient [knockout (KO)] and MIF-transgenic (Tg) mouse keratinocytes. Cells were exposed to HT at 44°C, and increased apoptosis was observed in MIF KO and wild-type (WT) cells compared with MIF Tg cells. To determine the mechanism, MIF-mediated changes in the cellular p53 level and its effects on p53-dependent death signaling (Bax and p21) and JNK signaling (p-JNK, JNK, p-Bad, and Bad) was investigated. MIF Tg cells exhibited substantially decreased levels of p53 after HT treatment compared with WT and MIF KO cells. In addition, HT treatment caused decreased expression of p-JNK and p-Bad in MIF Tg cells; however, no such changes were observed in MIF KO and WT cells. These results showed that the activation of JNK (p-JNK and p-Bad) and p53 may be involved in HT-induced apoptosis in keratinocytes and that enhanced endogenous MIF expression suppressed apoptosis.-Yoshihisa, Y., Rehman, M. U., Kondo, T., Shimizu, T. Role of macrophage migration inhibitory factor in heat-induced apoptosis in keratinocytes.
... Several cytokines or growth factors play important roles during tissue repair and promote wound healing (Sporn and Roberts, 1986). Shimizu found that keratinocytes migrated from the wound edges and their proliferation was essential for proper dermal wound repair (Shimizu et al., 1999). For the overall wound healing process, many cytokines and inflammatory mediators released from the keratinocytes influence events by regulating the growth, differentiation and metabolism of the other cells that are associated with the injured skin lesion (Sporn and Roberts, 1986). ...
Article
Full-text available
Estrogens play a crucial role in cutaneous wound healing by down-regulating macrophage migration inhibitory factor (MIF). We had previously reported the effect of young coconut juice (YCJ) known to contain the phytoestrogen, -sitosterol, on cutaneous wound healing in ovariectomized (ovx) rats. This research investigated the possible mechanisms of YCJ on cutaneous wound healing and it was found that it down regulated macrophage migration inhibitory factor (MIF). This resulted in ultrastructural changes that were observed using transmission electron microscopy (TEM). Four groups of female rats (6 in each group) were included in this study: sham-operated, ovariectomized (ovx), ovx that received estradiol benzoate (EB) injections intraperitoneally, and ovx that received YCJ orally. Two weeks after ovariectomy, two equidistant 1-cm full-thickness skin incisional wounds were made. At the end of the third week (7 day treatment) and the fourth week (14 day treatment) of study the rats were sacrificed, and their serum estradiol (E2) levels were measured by a chemiluminescent immunoassay. The skin from the wound was excised and examined by TEM and MIF immunohistochemical staining. The TEM study after 14 days of treatment showed that the size of the keratinocyte cells from the ovx+YCJ group was larger and these cells contained many more cytoplasmic processes than those of the ovx group. The MIF immunoreactivity was also lowest in the ovx+YCJ group. This study showed that there was an increased intercellular exchange via the cytoplasmic processes of the keratinocytes that could account for the promotion of cutaneous wound healing in the ovx rats receiving YCJ, and that the possible mechanism for this was via the down-regulation of MIF.