Summary
Global warming and rising temperature significantly increase the incidence of heat stress, which is known to affect the process of inflammation and aging. However, the effect of heat stress on skin melanogenesis is not fully known. We found that healthy foreskin tissues underwent significant pigmentation when exposed to 41°C. Furthermore, heat stress promoted melanogenesis in pigment cells by increasing the paracrine effects of keratinocytes. High-throughput RNA sequencing showed that heat stress activates the Hedgehog (Hh) signaling pathway in keratinocytes. The agonists of Hh signaling promote the paracrine effect of keratinocytes on melanogenesis. In addition, transient receptor potential vanilloid (TRPV) 3 agonists activate the Hh signaling in keratinocytes and augment its paracrine effect on melanogenesis. The heat-induced activation of Hh signaling is dependent on TRPV3-mediated Ca2+ influx. Heat exposure promotes melanogenesis by increasing the paracrine effects in keratinocytes via the TRPV3/Ca2+/Hh signaling pathway. Our findings provide insights into the mechanisms of heat-induced skin pigmentation.
Subject areas: Biological sciences, Molecular biology, Cell biology
Graphical abstract
Highlights
-
•
Heat promoted melanogenesis by increasing the paracrine effects in keratinocytes
-
•
Heat increased the paracrine effects in keratinocytes via the Hh signaling pathway
-
•
Heat-induced TRPV3-mediated Ca2+ influx activated the Hh signaling pathway
Biological sciences; Molecular biology; Cell biology
Introduction
Global warming has increased the risk of heat stress and related disorders, such as maternal health and neonatal outcomes,1 as well as cardiovascular and respiratory complications.2 Heat stress can also cause skin aging by damaging the DNA directly.3,4,5 Occupational exposure to high temperatures among soldiers, builders, and boiler workers is associated with considerable skin damage. Long-term skin exposure to direct heat leads to reticular pigmentation and telangiectasia formation, also known as erythema ab igne (EAI).6 However, so far, only a little is known about the actual effect of heat exposure on skin pigmentation and its underlying mechanism.
The Hedgehog (Hh) signaling pathway is an evolutionarily conserved pathway that plays a significant role in the normal embryonic development of invertebrates and vertebrates.7 Studies show that external stimuli like ultraviolet radiation b (UVB) exposure can activate the Hh signaling pathway8 and its key regulatory molecules including patched 1 (PTCH1), patched 2 (PTCH2), and Glioma-associated oncogene homologue 1 (GLI1).9 The upregulation of these genes is indicative of Hh pathway activation. The target genes of this pathway include vascular endothelial growth factor A (VEGFA),10 B-cell lymphoma 2 (BCL2),11 BHLH transcription factor (MYCN),12 members of the Wnt signaling pathway,13 etc.
The epidermal melanin unit (EMU) plays an important role in skin pigmentation.14 Melanocytes synthesize melanin, and so formed melanin gets transferred to the neighboring keratinocytes to protect the skin from ultraviolet (UV) damage.15 At the same time, keratinocytes affect melanin production by secreting paracrine factors in response to external stimuli, such as UV irradiation, inflammation, or drugs.16 Recent studies have suggested that paracrine factors, such as prostaglandin-endoperoxide synthase 2 (PTGS2) and vascular endothelial growth factor (VEGF), regulate melanin synthesis.17,18 The Wnt signaling pathway also plays an important role in melanogenesis and melanocyte development.19,20 The pathway is activated once the Wnt protein binds to its receptor, and it has crucial ligands named WNT3 and WNT7A.21
The exact pathogenesis of skin hyperpigmentation is unclear, although excessive UV radiation, exposure to particulate matter (PM) 2.5, and drugs have been implicated.22,23,24,25 By now, few studies have reported the effect of heat exposure on melanogenesis.26 In this study, we investigated the effects of heat exposure on skin pigmentation using in vitro and in vivo models and found that heat promotes melanogenesis by increasing the paracrine effects in keratinocytes via the transient receptor potential vanilloid (TRPV) 3/Ca2+/Hh signaling pathway.
Results
Heat exposure increased epidermal pigmentation in human skin
EAI is primarily induced by heat and manifests as local telangiectasia reticular erythema and pigmentation. Hematoxylin and eosin (H&E) staining and Fontana-Masson staining of human skin specimens confirmed the presence of more melanin particles in the basal layer of EAI skin than in normal skin (Figures 1A and 1B). To further establish the effects of heat on skin pigmentation, human skin tissues were exposed to heat for 5 days and stained as above. As shown in Figures 1C and 1D, heat exposure significantly increased the number of melanin granules.
Conditioned media from heat-treated HaCaT cells and keratinocytes promoted melanogenesis in pigment cells
The MNT1 and HaCaT cells were cultured at 37°C, 39°C, 41°C, 42°C, and 43°C for 24, 48, and 72 h, and the viability rates were evaluated by the Cell Counting Kit-8 (CCK-8) assay. As shown in Figures 2A and 2B, the viability of cells was unaffected at 37°C, 39°C, and 41°C. Therefore, we exposed the cells to these temperatures for 1 h daily over 3 consecutive days. Heat exposure significantly increased the melanin content (Figure 2C) as well as the expression of melanogenesis-related genes (Figure 2D) in the MNT1 cells, which was consistent with a previous report.27 The MNT1 and HaCaT cells were then co-cultured in 1:10 ratio and exposed to heat for 3 days. As shown in Figures 2E and 2F, heat exposure increased the melanin content and the expression of melanogenesis-related genes in the co-cultured cells. Consistent with this, MNT1 cells cultured in the conditioned medium of heat-treated HaCaT cells also exhibited higher melanin content and expression levels of melanogenesis-related genes compared to the 37°C-control group (Figures 2G–2I), and the increased melanogenesis was associated with the activation of the mitogen-activated protein kinase (MAPK) and Wnt/β-catenin pathways (Figures 2I and S1G). In addition, the conditioned medium of heat-exposed HaCaT cells also increased the number of Pmel17-expressing melanosomes along the dendrites of MNT1 cells (Figure 2J). On the other hand, we further validated our findings with primary cells. Human epidemal melanocytes (HEMs) cultured in the conditioned medium of heat-treated primary keratinocytes also exhibited higher melanin content (Figure S2A). Taken together, the conditioned medium of heat-treated HaCaT cells and keratinocytes significantly increased the melanin content and upregulated the melanogenesis-related genes of the pigment cells.
Heat-induced paracrine effects in HaCaT cells and keratinocytes were mediated via the Hh signaling pathway
To further explore the paracrine effects of heat on the HaCaT cells and keratinocytes, we exposed the cells to high temperatures for 3 days. We analyzed the changes in gene expression levels. Heat exposure significantly increased the expression of VEGF, PTGS2, WNT3, and WNT7A in HaCaT cells (Figure 3A) and keratinocytes (Figure S2B). The expression levels of other paracrine factors are shown in Figure S1A. Furthermore, high-throughput RNA sequencing revealed differentially expressed genes (DEGs) between the control and heat-treated cells (Figure 3B). The principal-component analysis (PCA) map of the genes is shown in Figure 3C. At the same time, gene set enrichment analysis (GSEA)-kyoto encyclopedia of genes and genomes (KEGG) revealed significant enrichment of the Hh, Wnt, and estrogen signaling pathways among others (Figure 3E). In addition, there was a significant positive correlation between heat exposure and the Hh, Wnt, estrogen, and GnRH signaling pathways (Figure 3F). Furthermore, we treated the HaCaT cells with the agonists of these signaling pathways (SAG-2.5 μM, CT99021-5 μM, DHEA-10 μM, and Elagolix Sodium-10 μM, respectively) for 24h and found that only the activation of the Hh signaling pathway significantly upregulated VEGF, PTGS2, WNT3, and WNT7A (Figure 3G). Inhibition of the Hh signaling pathway downregulated VEGF, PTGS2, WNT3, and WNT7A mRNA levels in HaCaT cells (Figure S1D). In contrast, the Hh pathway did not affect the fibroblast growth factor 2 (FGF2) levels (Figure S1B), indicating that other signaling pathways may activate it. The MNT1 cells were cultured in the conditioned medium of HaCaT cells supplemented with SAG (2.5 μM). As shown in Figure 3H, the melanin levels were significantly higher in the treated versus the untreated MNT1 cells. In addition, PTCH1 and GLI1 were significantly upregulated in the HaCaT cells and keratinocytes after heat treatment (Figures 3I and S2D). It indicates that heat exposure regulates the Hh signaling pathway in keratinocytes.
Heat-induced TRPV3-mediated Ca2+ influx activated the Hh signaling pathway in HaCaT cells and keratinocytes
Transient receptor potential vanilloid (TRPV) plays a key role in relaying heat or pain sensations. Different TRPV channels have different sensitivity to temperature. Our study found that heat exposure upregulated TRPV3 level in HaCaT cells (Figure 4A) and keratinocytes (Figure S2C). We also tested the influence of heat on other TRPV channels such as TRPV1, 2, and 4. As shown in Figure S1F, the expression level of TRPV1, 2, and 4 did not significantly changed after heat exposure. Furthermore, activation of TRPV3 using its specific agonist (camphor, 50 μM) led to increased Ca2+ influx into the HaCaT cells (Figure 4B). Likewise, heat exposure also increased intracellular calcium levels in the HaCaT cells, and supplementation of TRPV3 inhibitors (TRPV3 antagonist 74a, 100 μM) was able to reverse the increased Ca2+ influx in response to heat (Figure 4C). We further explored the role of other TRPV channels in heat-induced calcium influx. TRPV1 agonists (vanilloid, 25 μM) or TRPV4 agonists (GSK1016790A, 50 nM) induce increased Ca2+ influx into HaCaT cells (Figure S3A). However, TRPV1 antagonists (AMG-517, 0.25 nM) or TRPV4 antagonists (GSK2193874, 50 nM) could not reverse the increased Ca2+ influx induced by heat exposure (Figure S3B). These suggested that TRPV1 and TRPV4 might not play critical roles in heat-induced calcium influx in cells. The conditioned medium of the camphor-treated HaCaT cells also increased the melanin content in MNT1 cells (Figure 4D). The proteins of the Hh signaling pathway were also upregulated in the camphor-treated HaCaT cells (Figure 4E). Furthermore, heat failed to activate the Hh pathway effectively after a TRPV3 inhibitor was used in HaCaT cells (Figure S1E). Inhibition of the Hh signaling pathway did not affect heat-induced upregulation of TRPV3, which suggested TRPV3 might be upstream of Hh signaling pathway (Figure S1C). Taken together, the TRPV3/Ca2+/Hh signaling pathway is most likely involved in heat-induced melanogenesis.
Heat exposure activated the Hh signaling pathway via TRPV3 in human skin tissue
To further determine the biological relevance of the Hh signaling pathway, we exposed human skin pieces to heat for 5 days and analyzed the in situ expression of the pathway proteins. As shown in Figure S4, PTCH1 and GLI1 were both highly expressed in the heat-treated human skin. TRPV3 agonists and inhibitors were added to test whether heat activated the Hh pathway in human skin tissue through TRPV3. Human skin pieces were exposed to heat for 5 days after the addition of TRPV3 agonists (camphor, 50 μM) or inhibitors (TRPV3 antagonist 74a, 100 μM). We found that the TRPV3 agonists increased heat-induced pigmentation and the TRPV3 inhibitors decreased it (Figure 5A). Furthermore, the expressions of PTCH1 and GLI1 were higher after treatment of TRPV3 agonists, while those were lower after treatment of TRPV3 inhibitors (Figure 5B). The above results indicated that heat activated the Hh signaling pathway via TRPV3.
Discussion
This study shows that heat exposure induces paracrine effects in keratinocytes through the TRPV3/Ca2+/Hh signaling pathway, which promotes melanogenesis in melanocytes (Figure 6).
Previous studies have shown that the biological effects of heat and UVB on melanocytes are similar.27 Furthermore, heat exposure can even augment UVB-induced tyrosinase activation and melanogenesis.26 However, the specific mechanism underlying heat-induced melanogenesis has yet to be entirely understood. We found that heat directly affects melanogenesis, which is consistent with previous studies.27 The melanocytes and keratinocytes in the epidermis share a complex relationship. Keratinocytes secrete multiple paracrine factors that affect melanin production. Therefore, we evaluated these paracrine factors’ role in heat-induced melanogenesis and found that heat exposure increased the melanin content and the expression of melanogenesis-related genes in co-cultured MNT1 and HaCaT cells. Furthermore, the conditioned medium of the heat-treated HaCaT cells and keratinocytes showed a similar melanogenic effect on pigment cells. In this study, heat exposure significantly increased the expression of VEGF, PTGS2, WNT3, and WNT7A in HaCaT cells and keratinocytes. These paracrine factors have been reported to be involved in regulating melanin synthesis. VEGF interacts with VEGF receptors (VEGFRs) and promotes melanogenesis by activating MAPK signaling pathway.28 PTGS2 plays an important role in prostaglandin E2 (PGE2) production, which promotes melanogenesis by interacting with the EP receptor and activating the cyclic AMP (cAMP) signaling pathway in epidermal melanocytes.29,30 WNT3 and WNT7A might act as ligands in activating the Wnt/β-catenin pathways by binding to its receptors and promoting the expression of melanogenesis-related genes.31 Altogether, heat might promote melanogenesis by augmenting the paracrine effects of keratinocytes.
Nevertheless, the heat-induced paracrine effects of keratinocytes remain unclear. The Hh signaling pathway mediates thermal allodynia and hyperalgesia following skin damage due to UV irradiation.32 High-throughput RNA sequencing revealed a significant positive correlation between heat exposure and the Hh signaling pathway, indicating that heat can activate this pathway. Furthermore, activation of the Hh signaling pathway upregulated VEGF, PTGS2, WNT3, and WNT7A in the HaCaT cells. In addition, the conditioned medium of HaCaT cells treated with an Hh pathway agonist increased the melanin content of the MNT1 cells. These results suggest that heat-induced paracrine effects in keratinocytes are most likely mediated via the Hh signaling pathway.
Transient receptor potential (TRP) channels act as polymodal sensors that relay various chemical and physical stimuli and induce cellular and physiological responses.33 TRPV3 is a member of the vanilloid subfamily or TRPV channels and is highly expressed in skin keratinocytes where it forms Ca2+-permeable non-selective cation channels to regulate various cutaneous functions.34,35,36,37,38,39,40 TRPV3 plays an important role in many cutaneous sensations, including nociception, thermal sensing, and itching.36,38 Keratinocytes also express other TRPV channels, such as TRPV1 and TRPV4.41,42,43 TRPV1, a heat-gated channel, was found in human keratinocytes recently. Furthermore, the activation of epidermal TRPV1 was known to induce the release of proinflammatory mediators.44 TRPV4 channel is a physiological sensor for hypoosmolarity, mechanical deformation, and warm temperature.45 TRPV1 is activated by noxious heat (>43°C), while TRPV3 is activated at the warm temperature threshold of 33°C and exhibits increasing responses at higher noxious temperatures. TRPV4, identified originally as an osmosensory ion channel, is also activated by warm temperatures (25–34°C). Although both TRPV3 and TRPV4 are expressed in keratinocytes and are activated by a similar range of temperatures, these channels likely have distinct functions in the skin.46 Our study found that heat-induced TRPV3 can effectively induce Ca2+ influx and activate the Hh signaling pathway. The expression level of TRPV1 and 4 did not significantly change after heat exposure (39–41°C), and TRPV1 antagonists or TRPV4 antagonists could not reverse the increased Ca2+ influx induced by heat exposure (39–41°C). These results suggest that TRPV3 rather than TRPV1 or 4 may play a key role in heat-induced (39–41°C) calcium influx in keratinocytes. Furthermore, the conditioned medium of HaCaT cells treated with the TRPV3 agonist increased the melanin content in MNT1 cells. Thus, heat-induced TRPV3 may play a role in melanogenesis by activating the Hh signaling pathway.
Long-term or repeated exposure of skin to heat deepens erythema, resulting in reticular telangiectasia, skin atrophy, pigmentation, and diffuse hyperkeratosis, which are characteristic of EAI and similar to skin heterochromism.47 However, the exact pathological basis of EAI remains unclear. Our findings show that heat promotes melanogenesis, which may explain the pigmentation in EAI skin lesions. In addition, heat exposure also increased the expression of VEGF, a growth factor involved in normal and pathological angiogenesis, in the HaCaT cells.48 Therefore, heat-induced VEGF may alter the development of blood vessels in EAI. Overall, heat exposure (39–41°C) triggers melanogenesis by increasing the paracrine effects in keratinocytes via the TRPV3/Ca2+/Hh signaling pathway. Our study provides insights into the mechanisms of heat-induced skin diseases such as EAI and hints toward pathways that could be therapeutic targets.
Limitations of the study
The effect of heat on melanogenesis has yet to be verified in animal models, and experiments on cell culture and skin explants might differ from the actual physiological state. It would be more convincing to test whether Hh signaling is activated by heat in TRPV3 knockout mice. In addition, the effectors involved in the Hh signaling pathway during heat-induced melanogenesis need further investigation, and further experiments are required.
STAR★Methods
Key resources table
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
Rabbit monoclonal anti-TYR | ZENBIO | Cat#121392; SwissProtID:P14679 |
Rabbit monoclonal anti-MITF | ZENBIO | Cat#R24980; SwissProtID:O75030 |
Rabbit monoclonal anti-TYRP1 | ZENBIO | Cat#382326; SwissProtID:P17643 |
Rabbit monoclonal anti-DCT | ZENBIO | Cat#821374; SwissProtID:P40126 |
Rabbit monoclonal anti-RAB27A | Cell Signaling Technology | Cat#95394S; RRID: AB_2800247 |
Rabbit monoclonal anti-ERK | Cell Signaling Technology | Cat#4695S; RRID: AB_390779 |
Rabbit monoclonal anti- p-ERK | Cell Signaling Technology | Cat#4370S; RRID: AB_2315112 |
Rabbit monoclonal anti-JUK | Cell Signaling Technology | Cat#9252S; RRID: AB_2250373 |
Rabbit monoclonal anti-p-JUK | Cell Signaling Technology | Cat#9255S; RRID: AB_2307321 |
Rabbit monoclonal anti-p38 | Cell Signaling Technology | Cat#8690S; RRID: AB_10999090 |
Rabbit monoclonal anti-p-p38 | Cell Signaling Technology | Cat#4511S; RRID: AB_2139682 |
Rabbit monoclonal anti-β-catenin | ZENBIO | Cat#R22820; SwissProt ID: P35222 |
Rabbit monoclonal anti-GLI1 | ABclonal | Cat#A14675; RRID: AB_2761550 |
Rabbit monoclonal anti-PTCH1 | ABclonal | Cat#A0826; RRID: AB_2757415 |
Rabbit monoclonal anti-GAPDH | Cell Signaling Technology | Cat#5174S; RRID: AB_10622025 |
Mouse monoclonal anti-Pmel17 | Santa Cruz Biotechnology | Cat#sc-377325; RRID: AB_2889982 |
Goat Anti-mouse IgG(H+L) Fluor 594 | Affinity | Cat#S0005; RRID: AB_2843435 |
Chemicals, peptides, and recombinant proteins | ||
CT99021 | Selleck | S1263; CAS:252917-06-9 |
Elagolix Sodium | Selleck | S4896; CAS:832720-36-2 |
Smoothened Agonist (SAG) HCl | Selleck | S7779; CAS:2095432-58-7 |
Dehydroepiandrosterone(DHEA) | Selleck | S5508; CAS:853-23-6 |
Vismodegib | Selleck | S1082; CAS:879085-55-9 |
TRPV3 antagonist 74a | MedChemExpress | HY-131868; CAS:1432051-63-2 |
Camphor | Selleck | S3851; CAS: 76-22-2 |
GSK1016790A | Selleck | S8107; CAS:942206-85-1 |
GSK2193874 | Selleck | S8367; CAS:1336960-13-4 |
AMG-517 | Selleck | S7115; CAS:659730-32-2 |
Capsaicin(Vanilloid) | Selleck | S1990; CAS:404-86-4 |
Deposited data | ||
RNA-seq | This paper | GEO accession numbers: GSE229915 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE229915) |
Critical commercial assays | ||
Masson-Fontana | G-CLONE | 400-910-1997; Cat.No.RS8020 |
qPCR SYBR Green Master Mix | YEASEN | LOT:H8210010; Cat:11184ES08 |
HiScript II Q RT SuperMix for qPCR | Vazyme | R223-01 |
Fluo-4 AM | Beyotime | S1060 |
Experimental models: Cell lines | ||
MNT1 | Meisen CTCC | CTCC-006-0301 |
HaCaT | Otwo Biotech (ShenZhen) Inc. | HTX2089 |
Melanocytes | N/A | N/A |
Keratinocytes | N/A | N/A |
Oligonucleotides | ||
See the Table S1 | This paper | N/A |
Software and algorithms | ||
Image J | Image J software | https://imagej.nih.gov/ij/ |
Adobe Illustrator 2021 | N/A | https://www.adobe.com/products/illustrator.html |
CaseViewer 2.4(64-bit version) | The Digital Pathology Company | https://www.3dhistech.com/solutions/caseviewer/ |
GraphPad Prism 9.0 | GraphPad | https://www.graphpad.com/ |
Resource availability
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Qinghai Zeng (zengqinghai@csu.edu.cn).
Materials availability
This study did not generate new unique reagents.
Experimental model and subject details
Cell lines
The human melanoma cell line MNT1 cells were obtained and authenticated from the Meisen CTCC (Zhejiang Meisen Cell Technology Co., LTD). Human immortalized keratinocyte line HaCaT cells were obtained and and authenticated from Otwo Biotech (ShenZhen) Inc. MNT1 and HaCaT cells were cultured in DMEM (#C11995500BT, Gibco, NY) supplemented with 20% or 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin (#ABT920, G-CLONE). Primary human epidermal melanocytes (HEMs) and keratinocytes were separated from male volunteers’ foreskin (approved by donors and the Ethics Committee of the Third Xiangya Hospital, Central South University, Changsha, China). HEMs were cultured in 254 medium (M254500, Gibco) with 1% HMGS (M254500, Gibco) and 5% FBS, and keratinocytes were cultured in serum-free KGM. None of the cell lines were detected to be contaminated with mycoplasma. All cells were cultured in a humidified incubator at 37°C and 5% CO2.
Method details
Cell viability assay
Cell viability was tested by the Cell Counting Kit-8 (CCK-8) and was done according to the manufacturer’s instructions (#BS350A, Biosharp). Briefly, the cells were seeded in 96-well plates at the density of 2000 cells/well, or 5000 cells/well, and incubated at different temperatures in a temperature-regulated incubator (#HN-25BS, LICHEN). After 24h, 48h, and 72h of culture, 10μL CCK-8 reagent was added to each well, and the cells were incubated at 37°C for 1 hour. When the color of the medium turned orange, the reaction was terminated, and the absorbance was measured at 490 nm using a microplate reader (PerkinElmer EnVision Xcite, UK).
Masson-Fontana melanin staining
The adherent cultured cells were fixed with 4% paraformaldehyde for 15 min. After rinsing with water, the cells were stained with the Fontana ammonia-silver solution (#No.RS8020, G-GLONE) for 24 h in a dark chamber, rinsed again, and immersed in hyposulphite solution for 5 min. The slides were observed under an inverted microscope, and the melanin granules were counted.
Western blotting
The total cellular protein was extracted using RIPA Lysis Buffer (#No. EX6020, G-GLONE) supplemented with a protease inhibitor and phosphatase inhibitor cocktail (Roche) and quantified using a BCA protein assay kit (# KGPBCA, KeyGEN Biotec). After blocking with 1% BSA (#4240GR005, BioFroxx), the blots were incubated overnight with primary antibodies against TYR (#121392, ZENBIO), MITF (#R24980, ZENBIO), TYRP1 (#382326, ZENBIO), DCT (#821374, ZENBIO), RAB27A (#95394S, CST), ERK (#4695S, CST), p-ERK (#4370S, CST), JUK (#9252S, CST), p-JUK (#9255S, CST), p38 (#8690S, CST), p-p38 (#4511S, CST), β-catenin (#R22820, ZENBIO), GLI1 (#A14675, ABclonal), PTCH1 (#A0826, ABclonal) and GAPDH (#5174S, CST) at 4°C. All antibodies were diluted to1:1000, and the anti-GAPDH antibody was diluted to1:2000 with TBS-T buffer. The membranes were washed with TBS-T buffer and incubated with a goat anti-rabbit secondary antibody (1:10000; #56j9958, Affinity) for one hour. The positive bands were detected by enhanced chemiluminescence (ECL) using the ECL kit (biosharp), per the manufacturer's instructions.
Immunofluorescence
The adherent cells were fixed with 4% paraformaldehyde and incubated overnight with the anti-PMEL17 antibody (#sc-377325, SCBT), followed by fluorescent secondary antibodies (#S0005, Affinity). After counterstaining with DAPI (#BS097, Biosharp), the cells were observed under a confocal fluorescence microscope (LSM800; Zeiss, Oberkochin, Germany). The melanosomes with emitting red fluorescence were counted.
Quantitative reverse transcription-polymerase chain reaction (qRT-PCR)
The total cellular RNA was extracted using Fast total RNA extraction kit (FASTAGEN, Shanghai, China), and reverse-transcribed into cDNA using a reverse transcription kit (#R223-01, Vazyme), as per manufacturer’s instructions. The SYBR green qPCR Mix (#11184ES08, YEASEN) was used, and the reaction was performed on a real-time PCR instrument (Roche Light Cycler 480II, Basel, Switzerland). GAPDH was used as the internal control. The relative expression levels of MITF, TYR, TYRP1, DCT, RAB27A, MYO5A, VEGF, PTGS2, FGF2, ET-1, WNT3, WNT7A, TRPV1, TRPV2, TRPV3, and TRPV4, were calculated according to the ΔCt formula and normalized against GAPDH housekeeping gene. The primer sequences are listed in Table S1.
Human skin samples
Approved by the Ethics Committee of the Third Xiangya Hospital of Central South University and with the consent of the donors, skin tissue samples were collected from male adolescent circumcision in the Department of urology in the Third Xiangya Hospital of Central South University. In addition, the Erythema Ab Igne (EAI) specimens were donated by patients of the dermatology department of the Third Xiangya Hospital of Central South University. The foreskins were cut into pieces and carefully placed in DMEM (#C11995500BT, Gibco, NY) supplemented with 20% or 10% FBS (Biological Industries, Israel) and 1% penicillin-streptomycin (#ABT920, G-CLONE) antibiotic mixture. The skin pieces were allowed to float on the medium, with the epidermis facing up at the air/liquid interface and the dermis/subcutis facing down. The tissues were cultured at 37°C in a humidified CO2 incubator containing 5% CO2 and kept for 5 days, and exposed to different temperatures (37°C, 39°C and 41°C) every day for 1 hour.
Immunohistochemistry
The paraffin sections were deparaffinized, cleared with xylene, and hydrated through an ethanol gradient. After washing once with (Phosphate buffered saline) PBS, the slides were immersed in hot sodium citrate solution to retrieve antigens. The slides were then rinsed once with PBS, incubated with an endogenous peroxidase blocking agent at room temperature for 10 min, and washed thrice with PBS. The sections were then blocked with 5% BSA for 30 min and incubated overnight with anti-PTCH1 (1:100; #A0826, ABclonal) and anti-GLI1 (1:150; #A14675, ABclonal) antibodies at 4°C. The following day, the slides were washed with PBS and then incubated with 100μL of a reaction enhancement solution at room temperature for 20 min. The slides were then washed thrice with PBS, incubated with HRP-labeled goat anti-rabbit IgG antibody at 37°C for 30 min, washed thrice with PBS, developed with 3,3'-diaminobenzidine (DAB) substrate, and counterstained with hematoxylin. After dehydrating with an ethanol gradient and clarification with xylene, the specimens were sealed with a neutral resin and observed under an inverted microscope.
Cytoplasmic Ca2+ measurement
The suitably treated cells were incubated with 2μM Fluo-4-AM (#S1060, Beyotime) for 40 min. The reaction mixture was diluted by adding PBS, and the cells were left undisturbed for 25 min. The intensity of green fluorescence corresponding to cytoplasmic Ca2+ level was determined by confocal microscopy (LSM800; Zeiss, Oberkochin, Germany).
RNA high-throughput sequencing
RNA was extracted from HaCaT cells and cultured in the medium at 41°C for 3 days, and RNA sequencing was performed using the Illumina platform per the manufacturer's instructions. Three biological replicates were analyzed for each group.
Quantification and statistical analysis
All values are presented as mean ± SD and data were obtained from three independent experiments. Statistical analyses were performed on individual experiments, as indicated, with GraphPad Prism 9.0 software using an unpaired t-test, equal variance for comparison between two groups and one-way ANOVA for comparisons between more than two groups. A P value of ∗P<0.05 was considered as statistically significant.
Acknowledgments
This study was supported by financial grants from the National Natural Science Foundation of China (82073420), the Natural Science Foundation of Hunan Province (2021JJ20089), the Wisdom Accumulation and Talent Cultivation Project of the Third Xiangya Hospital of Central South University (YX202007), and the Science and Technology Innovation Program of Hunan Province (2021RC3035).
Author contributions
Conceived and designed the experiments and analyzed data: Q.Z., J.C., J.H., and H.Z.; Performed most of the experiments: L.Z., Y.Z., X.Z., and F.Z.; Performed bioinformatics analysis: C.F., L.J., Y.H., and L.Z.; Wrote the manuscript: L.Z. ; All authors contributed to and approved the paper.
Declaration of interests
The authors declare that they have no competing interests.
Inclusion and diversity
We support inclusive, diverse, and equitable conduct of research. We worked to ensure diversity in experimental samples through the selection of the cell lines.
Published: April 26, 2023
Footnotes
Supplemental information can be found online at https://doi.org/10.1016/j.isci.2023.106749.
Supplemental information
Data and code availability
-
•
RNA-seq data have been deposited at NCBI and are publicly available as of the date of publication. Accession numbers are listed in the key resources table.
-
•
This paper does not report original code.
-
•
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.
References
- 1.Yardman-Frank J.M., Fisher D.E. Skin pigmentation and its control: from ultraviolet radiation to stem cells. Exp. Dermatol. 2021;30:560–571. doi: 10.1111/exd.14260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.The L. Heatwaves and health. Lancet. 2018;392:359. doi: 10.1016/S0140-6736(18)30434-3. [DOI] [PubMed] [Google Scholar]
- 3.Dewey W.C., Hopwood L.E., Sapareto S.A., Gerweck L.E. Cellular responses to combinations of hyperthermia and radiation. Radiology. 1977;123:463–474. doi: 10.1148/123.2.463. [DOI] [PubMed] [Google Scholar]
- 4.Schieke S.M., Schroeder P., Krutmann J. Cutaneous effects of infrared radiation: from clinical observations to molecular response mechanisms. Photodermatol. Photoimmunol. Photomed. 2003;19:228–234. doi: 10.1034/j.1600-0781.2003.00054.x. [DOI] [PubMed] [Google Scholar]
- 5.Schroeder P., Haendeler J., Krutmann J. The role of near infrared radiation in photoaging of the skin. Exp. Gerontol. 2008;43:629–632. doi: 10.1016/j.exger.2008.04.010. [DOI] [PubMed] [Google Scholar]
- 6.Knöpfel N., Weibel L. Erythema Ab Igne. JAMA Dermatol. 2021;157:106. doi: 10.1001/jamadermatol.2020.3995. [DOI] [PubMed] [Google Scholar]
- 7.Varjosalo M., Taipale J. Hedgehog: functions and mechanisms. Genes Dev. 2008;22:2454–2472. doi: 10.1101/gad.1693608. [DOI] [PubMed] [Google Scholar]
- 8.Kim W., Kim E., Yang H.J., Kwon T., Han S., Lee S., Youn H., Jung Y., Kang C., Youn B. Inhibition of hedgehog signalling attenuates UVB-induced skin photoageing. Exp. Dermatol. 2015;24:611–617. doi: 10.1111/exd.12735. [DOI] [PubMed] [Google Scholar]
- 9.Bonifas J.M., Pennypacker S., Chuang P.T., McMahon A.P., Williams M., Rosenthal A., De Sauvage F.J., Epstein E.H., Jr. Activation of expression of hedgehog target genes in basal cell carcinomas. J. Invest. Dermatol. 2001;116:739–742. doi: 10.1046/j.1523-1747.2001.01315.x. [DOI] [PubMed] [Google Scholar]
- 10.Morrow D., Cullen J.P., Liu W., Guha S., Sweeney C., Birney Y.A., Collins N., Walls D., Redmond E.M., Cahill P.A. Sonic Hedgehog induces Notch target gene expression in vascular smooth muscle cells via VEGF-A. Arterioscler. Thromb. Vasc. Biol. 2009;29:1112–1118. doi: 10.1161/ATVBAHA.109.186890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bigelow R.L.H., Chari N.S., Unden A.B., Spurgers K.B., Lee S., Roop D.R., Toftgard R., McDonnell T.J. Transcriptional regulation of bcl-2 mediated by the sonic hedgehog signaling pathway through gli-1. J. Biol. Chem. 2004;279:1197–1205. doi: 10.1074/jbc.M310589200. [DOI] [PubMed] [Google Scholar]
- 12.Oliver T.G., Grasfeder L.L., Carroll A.L., Kaiser C., Gillingham C.L., Lin S.M., Wickramasinghe R., Scott M.P., Wechsler-Reya R.J. Transcriptional profiling of the Sonic hedgehog response: a critical role for N-myc in proliferation of neuronal precursors. Proc. Natl. Acad. Sci. USA. 2003;100:7331–7336. doi: 10.1073/pnas.0832317100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Katoh M., Katoh M. Notch ligand, JAG1, is evolutionarily conserved target of canonical WNT signaling pathway in progenitor cells. Int. J. Mol. Med. 2006;17:681–685. [PubMed] [Google Scholar]
- 14.Nordlund J.J. The melanocyte and the epidermal melanin unit: an expanded concept. Dermatol. Clin. 2007;25:271–281. doi: 10.1016/j.det.2007.04.001. [DOI] [PubMed] [Google Scholar]
- 15.Boissy R.E. Melanosome transfer to and translocation in the keratinocyte. Exp. Dermatol. 2003;12(Suppl 2):5–12. doi: 10.1034/j.1600-0625.12.s2.1.x. [DOI] [PubMed] [Google Scholar]
- 16.Wang Y., Viennet C., Robin S., Berthon J.Y., He L., Humbert P. Precise role of dermal fibroblasts on melanocyte pigmentation. J. Dermatol. Sci. 2017;88:159–166. doi: 10.1016/j.jdermsci.2017.06.018. [DOI] [PubMed] [Google Scholar]
- 17.Yuan X.H., Jin Z.H. Paracrine regulation of melanogenesis. Br. J. Dermatol. 2018;178:632–639. doi: 10.1111/bjd.15651. [DOI] [PubMed] [Google Scholar]
- 18.Kim J.Y., Shin J.Y., Kim M.R., Hann S.K., Oh S.H. siRNA-mediated knock-down of COX-2 in melanocytes suppresses melanogenesis. Exp. Dermatol. 2012;21:420–425. doi: 10.1111/j.1600-0625.2012.01483.x. [DOI] [PubMed] [Google Scholar]
- 19.Zhu J.W., Ni Y.J., Tong X.Y., Guo X., Wu X.P. Activation of VEGF receptors in response to UVB promotes cell proliferation and melanogenesis of normal human melanocytes. Exp. Cell Res. 2020;387 doi: 10.1016/j.yexcr.2019.111798. [DOI] [PubMed] [Google Scholar]
- 20.O'Connell M.P., Weeraratna A.T. Hear the Wnt Ror: how melanoma cells adjust to changes in Wnt. Pigment Cell Melanoma Res. 2009;22:724–739. doi: 10.1111/j.1755-148X.2009.00627.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhan Z., Zhang X., Huang J., Huang Y., Huang Z., Pan X., Quan G., Liu H., Wang L., Wu A.C. Improved gene transfer with functionalized hollow mesoporous silica nanoparticles of reduced cytotoxicity. Materials. 2017;10 doi: 10.3390/ma10070731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nie X., Liu H., Liu L., Wang Y.D., Chen W.D. Emerging roles of Wnt ligands in human colorectal cancer. Front. Oncol. 2020;10:1341. doi: 10.3389/fonc.2020.01341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Costin G.E., Hearing V.J. Human skin pigmentation: melanocytes modulate skin color in response to stress. FASEB J. 2007;21:976–994. doi: 10.1096/fj.06-6649rev. [DOI] [PubMed] [Google Scholar]
- 24.Yamaguchi Y., Hearing V.J. Physiological factors that regulate skin pigmentation. Biofactors. 2009;35:193–199. doi: 10.1002/biof.29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Shi Y., Zeng Z., Liu J., Pi Z., Zou P., Deng Q., Ma X., Qiao F., Xiong W., Zhou C., et al. Particulate matter promotes hyperpigmentation via AhR/MAPK signaling activation and by increasing alpha-MSH paracrine levels in keratinocytes. Environ. Pollut. 2021;278 doi: 10.1016/j.envpol.2021.116850. [DOI] [PubMed] [Google Scholar]
- 26.Gu W.J., Ma H.J., Zhao G., Yuan X.Y., Zhang P., Liu W., Ma L.J., Lei X.B. Additive effect of heat on the UVB-induced tyrosinase activation and melanogenesis via ERK/p38/MITF pathway in human epidermal melanocytes. Arch. Dermatol. Res. 2014;306:583–590. doi: 10.1007/s00403-014-1461-y. [DOI] [PubMed] [Google Scholar]
- 27.Nakazawa K., Sahuc F., Damour O., Collombel C., Nakazawa H. Regulatory effects of heat on normal human melanocyte growth and melanogenesis: comparative study with UVB. J. Invest. Dermatol. 1998;110:972–977. doi: 10.1046/j.1523-1747.1998.00204.x. [DOI] [PubMed] [Google Scholar]
- 28.Zhu J.W., Ni Y.J., Tong X.Y., Guo X., Wu X.P., Lu Z.F. Tranexamic acid inhibits angiogenesis and melanogenesis in vitro by targeting VEGF receptors. Int. J. Med. Sci. 2020;17:903–911. doi: 10.7150/ijms.44188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Li M., Gao Y., Li C., Liu L., Li K., Gao L., Wang G., Zhang Z., Gao T. Association of COX2 functional polymorphisms and the risk of vitiligo in Chinese populations. J. Dermatol. Sci. 2009;53:176–181. doi: 10.1016/j.jdermsci.2008.09.010. [DOI] [PubMed] [Google Scholar]
- 30.Starner R.J., McClelland L., Abdel-Malek Z., Fricke A., Scott G. PGE(2) is a UVR-inducible autocrine factor for human melanocytes that stimulates tyrosinase activation. Exp. Dermatol. 2010;19:682–684. doi: 10.1111/j.1600-0625.2010.01074.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Guo H., Yang K., Deng F., Ye J., Xing Y., Li Y., Lian X., Yang T. Wnt3a promotes melanin synthesis of mouse hair follicle melanocytes. Biochem. Biophys. Res. Commun. 2012;420:799–804. doi: 10.1016/j.bbrc.2012.03.077. [DOI] [PubMed] [Google Scholar]
- 32.Babcock D.T., Shi S., Jo J., Shaw M., Gutstein H.B., Galko M.J. Hedgehog signaling regulates nociceptive sensitization. Curr. Biol. 2011;21:1525–1533. doi: 10.1016/j.cub.2011.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Moore C., Liedtke W.B. In: Neurobiology of TRP Channels. Emir T.L.R., editor. 2017. Osmomechanical-sensitive TRPV channels in mammals; pp. 85–94. [DOI] [PubMed] [Google Scholar]
- 34.Yang P., Zhu M.X. TRPV3. Handb. Exp. Pharmacol. 2014;222:273–291. doi: 10.1007/978-3-642-54215-2_11. [DOI] [PubMed] [Google Scholar]
- 35.Luo J., Hu H. Thermally activated TRPV3 channels. Curr. Top. Membr. 2014;74:325–364. doi: 10.1016/B978-0-12-800181-3.00012-9. [DOI] [PubMed] [Google Scholar]
- 36.Wang Y., Li H., Xue C., Chen H., Xue Y., Zhao F., Zhu M.X., Cao Z. TRPV3 enhances skin keratinocyte proliferation through EGFR-dependent signaling pathways. Cell Biol. Toxicol. 2021;37:313–330. doi: 10.1007/s10565-020-09536-2. [DOI] [PubMed] [Google Scholar]
- 37.Szöllősi A.G., Vasas N., Angyal Á., Kistamás K., Nánási P.P., Mihály J., Béke G., Herczeg-Lisztes E., Szegedi A., Kawada N., et al. Activation of TRPV3 regulates inflammatory actions of human epidermal keratinocytes. J. Invest. Dermatol. 2018;138:365–374. doi: 10.1016/j.jid.2017.07.852. [DOI] [PubMed] [Google Scholar]
- 38.Zhao J., Munanairi A., Liu X.Y., Zhang J., Hu L., Hu M., Bu D., Liu L., Xie Z., Kim B.S., et al. PAR2 mediates itch via TRPV3 signaling in keratinocytes. J. Invest. Dermatol. 2020;140:1524–1532. doi: 10.1016/j.jid.2020.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Vasas N., Pénzes Z., Kistamás K., Nánási P.P., Molnár S., Szegedi A., Szöllősi A.G., Bíró T. Transient receptor potential vanilloid 3 expression is increased in non-lesional skin of atopic dermatitis patients. Exp. Dermatol. 2022;31:807–813. doi: 10.1111/exd.14530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Park C.W., Kim H.J., Choi Y.W., Chung B.Y., Woo S.Y., Song D.K., Kim H.O. TRPV3 channel in keratinocytes in scars with post-burn pruritus. Int. J. Mol. Sci. 2017;18 doi: 10.3390/ijms18112425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tóth B.I., Dobrosi N., Dajnoki A., Czifra G., Oláh A., Szöllosi A.G., Juhász I., Sugawara K., Paus R., Bíró T. Endocannabinoids modulate human epidermal keratinocyte proliferation and survival via the sequential engagement of cannabinoid receptor-1 and transient receptor potential vanilloid-1. J. Invest. Dermatol. 2011;131:1095–1104. doi: 10.1038/jid.2010.421. [DOI] [PubMed] [Google Scholar]
- 42.Ständer S., Moormann C., Schumacher M., Buddenkotte J., Artuc M., Shpacovitch V., Brzoska T., Lippert U., Henz B.M., Luger T.A., et al. Expression of vanilloid receptor subtype 1 in cutaneous sensory nerve fibers, mast cells, and epithelial cells of appendage structures. Exp. Dermatol. 2004;13:129–139. doi: 10.1111/j.0906-6705.2004.0178.x. [DOI] [PubMed] [Google Scholar]
- 43.Kida N., Sokabe T., Kashio M., Haruna K., Mizuno Y., Suga Y., Nishikawa K., Kanamaru A., Hongo M., Oba A., Tominaga M. Importance of transient receptor potential vanilloid 4 (TRPV4) in epidermal barrier function in human skin keratinocytes. Pflugers Arch. 2012;463:715–725. doi: 10.1007/s00424-012-1081-3. [DOI] [PubMed] [Google Scholar]
- 44.Li W.H., Lee Y.M., Kim J.Y., Kang S., Kim S., Kim K.H., Park C.H., Chung J.H. Transient receptor potential vanilloid-1 mediates heat-shock-induced matrix metalloproteinase-1 expression in human epidermal keratinocytes. J. Invest. Dermatol. 2007;127:2328–2335. doi: 10.1038/sj.jid.5700880. [DOI] [PubMed] [Google Scholar]
- 45.Sokabe T., Fukumi-Tominaga T., Yonemura S., Mizuno A., Tominaga M. The TRPV4 channel contributes to intercellular junction formation in keratinocytes. J. Biol. Chem. 2010;285:18749–18758. doi: 10.1074/jbc.M110.103606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dhaka A., Viswanath V., Patapoutian A. Trp ion channels and temperature sensation. Annu. Rev. Neurosci. 2006;29:135–161. doi: 10.1146/annurev.neuro.29.051605.112958. [DOI] [PubMed] [Google Scholar]
- 47.Kettelhut E.A., Traylor J., Roach J.P. StatPearls; 2022. Erythema Ab Igne. [PubMed] [Google Scholar]
- 48.Melincovici C.S., Boşca A.B., Şuşman S., Mărginean M., Mihu C., Istrate M., Moldovan I.M., Roman A.L., Mihu C.M. Vascular endothelial growth factor (VEGF) - key factor in normal and pathological angiogenesis. Rom. J. Morphol. Embryol. 2018;59:455–467. [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
-
•
RNA-seq data have been deposited at NCBI and are publicly available as of the date of publication. Accession numbers are listed in the key resources table.
-
•
This paper does not report original code.
-
•
Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request.