Abstract Disrupted N^6-methyladenosine (m^6A) modification modulates various inflammatory disorders. However, the role of m^6A in regulating cutaneous inflammation remains elusive. Here, we reveal that the m^6A and its methyltransferase METTL3 are down-regulated in keratinocytes in inflammatory skin diseases. Inducible deletion of Mettl3 in murine keratinocytes results in spontaneous skin inflammation and increases susceptibility to cutaneous inflammation with activation of neutrophil recruitment. Therapeutically, restoration of m^6A alleviates the disease phenotypes in mice and suppresses inflammation in human biopsy specimens. We support a model in which m^6A modification stabilizes the mRNA of the lipid-metabolizing enzyme ELOVL6 via the m^6A reader IGF2BP3, leading to a rewiring of fatty acid metabolism with a reduction in palmitic acid accumulation and, consequently, suppressing neutrophil chemotaxis in cutaneous inflammation. Our findings highlight a previously unrecognized epithelial-intrinsic m^6A modification–lipid metabolism pathway that is essential for maintaining epidermal and immune homeostasis and lay the basis for potential therapeutic targeting of m^6A modulators to attenuate inflammatory skin diseases. __________________________________________________________________ Disrupted epithelial m^6A modification–lipid metabolism pathway drives skin inflammation via enhancing neutrophil chemotaxis. INTRODUCTION RNA modification plays a pivotal role in posttranscriptional regulation and notably influences the fate and function of RNA molecules ([86]1). Among various forms of RNA modifications identified to date, N^6-methyladenosine (m^6A) is the most abundant and prevalent modification on mRNA that profoundly affects RNA fate-related decisions, such as its splicing, decay, stabilization, and translation ([87]2). m^6A modification is catalyzed by methyltransferases (which are referred to as writers), removed by demethyltransferases (erasers), and recognized by m^6A-binding proteins (readers). Among all the known RNA methyltransferases, METTL3 is considered as the only S-adenosylmethionine–dependent methyltransferase that has catalytic effects on mRNA in mammals ([88]3). Numerous studies have elucidated the physiological and pathological roles of METTL3-mediated m^6A modification in specific cell types ([89]4). Increasing evidence underscores the pivotal and diverse roles of m^6A modification and its associated biochemical machinery in regulating various types of immune responses. For instance, inhibiting METTL3 enhances T cell–mediated cancer killing through activation of a cell-intrinsic interferon response ([90]5). In renal tubular epithelial cells, the lack of METTL3-mediated TAB3 m^6A modification leads to increased renal inflammation ([91]6). Reduced m^6A modification resulting from reduced METTL3 expression in monocyte macrophages enhances T helper 2 (T[H]2) cell responses and exacerbates allergic airway inflammation by promoting M2 macrophage activation in vivo ([92]7). Similarly, loss of METTL3-mediated m^6A modification in mast cells exacerbates their effector function in response to immunoglobulin E (IgE) and antigenic complexes ([93]8). In skin diseases, heterozygous knockout (KO) of Mettl3 worsens the severity of psoriasis ([94]9), while depletion of Mettl3 in CD4^+ T cells or γδ T cells alleviates psoriasis by inhibiting interleukin-17A (IL-17A) production ([95]10, [96]11). Psoriasis and atopic dermatitis (AD) are two kinds of highly prevalent chronic inflammatory skin diseases that share common features of epidermal hyperplasia and abundant infiltrates ([97]12–[98]14). Keratinocytes, in addition to various immune cells, play an essential role in their pathogenesis, particularly in driving the production of inflammatory molecules such as cytokines, chemokines, and lipid mediators ([99]12). Although aberrant responses of keratinocytes to T cell–derived cytokines are an intrinsic common pathological mechanism of psoriasis and AD ([100]12), other common mechanisms modulating skin inflammation remain unclear. Recent studies have highlighted the importance of m^6A modifications in skin epithelial progenitors. Xi et al. ([101]15) found that m^6A modification can affect cellular fate choices and tissue architecture during skin morphogenesis by enhancing translation of key morphogenetic regulators. Deletion of Mettl3 in epidermal progenitors results in impaired development and self-renewal, affecting hair follicle morphogenesis, cell adhesion, and polarity, which is associated with altered expression of histone-modifying enzymes ([102]16). Nevertheless, the specific impact of m^6A modification in epidermal keratinocytes on regulating cutaneous inflammation is not well defined. Given the versatility of the m^6A modification, we aimed to investigate its critical role in propagating inflammation in the skin, particularly within epidermal keratinocytes. Herein, we investigated the m^6A modification and its methyltransferase METTL3 in psoriasis and AD, using a genetic ablation approach to delineate the transcriptional and fatty acid metabolic rewiring that occurs during cutaneous inflammation resembling psoriasis or AD subsequent to depletion of the m^6A methyltransferases METTL3, which might elucidate metabolite-facilitated activation of the innate immunity. Our findings also underscore an m^6A-dependent effect in maintaining epidermal and immune homeostasis in the skin, with substantial implications for the treatment of psoriasis, AD, and other inflammatory diseases. RESULTS METTL3-m^6A pathway is down-regulated in psoriasis and AD To underline the dynamic of RNA m^6A in psoriasis, we analyzed the m^6A level by performing m^6A enzyme-linked immunosorbent assay (ELISA) and immunofluorescence staining. The results revealed a lower number of m^6A signals in the lesional skin of patients with psoriasis compared to the healthy skin ([103]Fig. 1A and fig. S1A). To identify the enzymes responsible for this m^6A reprogramming in skin inflammation, we analyzed the expression of m^6A writers and erasers in the lesional skin of patients with psoriasis and in the skin of healthy donors using publicly available RNA sequencing (RNA-seq) or microarray datasets [accession nos. [104]GSE13355 ([105]17) and [106]GSE121212 ([107]18)], quantitative polymerase chain reaction (PCR) with reverse transcription (RT-qPCR), and immunofluorescence staining. Our results showed a significant reduction in the key RNA methyltransferase METTL3 (both mRNA and protein levels) in the skin of patients with psoriasis, consistent with the decreased m^6A abundance in psoriatic lesions ([108]Fig. 1, B and C, and fig. S1B). In addition, the expression of METTL3 in the skin lesions of patients with psoriasis was restored after receiving biologic therapies, such as infliximab (anti–TNF-α) and secukinumab (anti–IL-17A) (fig. S1, C and D), supporting the strong association between METTL3-mediated m^6A and psoriasis development. Decreases in m^6A abundance and METTL3 expression were also detected in the epidermis keratinocytes of AD lesional skin and could be partially restored after crisaborole treatment ([109]Fig. 1D and fig. S1, E to G), indicating that the reduction in METTL3-mediated m^6A might be conservative changes in both psoriasis and AD. Fig. 1. METTL3-m^6A pathway is down-regulated in keratinocytes of psoriasis and AD. [110]Fig. 1. [111]Open in a new tab (A) Representative images of m^6A immunofluorescence staining in the lesional skin of patients with psoriasis or healthy control. (B and C) RT-qPCR of the indicated genes (B) and immunochemistry staining of indicated proteins (C) in the lesional skin of patients with psoriasis or the healthy skin. Scale bar, 100 μm. (D) Representative images of m^6A and METTL3 immunofluorescence staining in the lesional skin of patients with AD or healthy control. (E and F) Representative images of m^6A (E) and METTL3 (F) immunofluorescence staining in the dorsal skin from WT mice topically treated with IMQ for 6 days consecutively. (G and H) RT-qPCR analysis of Mettl3 mRNA (G) and immunoblot of METTL3 (H) in the epidermis from WT mice topically treated with IMQ. (I) Immunoblot of METTL3 in the dorsal skin treated with IMQ for 6 days consequently or in the recovery skin (on day 30). (J and K) Representative images of m^6A (J) and METTL3 (K) immunofluorescence staining in the dorsal skin from WT mice topically treated with MC903 for 8 days consecutively. (L and M) RT-qPCR analysis of Mettl3 mRNA (L) and immunoblot of METTL3 (M) in the epidermis from WT mice topically treated with MC903. (N) Immunoblot of METTL3 in ear skin treated with MC903 for 12 days consequently or in the recovery skin (on day 30). Scale bar, 50 μm [(A), (D), (E), (F), (J) and (K)]; scale bar, 100 μm (C). Pso, psoriasis; KRT14, keratin 14; IMQ, imiquimod; AD, atopic dermatitis; GAPDH, glyceraldehyde-3-phosphate dehydrogenase. *P < 0.05; **P < 0.01; ***P < 0.001. The P values were analyzed by unpaired, two-tailed Student’s t test [(B), (G), and (L)]. Data are presented as means ± SEM. Next, we evaluated the m^6A levels in the mouse models induced by imiquimod (IMQ, a Toll-like receptor 7 agonist)–induced psoriasis-like skin inflammation and calcipotriol (MC903)–induced AD-like skin inflammation. In both IMQ-induced and MC903-induced skin inflammation, decreases in m^6A modification and METTL3 expression were observed primarily within the epidermis rather than the dermis ([112]Fig. 1, E to H and J to M, and fig. S1, H, I, and K). Intriguingly, the decrease in METTL3 expression was transient as it was restored within 30 days after the initial IMQ or MC903 treatment, at which point the skin regained homeostasis ([113]Fig. 1, I and N, and fig. S1, J and L). These findings suggest that a similar pattern was observed in the mouse models of psoriasis and AD as in the lesional skin of patients with psoriasis and AD. Last, the decrease in METTL3 expression not only was limited to the skin inflammation induced by IMQ or MC903 but also occurred in inflammation induced by tumor necrosis factor–α (TNF-α) and IL-17A (fig. S1, M and N). These data suggest that a reduction in the METTL3-m^6A axis may be prevalent in skin inflammation. Inducible keratinocyte-specific Mettl3-deficient mice develop spontaneous skin lesions It has been previously reported that loss of m^6A in preimplantation epithelial cells leads to early implantation lethality ([114]19). Notably, mice with decreased m^6A modification in keratinocytes [Mettl3^f/f; keratin 14 (KRT14)–Cre mice] have a limited life span, and most do not survive beyond postnatal day 6 ([115]15). Their poor survival is possibly related to a reduction in tongue filiform papillae rather than concomitant alterations in the skin barrier function or inflammation ([116]15). Therefore, to investigate the role of epidermal m^6A modification in vivo, we generated an inducible, keratinocyte-specific Mettl3-ablation mouse model by crossing Mettl3^flox/flox mice with KRT14-CreER^T2 mice in which the deletion of Mettl3 in keratinocytes was induced following tamoxifen injection, hereafter referred to as Mettl3 cKO mice ([117]Fig. 2, A and B). As expected, marked reductions in both protein and mRNA levels of METTL3 as well as in m^6A modification levels were observed in the epidermis of Mettl3 cKO mice ([118]Fig. 2, C and D, and fig. S2, A and B). Skin lesions exhibiting erythema, scaling, and hair loss developed spontaneously in Mettl3 cKO mice 2 weeks after the initial tamoxifen injection, mainly at the base of the tail and in the palm area ([119]Fig. 2, E to H). Histological examination of the skin lesions revealed epidermal hyperplasia, hyperkeratosis, parakeratosis, and extensive cellular infiltration in the dermis of Mettl3 cKO mice ([120]Fig. 2, G and H). Furthermore, the lesional skin section of Mettl3 cKO mice showed a significant increase in the number of epidermal cells positive for Ki67^+ (a marker of cell proliferation) and a marked decrease in the expression of loricrin (a marker of terminal differentiation) in the epidermis ([121]Fig. 2I and fig. S2, C and D). These data provide strong evidence that METTL3-mediated m^6A in epidermal keratinocytes is crucial for maintaining skin homeostasis in adult mice. Fig. 2. Deletion of Mettl3 in the epidermal keratinocytes of adult mice leads to spontaneous skin inflammation. [122]Fig. 2. [123]Open in a new tab (A and B) Mouse construct (A) and schematic diagram (B) for CreER-inducible Mettl3 conditional ablation specific in keratinocytes and harvest time in (C) and (D). (C and D) Immunofluorescence staining of METTL3 (C) or m^6A (D) in the dorsal skin from Mettl3 cKO (KRT14-CreER^T2; Mettl3^fl/fl) or WT (Mettl3^fl/fl) mice. Scale bars, 50 μm. (E) Schematic diagram for experimental design for inducible keratinocyte-specific ablation of Mettl3 observation in (F) to (N). (F) Incidence of skin lesions in the tail or palm of Mettl3 cKO and WT mice over time (n = 20). (G and H) Dermoscopic images, H&E staining of the tail (G) or palm (H) skin lesions, and their quantitation of acanthosis and dermal cellular infiltrates on the lesional skin of Mettl3 cKO and WT mice skin in the indicated locations. Scale bars, 100 μm. (I) Immunofluorescence staining of loricrin in lesional skins of Mettl3 cKO mice or WT mice skin in the indicated locations. Scale bars, 50 μm. Dashed line indicates the border between the epidermis and dermis. (J and K) Top 10 GO terms (J) and KEGG pathways (K) enriched in DEGs identified by RNA-seq between the spontaneously inflamed tail skin of Mettl3 cKO mice and the normal tail skin of WT mice. (L) Heatmap showing expression of selected genes [Fragments Per Kilobase of transcript per Million mapped reads (FPKM)] involved in immune responses in WT and Mettl3 cKO skin. (M) GSEA of genes in Mettl3 cKO skin relative to control skin with DEGs in the human psoriatic skin (left) or AD skin (right) ([124]GSE121212) enriched. TAM, tamoxifen; AD, atopic dermatitis; ECM, extracellular matrix; NES, normalized enrichment score. *P < 0.05; **P < 0.01; ***P < 0.001. The P values were analyzed by unpaired, two-tailed Student’s t test [(G) and (H)]. Data are presented as means ± SEM. To seek the molecular insights into the formation of spontaneously inflamed skin lesions in Mettl3 cKO mice, we conducted transcriptome analysis and compared the results to those in wild-type (WT) mice. Through gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, we identified that the differentially expressed genes (DEGs), selected based on the criteria of adjusted P < 0.05 and log[2] fold change (|log[2]FC|) > 2 from RNA-seq, were significantly enriched in pathways related to innate immune response, proliferation, and activation of T cells, cytokine–cytokine receptor interaction, and neutrophil chemotaxis ([125]Fig. 2, J and K). Furthermore, several specific inflammatory signaling pathways, such as the cytokine-related signaling pathway and the nuclear factor κB (NF-κB) signaling pathway, were also enriched ([126]Fig. 2K). The heatmap showed increased expression of several immune response–related genes in the spontaneously inflamed lesional skin of Mettl3 cKO mice compared with the normal skin of WT mice ([127]Fig. 2L). Moreover, RT-qPCR analysis confirmed elevated expression of multiple inflammatory factors, including Tnf, Il23a, Cxcl1, Cxcl2, Cxcl3, Il6, Tslp, Il33, S100a8, and S100a9, in the skin lesions of Mettl3 cKO mice (fig. S2E). These factors are crucial for T[H]17 (e.g., Il23) and T[H]2 (e.g., Tslp and Il33) immune responses. Furthermore, as the phenotypes closely resemble the phenotypes of skin lesions from patients with psoriasis or AD, we next performed gene set enrichment analysis (GSEA) by comparing transcriptomic data from spontaneously inflamed skin lesions of Mettl3 cKO mice with the data from the normal skin tissue and the data from the GEO database [accession no. [128]GSE121212 ([129]18)] from psoriatic or AD skin samples relative to normal skin samples. It was shown that the down-regulated transcriptional signatures of skin lesions in Mettl3 cKO mice significantly overlapped with the down-regulated DEGs identified in psoriatic or AD skin samples relative to normal skin samples ([130]Fig. 2M), implying the molecular resemblance between these spontaneous mouse skin lesions and both psoriasis and AD skin in human patients. Mettl3 deficiency in epidermal keratinocytes exacerbates both psoriasis-like and AD-like skin inflammation To directly examine the impact of METTL3-mediated m^6A methylation in epidermal keratinocytes on psoriasis-like and AD-like skin inflammation, we next compared the IMQ-induced or MC903-induced skin responses in Mettl3 cKO and WT mice. IMQ elicited more severe psoriasis-like skin symptoms in epidermal m^6A-diminished Mettl3 cKO mice ([131]Fig. 3, A and B, and fig. S3A). This was evident from the higher psoriasis area and severity index (PASI) score and greater transepidermal water loss (TEWL) as well as the more prominent acanthosis and elevated dermal inflammatory cell infiltration ([132]Fig. 3, B to D, and fig. S3B). In addition, compared to WT mice, Mettl3-deficient mice showed a more pronounced increase in the percentage of epidermal Ki67^+ cells and a marked decrease in the expression of the terminal differentiation markers, loricrin and filaggrin, in the epidermis after IMQ application (fig. S3, C to E). Furthermore, the mRNA expression of psoriasis-related inflammatory cytokines/chemokines, such as Il17a, Tnf, Il6, Mmp9, Cxcl1, Cxcl2, Cxcl3, Cxcl5, Cxcl15, and Csf3, was elevated in the skin of IMQ-treated Mettl3 cKO mice compared with WT mice ([133]Fig. 3E and fig. S3F). Fig. 3. Mettl3 deficiency in epidermal keratinocytes exacerbates skin inflammation by enhancing neutrophil infiltration. [134]Fig. 3. [135]Open in a new tab (A) Schematics of IMQ treatment strategy. (B) Representative images and PASI score of the skin after treated with IMQ for 6 days (left) or for the indicated time (right). (C and D) H&E staining images (C) and their quantification (D) of the skin treated with IMQ for 6 days. (E) Heatmap showing the expression of indicated genes in the skin after 3 days of IMQ treatment. (F) Schematics of MC903 treatment strategy. (G) Representative images and EASI score of the skin after 6 days (left) or for the indicated time (right). (H and I) H&E staining images (H) and their quantification (I) of the skin. (J) Heatmap showing the expression of indicated genes in the skin by performing RT-qPCR. (K) Flow cytometry of CD45^+ cells and neutrophils in the skin. (L and M) Immunofluorescence staining of Ly6G and CD11b in the skin after IMQ (L) or with MC903 (M) treatment. (N and O) Heatmap showing the expression of indicated genes in the epidermis after IMQ treatment for 3 days (N) or in the 6-day MC903-treated mouse skin (O). (P) Schematics of neutrophil depletion strategy. (Q) Representative images of the skin after IMQ treatment for 6 days (left) or for the indicated time (right). (R and S) H&E staining images (R) and their quantifications (S) of the skin. (T and U) Immunofluorescence staining of METTL3 (T) and m^6A (U) in the lesional skin from patients with generalized pustular psoriasis (GPP) or the healthy skin. Scale bars, 100 μm [(C), (H), (L), (M), and (R)] and 50 μm [(T) and (U)]. Ab, antibody. *P < 0.05; **P < 0.01; ***P < 0.001. The P values were determined by two-way ANOVA [(B) and (Q)] or unpaired, two-tailed Student’s t test [(D), (I), (K), and (S)]. Data are presented as means ± SEM. Similar to the above results, we found a protective role of epidermally expressed Mettl3 against the MC903-induced AD-like skin inflammation. MC903 treatment of the ear skin in Mettl3 cKO mice led to an increase in erythema and scaling that was accompanied by a significant increase in skin thickness, acanthosis, and dermal cell infiltration compared to WT mice (fig. S3, G to K). In addition, MC903-treated Mettl3 cKO mice also developed skin lesions in areas that were not in direct contact with MC903 (e.g., eyes, face, and tail root): They exhibited hair loss and skin erythema covered with scaling (fig. S3, L and M). Similarly, MC903 treatment of the dorsal skin resulted in more severe skin phenotypes, including more pronounced keratinocyte terminal differentiation defects, in Mettl3 cKO mice than in the WT mice ([136]Fig. 3, F to I, and fig. S3, N to P). In addition, the expression of the AD-related inflammatory factors (Tslp, Il33, and Il6), but not Il4 and Il13, was elevated in the skin of MC903-treated Mettl3 cKO mice ([137]Fig. 3J and fig. S3Q). Notably, our results indicate no discernible disparity in TEWL between Mettl3 cKO mice and WT mice 9 days after tamoxifen injection treatment (fig. S3, B and N, day 0), consistent with previous evidence indicating that mice with specific Mettl3 knockdown in keratinocytes do not exhibit barrier impairment ([138]15). Collectively, our data demonstrate through IMQ-induced and MC903-induced mouse models with Mettl3 deficiency that decreases in METTL3 and m^6A modification of RNA in epidermal keratinocytes are also critically important for maintaining skin homeostasis and suppressing skin inflammation in response to psoriasis-associated or AD-associated stimuli. Disrupted m^6A in epidermal keratinocytes leads to an excessive influx of neutrophils in skin inflammation To further understand the nature of the skin inflammation in Mettl3 cKO mice, we conducted flow cytometry analysis to profile immune cell compositions in the skin lesions of Mettl3 cKO mice and their sex-matched WT littermates (fig. S4). A drastic increase in the number of total immune (CD45^+) cells was observed in IMQ-treated Mettl3 cKO mice compared with WT mice, and neutrophils represented the major cell type (>50% of immune cells) that underwent population expansion ([139]Fig. 3K and fig. S5, A to C). Immunofluorescence staining with antibodies against the Ly6G and CD11b confirmed a more rapid accumulation of neutrophils in the skin lesions of Mettl3 cKO mice ([140]Fig. 3L). In contrast, the T cell numbers declined in Mettl3 cKO mice, while the numbers of other types of immune cells, including macrophages and dendritic cells, were not significantly altered (fig. S5, D to L). A higher degree of neutrophil recruitment was also observed in MC903-treated Mettl3 cKO mice than in WT mice ([141]Fig. 3M). These results highlight neutrophil infiltration and accumulation as a major event that occurs during exacerbated psoriasis-like and AD-like skin inflammation in Mettl3 cKO mice. To investigate the mechanism underlying enhanced neutrophil accumulation, we performed RT-qPCR analysis of genes related to neutrophil chemotaxis and adhesion (such as Cxcl1, Cxcl2, Cxcl3, Cxcl5, Cxcl15, Csf3, and Icam1) in the skin lesions of both the IMQ-induced and MC903-induced inflammation mouse models. The expression of these genes was significantly elevated in the skin lesions of Mettl3 cKO skin compared to the skin lesions of WT mice skin in both models ([142]Fig. 3, N and O, and fig. S6, A and B). To assess the potential keratinocyte-intrinsic function of METTL3, we treated HaCaT cells, an immortalized human keratinocyte cell line, with a potent METTL3 inhibitor—STM2457 ([143]20). As expected, this led to a significant reduction in m^6A levels (fig. S6C). When HaCaT cells were treated with STM2457 along with a cocktail of inflammatory factors (the M5 cocktail, which includes IL-17A, TNF-α, IL-1α, IL-22, and oncostatin M) ([144]21) that is known to mimic a psoriatic inflammatory environment, it resulted in significantly elevated expression of neutrophil chemotaxis–related and adhesion-related genes, such as CXCL1, CXCL2, CXCL3, CXCL6, CSF3, and ICAM1, in comparison to the dimethyl sulfoxide (DMSO) control (fig. S6D). These results are indicative of a keratinocyte-specific role of METTL3, and by inference m^6A modification of mRNAs, in regulating the expression of genes involved in neutrophil recruitment. To examine the functional contribution of neutrophils to IMQ-induced skin inflammation in Mettl3 cKO mice, we used a neutralizing antibody against Ly6G to induce the depletion of neutrophils ([145]Fig. 3P and fig. S6E). Administration of the Ly6G antibody partially rescued the exacerbation of IMQ-induced skin inflammation in Mettl3 cKO mice, as evidenced by a significant decline in both the PASI score and TEWL value ([146]Fig. 3Q and fig. S6F). Moreover, histological analysis and immunofluorescence staining revealed a significant reduction in acanthosis, dermal cell infiltration, and epidermal Ki67^+ cell numbers in response to neutrophil depletion ([147]Fig. 3, R and S, and fig. S6G). Pustular psoriasis is a severe type of psoriasis characterized by the presence of pustules filled with neutrophils ([148]22). As anticipated, we found a significant decrease in m^6A modification and METTL3 expression in the keratinocytes of pustular psoriasis compared to those of the healthy skin ([149]Fig. 3, T and U). Accordingly, our mechanistic experiments indicate that METTL3, and by inference m^6A modification, had a regulatory effect on the expression of genes related to neutrophil recruitment. Thus, METTL3/m^6A may play a protective role in inflammatory skin conditions through inhibition of neutrophil infiltration. Therapeutic potential of an m^6A activator in cutaneous inflammation Methyl piperidine-3-carboxylate (MP3C) has been recently reported to be an agonist that targets the m^6A methyltransferase METTL3-METTL14-WTAP complex ([150]23). We next investigated if MP3C could activate m^6A methyltransferase activity in skin lesions and alleviate the inflammatory skin conditions. The efficacy of MP3C in activating m^6A methyltransferase activity was confirmed by quantifying the levels of m^6A in RNA isolated from HaCaT cells after treatment with or without MP3C (fig. S7A). In the next set of experiments, we evaluated the therapeutic effectiveness of MP3C in models of skin inflammation induced by IMQ or MC903. Our results demonstrated that topical application of MP3C effectively reduced cutaneous inflammation induced by IMQ or MC903 ([151]Fig. 4, A to D and G to J, and fig. S7, B and C), with an increase in m^6A abundance in the epidermis of lesional skin compared to the control group (fig. S7, D and I). In addition, enhanced filaggrin and loricrin expressions, as well as decreased inflammatory factor expression, were observed in psoriasis-like or AD-like skin lesions after topical application of MP3C ([152]Fig. 4, E and K, and fig. S7, E to G and J to L). Notably, CD45^+ cells and neutrophil numbers were significantly diminished after topical application of MP3C to IMQ-induced psoriasis-like skin lesions ([153]Fig. 4F and fig. S7H). This is consistent with the flow cytometry results for skin lesions in Mettl3 cKO mice. Fig. 4. Activation of the m^6A modification dampens the severity of skin inflammation. [154]Fig. 4. [155]Open in a new tab (A) Schematic diagram of strategy for application of MP3C or EtOH and IMQ. (B) Representative images (left) and PASI score (right) of mice treated with MP3C or EtOH after IMQ treatment for 6 days or at the indicated time. (C and D) H&E staining images (C) and their quantification (D) of mice treated with MP3C or EtOH after 6-day IMQ application. (E) RT-qPCR analysis of the expression of indicated genes in the 6-day IMQ-treated skin from mice with MP3C or EtOH application. (F) Flow cytometry of CD45^+ cells and neutrophils in the 6-day IMQ-treated skin from mice with MP3C or EtOH application. (G) Schematic diagram of strategy for application of MP3C or EtOH and MC903. (H) Representative images and EASI score of mice treated with MP3C or EtOH after MC903 treatment for 6 days (left) or at the indicated time (right). (I and J) H&E staining images (I) and their quantifications (J) of mice treated with MP3C or EtOH after 6-day MC903 application. (K) RT-qPCR analysis of the expression of indicated genes in the 6-day MC903-treated mouse skin with MP3C or EtOH application. (L) Schematic depicting the treatment of skin biopsies obtained from healthy donors (left) and RT-qPCR analysis of specified genes in these biopsies after a 6-hour treatment (right). (M) Schematic depicting the treatment of lesional psoriatic skin biopsies (left) and RT-qPCR analysis of specified genes in these biopsies after a 24-hour treatment (right). The figures were created with [156]BioRender.com [(L) and (M)]. Scale bar, 100 μm [(C) and (I)]. *P < 0.05; **P < 0.01; ***P < 0.001. The P values were determined by two-way ANOVA [(B), (H), and (L)], unpaired two-tailed Student’s t test [(D), (F), and (J)], or paired Student’s t test (M). Data are means ± SEM. To address the translational relevance of MP3C in human inflammatory skin diseases, we conducted experiments using cultured skin biopsy tissues obtained from healthy donors. We found that M5 stimulation markedly increased the expression of inflammatory factors, including chemoattractants for neutrophils, T[H]17-related cytokines, and T[H]2-related cytokines such as TSLP and IL33. However, treatment with MP3C effectively suppressed these inflammatory responses ([157]Fig. 4L). Incubation of lesional psoriasis skin cultures with MP3C resulted in a decreased expression of several psoriasis-related genes, chemoattractants for neutrophils, and adhesion molecule–encoding genes, including CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL8, IL17A, ITNF, and IL6 ([158]Fig. 4M). These data suggest that MP3C has anti-inflammatory properties in human skin biopsies and may reduce inflammatory responses in the lesional skin from patients with psoriasis under ex vivo conditions. In summary, our study provides evidence that activation of m^6A modification could ameliorate skin inflammation and might serve as a potential therapeutic strategy for targeting m^6A in psoriasis and AD. Mettl3 deficiency in keratinocytes leads to fatty acid metabolic reprogramming To gain molecular insight into the effects of m^6A deficiency on skin inflammation, particularly its effects on epidermal keratinocytes, we performed methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA-seq analysis on the lesional skin of patients with psoriasis and the healthy skin as well as the epidermis of Mettl3 cKO and WT mice treated with IMQ for 3 or 6 days ([159]Fig. 5A). Our analysis revealed a significant positive correlation between mRNA expression and m^6A modification levels in the lesional skin of psoriatic patients versus the healthy skin tissue ([160]Fig. 5B). Furthermore, Metascape analysis ([161]24) showed that genes down-regulated or hypomethylated in the psoriatic lesional skin were enriched in long-chain fatty acid metabolic process or the lipid biosynthesis process ([162]Fig. 5C). Fig. 5. Lipid metabolism undergoes rewiring in psoriasis or in skin inflammation by Mettl3 depletion. [163]Fig. 5. [164]Open in a new tab (A) Schematic illustration of sample collection for MeRIP-seq in psoriasis. The figure was created with [165]BioRender.com. (B) The volcano plot displays changes of RNA expression and m^6A modification levels between the lesional skin of patients with psoriasis and the healthy skin. Dashed lines define log[2] FC > 1 and log[2] FC < −1. Purple and green dots were peaks with |FC| > 2 and adjusted P value < 0.05. (C) Top 10 enrichment clusters of down-regulated genes and hypomethylated genes in the lesional skin of patients with psoriasis analyzed using the Metascape database. One row per cluster uses a discrete color scale to represent statistical significance. (D and E) Volcano plot depicts significantly DEGs and DMPs (adjusted P < 0.05 and |FC| > 2) from MeRIP-seq between the epidermis of Mettl3 cKO and WT mice after IMQ treatment for 3 or 6 days (D) and the heatmap showing the top 20 enrichment clusters analyzed using the Metascape database (E). (F) GSEA performed by CEMiTool showing module activity for each sample group (n = 3). (G) Depiction of the gene interaction network of module 3 from G in (F), which related to lipid metabolism. (H) Top 10 of GO terms enrichment of down-regulated DEGs in spontaneous skin lesions of Mettl3 cKO mice relative to WT skin. (I) Absolute quantification of fatty acids in the epidermis of Mettl3 cKO or WT mice after IMQ treatment using GC-MS. Lipid metabolism–related pathways are marked in red [(C), (E), and (H)]. PCC, Pearson’s correlation coefficient; D, day; IMQ, imiquimod; NES, normalized enrichment score; DEG, differentially expressed gene; DMP, differentially methylated peak; Hyper, hypermethylated gene; Hypo, hypomethylated gene; Down, down-regulated gene; Up, up-regulated gene. GC-MS, gas chromatography–mass spectrometry; CoA, coenzyme A. *P < 0.05. The P values were determined by two-tailed Student’s t test (I). Data are presented as means ± SEM. As observed in the human skin samples, a strong positive correlation was also observed between mRNA expression and m^6A modification levels, with most genes showing reduced mRNA levels and m^6A hypomethylation in the epidermis of Mettl3 cKO mice ([166]Fig. 5D). Metascape analysis of the genes that were down-regulated or hypomethylated in the epidermis of IMQ-treated Mettl3 cKO mice revealed lipid or fatty acid metabolism–associated clusters as being among the top 10 enriched clusters across multiple gene lists ([167]Fig. 5E). Furthermore, we performed a modular gene coexpression network analysis using CEMiTool ([168]25), through which seven individual modules were identified in our dataset ([169]Fig. 5F). Of note, the expression of module 3 was decreased in the epidermis of Mettl3 cKO mice after IMQ treatment compared to the epidermis of WT mice, and numerous pathways relevant to fatty acid metabolism or biosynthesis were enriched in this module ([170]Fig. 5G). In addition, GO enrichment analysis of DEGs down-regulated in the spontaneous skin lesions of Mettl3 cKO mice relative to the normal skin showed that lipid and fatty acid metabolic processes were the top terms ([171]Fig. 5H). On the basis of the above results, we aimed to investigate the impact of METTL3-mediated m^6A plays a role in skin inflammation via induction of fatty acid metabolism reprogramming and analyzed the fatty acid composition in the epidermis of Mettl3 cKO mice and WT mice after IMQ treatment. Our results revealed that the levels of palmitic acid (PA), stearic acid (SA), elaidic acid (EA), and oleic acid (OA) were significantly higher in the epidermis of Mettl3 cKO mice than in the epidermis of WT mice ([172]Fig. 5I). Conversely, a decreasing tendency was observed in the levels of very-long-chain fatty acids, such as erucic and lignoceric acids ([173]Fig. 5I). This suggests a potential disruption in fatty acid carbon chain elongation due to the loss of METTL3. The above results provide compelling evidence for the involvement of METTL3 in regulating the mRNA m^6A modification in genes associated with fatty acid metabolism in epidermal keratinocytes. Consequently, the loss of METTL3/m^6A leads to the reprogramming of fatty acid in the epidermis. ELOVL6 is a critical downstream target of METTL3 in cutaneous inflammation To identify the functionally important target of METTL3 in skin inflammation, we performed a Venn diagram analysis using our RNA-seq and MeRIP-seq data to identify the potential target genes involved in lipid metabolism that exhibit down-regulation and hypomethylation in Mettl3 cKO compared to WT mice. Nine potential target genes were identified, namely, Acot1, Acox3, Agpat4, Cers4, Elovl3, Elov6, Hmgcr, Mgll, and Lpcat3 ([174]Fig. 6A). Among these genes, Elovl6 and Elovl3 mRNA were previously reported to play critical regulatory roles in fatty acid metabolism ([175]26). Specifically, ELOVL6 converts C16 saturated and monounsaturated fatty acids to C18 species ([176]27), while ELOVL3 catalyzes the synthesis of C20 to C24 fatty acids ([177]28). Fig. 6. Elovl6 is identified as a target gene of METTL3. [178]Fig. 6. [179]Open in a new tab (A) Venn diagram of genes associated with metabolism of lipids and hypomethylated genes in cKO mice versus WT mice after IMQ treatment. (B) Integrative Genomics Viewer (IGV) tracks showing m^6A modification on Elovl6 mRNAs in the IMQ-treated epidermis. (C and D) SN ratio or Elovl6 mRNA level of the epidermis of cKO mice relative to WT mice treated with IMQ for 3 days (C) or with MC903 for 6 days (D). (E) Immunofluorescence staining of ELOVL6 in the skin treated with IMQ for 6 days. (F and G) Expression of ELOVL6 in the 6-day IMQ-treated epidermis from C57BL6/J mice with MP3C or EtOH application. (H) Signal-to-Noise (SN) ratio of Elovl6 RNA in the epidermis. (I and J) Immunofluorescence staining of ELOVL6 in the MC903-treated skin from WT or cKO mice (I) or from C57BL6/J mice with MP3C or EtOH application (J). (K and L) ELOVL6 expression in the tail (K) or palm (L) skin of WT and cKO mice. (M and N) IGV tracks showing m^6A modification on ELOVL6 mRNAs in the human skin tissue (M) or in HEKa (N). (O) SN ratio of ELOVL6 RNA in HEKa relative to the DSMO group. (P) Immunoblot of ELOVL6 in HEKa after STM2457 treatment. (Q) Actinomycin D chase experiments for ELOVL6 mRNA expression in HaCaT cells. (R and S) Expression of ELOVL6 after IGF2BP1-3 was silenced in HaCaT cells. (T) RIP-qPCR tested the binding of IGF2BP3 and ELOVL6 mRNA in HaCaT cells treated with STM2457 or DMSO. Scale bar, 50 μm. *P < 0.05; **P < 0.01; ***P < 0.001. The P values were determined by two-way ANOVA [(C), (D), (Q), and (T)], unpaired, two-tailed Student’s t test [(C), (D), (F), (H), (K), and (O)], or one-way ANOVA (R). Data are presented as means ± SEM. Next, we sought to confirm if ELOVL3 and ELOVL6 are direct targets of METTL3 in keratinocytes of skin inflammation. We found that significant reductions in m^6A modification and the expression levels of Elovl6 were observed in the epidermis of Mettl3 cKO mice compared to WT mice in both IMQ-induced and MC903-induced skin inflammation ([180]Fig. 6, B to E and I, and fig. S8A), while MP3C treatment restored the m^6A modification and the expression levels of Elovl6 ([181]Fig. 6, F to H and J). Moreover, reduced mRNA and/or protein levels of Elovl6 were also observed in the epidermis of spontaneously developed skin lesions in Mettl3 cKO mice compared to WT mice ([182]Fig. 6, K and L). Furthermore, we found that the m^6A level of ELOVL6 was significantly decreased in the lesional skin of patients with psoriasis or AD according to the results of MeRIP-seq analysis (P value = 0.05 for psoriatic versus healthy control; P value = 0.03 for AD versus healthy control) ([183]Fig. 6M). To determine if ELOVL6 mRNA is a bona fide target of METTL3-mediated m^6A modification in human epidermal keratinocytes, we used both adult human keratinocytes. As we expected, treatment of human epidermal keratinocytes with STM2457 resulted in down-regulation of the m^6A level in ELOVL6 mRNA, accompanied by a decrease in ELOVL6 protein levels ([184]Fig. 6, N to P, and fig. S8, B to E). In contrast, although reduced m^6A modification and mRNA expression of Elovl3 were observed in the epidermis of inflammatory skin lesions from Mettl3 cKO mice (fig. S8, F to H), the m^6A signal was barely detectable in ELOVL3 mRNA in HEKa and HaCaT cells (fig. S8, I and J), and ELOVL3 mRNA expression was not suppressed by STM2457 treatment (fig. S8K). These data demonstrated that ELOVL6, but not ELOVL3, is a direct target of METTL3-mediated m^6A modification in epidermal keratinocytes. Then, we examined the impact of m^6A on ELOVL6 mRNA stability. As expected, in STM2457 pretreated cells, ELOVL6 mRNA stability decreased after actinomycin D treatment, which led to the inhibition of gene transcription ([185]Fig. 6Q). The potential function of m^6A readers in ELOVL6 mRNA stability was further explored. Among various m^6A reader proteins, insulin-like growth factor 2 mRNA-binding proteins (IGF2BPs, including IGF2BP1/2/3) have been previously reported to enhance the stability and translation of target RNAs ([186]2, [187]29). To investigate if IGF2BPs might be required for ELOVL6 mRNA stability in keratinocytes, we individually knocked down IGF2BP1/2/3 respectively in HaCaT cells and found that silencing IGF2BP3 significantly inhibited ELOVL6 expression ([188]Fig. 6, R and S, and fig. S8L). In addition, RIP-qPCR revealed the direct binding of IGF2BP3 to ELOVL6 mRNA, and this binding was found to be inhibited by STM2457 ([189]Fig. 6T). This is indicative of a dependence on METTL3 activity/m^6A modification. Together, these results identify IGF2BP3 as a potential reader of m^6A-modified ELOVL6 mRNA that promotes its stability. ELOVL6 inhibition aggravates the severity of inflammatory skin conditions To further investigate the role of ELOVL6 in the pathogenesis of psoriasis and AD, we first determined the expression level of ELOVL6 in psoriasis and AD. We found a significant decrease in the mRNA levels of ELOVL6 in the lesional skin compared to nonlesional skin in patients with both psoriasis and AD by analyzing a publicly available RNA-seq dataset [accession no. [190]GSE121212 ([191]18)] ([192]Fig. 7A). Our immunofluorescence staining results further confirmed this markedly reduced expression of the ELOVL6 protein, particularly in epidermal keratinocytes from patients with psoriasis (including psoriasis vulgaris and pustular psoriasis) and AD ([193]Fig. 7B). Consistent with the decreased expression of METTL3 observed in the inflamed murine skin ([194]Fig. 1, G, H, L, and M), the mRNA and protein expression levels of ELOVL6 were found to be significantly reduced in the epidermis of WT mice treated with both IMQ and MC903 compared to their vehicle control groups ([195]Fig. 7, C and D). Fig. 7. Inhibition of ELOVL6 exacerbates skin inflammation. [196]Fig. 7. [197]Open in a new tab (A) Expression of ELOVL6 mRNA reanalyzed from the RNA-seq analysis ([198]GSE121212) of the paired lesional and nonlesional skin from patients with psoriasis (n = 27) or AD (n = 21). (B) Immunofluorescence staining of ELOVL6 in the lesional skin of patients with PSV, GPP, and AD or the healthy skin. (C and D) RT-qPCR analysis (left) and immunofluorescence staining (right) of ELOVL6 in the epidermis from WT mice treated with IMQ (C) or MC903 (D). (E) Schematic diagram of strategy for application of ELOVL6-IN and IMQ. (F) Representative images (left) and PASI score (right) of mice treated with ELOVL6-IN after IMQ treatment for 5 days (left) or at the indicated time (right). (G and H) H&E staining images (G) and their quantifications (H) of the mouse skin. (I) Heatmap showing the expression of indicated genes in the mouse skin by performing RT-qPCR. (J) Flow cytometry of CD45^+ cells and neutrophils in the mouse skin. (K) Schematic diagram of strategy for application of ELOVL6-IN and MC903. (L) Representative images and EASI score of the dorsal skin of mice treated with ELOVL6-IN after application of MC903 for 5 days (left) or at the indicated times (right). (M and N) H&E staining images (M) and their quantifications (N) of the mouse skin. (O) Heatmap showing the expression of indicated genes after the 5-day MC903-treated mouse skin by performing RT-qPCR. Scale bars, 100 μm [(F) and (M)] and 50 μm [(B) to (D)]. The dashed line indicates the border between the epidermis and dermis [(C) and (D)]. *P < 0.05; **P < 0.01; ***P < 0.001. The P values were determined by paired, two-tailed Student’s t test (A), unpaired, two-tailed Student’s t test [(C), (D), (H), (J), and (N)], or two-way ANOVA [(F) and (L)]. Data are presented as means ± SEM. To assess the potential role of ELOVL6 in regulating cutaneous inflammation, we topically applied a selective ELOVL6 inhibitor ([199]30) to the dorsal skin of C57BL/6J mice ([200]Fig. 7E). After the IMQ treatment, mice treated with the ELOVL6 inhibitor displayed significantly more pronounced psoriasis-like skin features compared to the vehicle control mice ([201]Fig. 7, F to H, and fig. S9, A to D). Furthermore, the skin of mice treated with the ELOVL6 inhibitor exhibited more marked up-regulation of psoriasis-related genes (such as Il17a, Il6, Mmp9, and S100a8) and neutrophil chemotaxis/adhesion–related genes (such as Cxcl1, Cxcl2, Cxcl5, Csf3, and Icam1) than the skin of mice in the control group ([202]Fig. 7I and fig. S9E). Consistent with these findings, the results of flow cytometry also revealed a remarkable increase in the number of CD45^+ cells and neutrophils in the ELOVL6 inhibitor–treated skin after IMQ application ([203]Fig. 7J and fig. S9F). Similarly, ELOVL6 inhibition resulted in worsened skin inflammation in response to MC903 treatment, as indicated by higher EASI scores and TEWL values, more severe epidermal hyperplasia, dermal cell and neutrophil infiltration, decreased expression of terminal differentiation markers, and elevated expression of AD-related or neutrophil-related genes such as Tslp, Il33, Il13, Il6, Cxcl1, Cxcl3, Cxcl5, Cxcl15, Csf3, and Icam1 ([204]Fig. 7, K to O, and fig. S9, G to K). These results imply that inhibition of ELOVL6 function aggravates both psoriasis-like and AD-like skin inflammation. To further understand the role of ELOVL6 in epidermal function, we induced silencing or overexpression of ELOVL6 in HaCaT cells and evaluated the changes in the inflammatory response (fig. S10, A and B). Silencing ELOVL6 in human keratinocytes resulted in increased mRNA expression of neutrophil chemoattractants such as CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL8, CSF3, and ICAM1 in response to M5 stimulation (fig. S10C). In contrast, overexpression of ELOVL6 partially suppressed the elevated expression of these neutrophil chemoattractants in M5-treated keratinocytes, whereas METTL3 activity was inhibited by STM2457 treatment (fig. S10D). On the basis of these data combined with the observed in vivo effect of ELOVL6 inhibition, we deduced that ELOVL6, similar to its upstream regulator METTL3, plays a role in protecting the skin from both psoriasis-like and AD-like inflammatory manifestations, partly by regulating neutrophil chemoattraction in epidermal keratinocytes. PA exacerbates IMQ-induced or MC903-induced skin inflammation To underline the role of ELOVL6 in fatty acid metabolism rewiring in keratinocytes during cutaneous inflammation, we conducted a lipidomics study to compare the fatty acid composition of the epidermis in mice treated with the ELOVL6 inhibitor or its vehicle after IMQ application as well as human keratinocytes after silencing ELOVL6 expression under inflammatory conditions ([205]Fig. 8A and fig. S10E). The predominant fatty acid species in the mouse epidermis were identified as PA, SA, OA, and linoleic acid (LA) ([206]Figs. 5I and [207]8A). Furthermore, the epidermis of the ELOVL6 inhibitor–treated mice showed notably elevated levels of PA, SA, and OA compared to the controls, which was consistent with the changes observed in the epidermis of Mettl3 cKO mice ([208]Figs. 5I and [209]8A). We also found that ELOVL6 knockdown in human keratinocytes resulted in a significant elevation of the levels of C16 fatty acid but not C18 fatty acid. This effect was associated with a marked decrease in the ELOVL6 activity index, which was calculated using the following formula: (SA [C18:0] + OA [C18:1])/(PA [C16:0]) + (palmitoleic acid [C16:1]) (fig. S10, E and F) ([210]31). This implies the importance of ELOVL6 in prolonging the C16 fatty acid profile in human keratinocytes. Moreover, ELOVL6-silenced HaCaT cells had a greater PA composition than control cells, with no significant difference observed in the proportion of SA, EA, or OA (fig. S10E). On the basis of these findings, we hypothesized that PA might play an essential role in cutaneous inflammation induced by the METTL3-m^6A-ELOVL6 axis. Fig. 8. Topical treatment of PA results in worsening of skin inflammation. [211]Fig. 8. [212]Open in a new tab (A) Quantitation of FAs in the epidermis of mice topically treated with ELOVL6-IN or its vehicle using GC-MS. (B) Schematic diagram of strategy for application of PA and IMQ. (C and D) Representative images (C) and PASI score (D) of the mouse skin treated with PA or its vehicle after IMQ treatment for 6 days (C) or at the indicated time (D). (E and F) H&E staining images and their quantifications (F) of mice treated with PA or its vehicle after 6-day IMQ application. (G) Flow cytometry of CD45^+ cells and neutrophils in the lesional skin of mice treated with PA or its vehicle after 6-day IMQ application. (H) RT-qPCR analysis of the expression of indicated genes in the 6-day IMQ-treated skin from mice with PA or its vehicle application. (I) Schematic diagram of strategy for application of PA and MC903. (J and K) Representative images (J) and EASI score (K) of the mouse skin treated with PA or vehicle after application of MC903 for 6 days (J) or at the indicated times (K). (L and M) H&E staining images (L) and their quantifications (M) of the mouse skin treated with PA or its vehicle in 6-day MC903-induced skin inflammation. (N) RT-qPCR analysis of the expression of indicated genes in the 6-day MC903-treated skin from mice with PA or its vehicle application. (O) Proposed model of the regulatory and functional role of the METTL3-m^6A-ELOVL6 axis during skin inflammation. The figure was created with [213]BioRender.com. *P < 0.05; **P < 0.01; ***P < 0.001. Scale bar, 100 μm [(E) and (L)]. The P values were determined by unpaired, two-tailed Student’s t test [(A), (F), (G), (H), (M), and (N)] or two-way ANOVA [(D) and (K)]. Data are presented as means ± SEM. Previously, PA, but not myristic acid (C14:0), palmitoleic acid (C16:1), or OA, has been shown to augment phorbol 12-myristate 13-acetate–induced neutrophilic folliculitis in mice when topically applied ([214]32). Moreover, PA could promote the secretion of proinflammatory cytokines, such as IL-6, IL-1, and TNF-α, in human keratinocytes ([215]33). Our detection of increased levels of PA in the METTL3-deficient or ELOVL6-deficient epidermis ([216]Figs. 5I and [217]8A) prompted us to investigate if topical application of PA exacerbates IMQ-induced or MC903-induced skin inflammation. Topical application of PA remarkably aggravated the skin symptoms induced by either IMQ or MC903, which were manifested in the form of clinical sores, disruption of barrier function, epidermal proliferation, dermal cell infiltration, and keratinocyte differentiation ([218]Fig. 8, B to F and I to M, and fig. S11, A to D and G to I). Moreover, PA treatment resulted in a significant increase in the expression of both inflammation-related genes (such as Il6, Mmp9, Cxcl2, Cxcl5, Cxcl15, and Csf3) and the abundance of neutrophils in IMQ-induced skin lesions ([219]Fig. 8, G and H, and fig. S11, F and E). Similarly, PA application was found to enhance neutrophil accumulation as well as the expression of inflammatory factors in MC903-induced AD-like skin inflammation ([220]Fig. 8N and fig. S11, J and K). PA treatment of HaCaT cells led to elevated expression of neutrophil chemotaxis/adhesion–related genes (fig. S11L). Together, these data suggest that epidermal keratinocytes respond to elevated levels of PA by up-regulating the expression of genes involved in neutrophil recruitment and, consequently, cutaneous inflammation. Overall, our results provide strong evidence for the down-regulation of the epidermal METTL3-m^6A-ELOVL6-PA axis in cutaneous inflammation and underscore the normalization of this axis to be an important aspect of therapeutic corrections for inflammatory skin diseases such as psoriasis and AD. DISCUSSION Our findings highlight the crucial involvement of an m^6A modification–fatty acid metabolism pathway detected in epidermal keratinocytes in preserving skin homeostasis and mitigating excessive inflammation in response to disease-inducing stimuli. More specifically, we suggest a working model in which METTL3, expressed by keratinocytes in the healthy skin, is responsible for maintaining appropriate levels of m^6A modification on critical target genes, most notably ELOVL6 mRNA, that regulate fatty lipid metabolism. In disease states such as psoriasis and AD, decreased epidermal METTL3 expression as a result of genetic deletion and/or aberrant inflammation results in decreased ELOVL6 mRNA levels, probably due to an m^6A/IGF2BP3-dependent decrease in stability. Consequently, PA extension is impeded and it accumulates, triggering the up-regulation of genes related to neutrophil recruitment or cell adhesion and culminating in enhanced neutrophil aggregation in the skin. These events collectively contribute to the escalation of the skin inflammatory cycle and aggravation of disease severity ([221]Fig. 8O). Recently, accumulating research has demonstrated the impact of m^6A modification in psoriasis and AD. It seems that the role of m^6A modification in cutaneous inflammation is dependent on cell types. For example, a proinflammatory effect of m^6A modification was demonstrated in γδT cells and CD4^+ T cells ([222]10, [223]11), whereas an anti-inflammatory function was identified in macrophages and keratinocytes that was a result of alteration of the fates of noncoding RNAs ([224]34–[225]36). Furthermore, a recent study reported the aggregated IMQ-induced psoriasis-like skin inflammation in Mettl3^+/− mice compared to WT controls ([226]9). Our study not only supports the protective role of Mettl3 and m^6A in murine epidermal keratinocytes but also shows that this protective role is so crucial that its loss in keratinocytes can be sufficient to initiate skin inflammation even in the absence of any external stimuli. This is evident from the formation of spontaneous psoriasis-like and AD-like lesions in Mettl3-deficient mice. Furthermore, our data reveal mRNAs as important targets of Mettl3-mediated m^6A modification in keratinocytes. Our findings propose a mechanism by which the absence of m^6A modification in epidermal keratinocytes promotes cutaneous inflammation, at least in part, via disruption of lipid metabolism, particularly fatty acid metabolism. However, it should be noted that the ability of m^6A modifications to modulate fatty acid metabolic reprogramming is not unprecedented ([227]37–[228]40), and disruptions in fatty acid metabolism are known to contribute to the development of various inflammatory diseases, including asthma, rheumatoid arthritis, and autoimmune skin diseases such as psoriasis and AD ([229]41–[230]44). However, our work pinpoints m^6A modification as an important means to regulate fatty acid metabolism in the context of skin inflammation. We have identified ELOVL6, a member of the elongation of very-long-chain fatty acids (ELOVL) family of enzymes, as a key target of METTL3-mediated m^6A modification and have provided in vivo evidence for its functional involvement in suppressing psoriasis-like and AD-like skin inflammation. The ELOVL family of enzymes plays a role in the formation and maintenance of healthy skin structure and improvement of skin barrier function by catalyzing the formation of long-chain fatty acids ([231]45). Accordingly, depletion of Elovl6 has previously been shown to aggravate the mechanical damage to the skin and subsequently cause dermatitis by promoting the cellular death of keratinocytes ([232]46). ELOVL6 is known to be responsible for the elongation of PA (C16:0) and palmitoleate (C16:1n-7) ([233]22). Consistent with this known biochemical activity, we found that inhibition of ELOVL6 promoted PA accumulation in the epidermis. While we cannot exclude barrier dysregulation and keratinocyte death as possible causes of the exacerbated inflammation and neutrophil aggregation based on the findings so far, our results do indicate that the effects of PA application closely resemble those of METTL3 deficiency or ELOVL6 deficiency at the tissue, cellular, and molecular levels and suggest a more direct mechanism of PA downstream of the METTL3-m^6A-ELOVL6 axis that regulates epidermal-neutrophil cross-talk and neutrophil chemotaxis during skin inflammation. Our in vitro experiments with human keratinocytes support this notion and implicate a keratinocyte-intrinsic mechanism whereby PA induces the expression of neutrophil chemotaxis/adhesion–related genes in these cells. These data propose an innovative mechanism for regulating metabolism and immunity in the context of skin conditions. Neutrophilic inflammation is a characteristic feature of both psoriasis and AD pathologies ([234]47). The number of neutrophils in the lesional skin from patients with psoriasis and AD was higher than that in the healthy skin, and both were correlated with the expression levels of neutrophil chemoattractant genes ([235]48). Although the role of neutrophils in AD is still under debate, a growing number of recent studies indicate that neutrophils may play a crucial role in chronic recurrent AD ([236]49, [237]50). In the present study, down-regulation of the METTL3-m^6A-ELOVL6 axis was detected in the epidermal keratinocytes of patients with psoriasis vulgaris, psoriasis pustulosa, and AD. These findings are consistent with our observation of increased neutrophil infiltration in the inflamed skin of Mettl3 cKO mice, ELOVL6 inhibitor–treated mice, and PA-treated mice. Thus, the METTL3-m^6A-ELOVL6 axis may be instrumental in driving inflammation in human patients with skin diseases associated with neutrophil involvement. The anti-inflammatory properties of ELOVL6 are not restricted to the skin as similar effects have also been observed in allergic airway inflammatory conditions such as asthma ([238]26). Thus, the regulatory mechanisms elucidated in this study may extend beyond the skin and have broad implications for autoimmune diseases affecting other cell types and tissues too. Consistent with our findings, previous studies have also indicated higher levels of PA in the serum of patients with psoriasis than in the serum of healthy controls ([239]51, [240]52). Similarly, an inclination toward elevated PA levels was observed in the epidermis of lesioned skin in patients with AD compared to nonlesional skin ([241]53). Application of fatty acids, including PA, has been associated with hyperkeratinization and exacerbation of neutrophilic folliculitis ([242]32). These studies further corroborate the role of PA in autoimmune skin disorders. In addition, a study reported that deficiency in METTL3-mediated m^6A modification in tumor cells enhances the recruitment of tumor-associated neutrophils and, consequently, affects tumor pathogenesis ([243]54). However, if abnormalities in lipid metabolism contribute to this process remains unexplored. Hence, our study may serve as a useful reference for further mechanistic studies in cancer and other inflammatory diseases that are characterized by neutrophil involvement. Our analysis of MeRIP-seq data reveals that, in addition to genes related to fatty acid metabolism, one gene involved in ceramide synthesis, such as Cers4, may also play a significant role in skin inflammation regulated by RNA m^6A methylation in keratinocytes ([244]Fig. 6A). Ceramides, which are essential components of skin lipids, have been linked to various inflammation conditions, including asthma, Alzheimer’s disease, and skin inflammation ([245]55–[246]58). In inflammatory skin diseases including psoriasis and AD, aberrant ceramide expression profiles are recognized for their disruptive effects on epidermal barrier maintenance, epidermal self-renewal, and cutaneous immune responses ([247]59). Future investigations will further extend the repertoire of m^6A-regulated genes that contribute to skin inflammation, enhancing a comprehensive understanding of m^6A modification in skin inflammation. Overall, our findings underscore the crucial role of disrupted m^6A modification in keratinocytes and consequent reprogramming of fatty acid metabolism and increased neutrophil accumulation in autoimmune skin disorders such as psoriasis and AD. These insights have important implications for potential therapies targeting m^6A modification in such diseases as well as other conditions related to epithelial inflammation. MATERIALS AND METHODS Patients The skin samples used in this study were obtained from patients with psoriasis vulgaris, patients with psoriasis pustulosa, patients with AD, and healthy donors. RNA extraction or paraffin sectioning was performed on these samples. The procedure for the collection of samples, including skin biopsies, was approved by the Ethical Review Committees of Shanghai Skin Hospital (no. 2020-04) and Shanghai Tenth People’s Hospital (no. 2021KN114). Informed consent was obtained for all the procedures. All the patients were volunteers and did not receive compensation for their participation in this study and provision of their skin samples. Mice and tamoxifen-mediated deletion of Mettl3 in mice C57BL/6J mice were obtained from the Shanghai Laboratory Animal Center. Mettl3^fl/fl mice were generated by Shanghai Model Organisms Center Inc. The KRT14-CreER^T2 transgenic mice (catalog no. NM-KI-190024) were obtained from Shanghai Model Organisms Center Inc. Mettl3^fl/fl mice were crossed with the KRT14-CreER^T2 transgenic mice to generate KRT14-CreER^T2; Mettl3^fl/fl mice. To achieve conditional KO of Mettl3, 5-week-old KRT14-CreER^T2 and KRT14-CreER^T2; Mettl3^fl/fl; mice were intraperitoneally injected with tamoxifen (catalog no. T5648, Sigma-Aldrich) (75 μg per g body weight), which is dissolved in corn oil, for five consecutive days. After 9 days of rest, mice were euthanized to test the KO efficiency. All animal studies were performed with sex-matched and age-matched mice. Mice were housed in groups with a 12-hour light/12-hour dark cycle. They had ad libitum access to food and water. All animal handling and surgical procedures were conducted according to national guidelines and approved by the Tongji University Animal Care and Use Committee (protocol no. 2020-029). Histological analysis and immunofluorescence staining For histological analysis, skin tissue was fixed in 4% paraformaldehyde, embedded in paraffin, sectioned (4 μm), and stained with hematoxylin and eosin (H&E). Sites were randomly selected and evaluated using the ImageJ 1.52a software for analysis of epidermal hyperplasia (acanthosis) or dermal cell infiltration (number of dermal cells per 10,000 μm^2 of tissue). For immunohistochemical analyses, paraffin-embedded sections were deparaffinized in xylene and rehydrated before antigen retrieval using boiling in an EDTA buffer [1 mM EDTA containing 0.05% Tween 20 (pH 9.0)]. Next, the sections were incubated in H[2]O[2] for 10 min, permeabilized with 0.5% Triton X-100 for 15 min, and washed with phosphate-buffered saline (PBS), and this was followed by blocking in 3% bovine serum albumin in PBS at room temperature for 1 hour and overnight staining at 4°C with primary antibodies. The next day, a two/three-color fluorescence kit (Recordbio Biological Technology, Shanghai, China) was used to stain the sections based on the tyramide signal amplification technology according to the manufacturer’s instructions. The fluorescence-stained sections were then mounted in ProLong Gold Antifade reagent with DAPI (4′,6-diamidino-2-phenylindole) (catalog no. 4083, Cell Signaling Technology) and visualized under a microscope (Leica). Detailed information about the antibodies and their usage in this study is presented in table S3. RNA isolation and RT-qPCR The skin tissues were homogenized in TRIzol (Takara) after they were ground in the presence of liquid nitrogen, while the cells were resuspended directly in TRIzol. Total RNA was isolated using the RevertAid First Strand cDNA Synthesis Kit (Takara) according to the manufacturer’s instructions and reverse transcribed into cDNA. RT-qPCR was performed using SYBR Green master mix (Takara) on a StepOnePlus Real-Time PCR System (Applied Biosystem). Western blotting Skin tissues and cells were lysed in a radioimmunoprecipitation buffer (catalog no. 89900, Invitrogen) with a protease inhibitor (catalog no. 78442, Thermo Fisher Scientific). The lysates were heated at 99.9°C for 10 min, and the protein samples were separated by SDS–polyacrylamide gel electrophoresis gel and transferred to polyvinylidene membranes (catalog no. IPVH00010, Millipore). The membranes were blocked with 5% nonfat milk prepared in Tris buffer saline plus 0.1% Tween 20 (TBST) for 1 hour at room temperature and then incubated with primary antibodies overnight at 4°C, and this was followed by incubation with goat anti-rabbit IgG (H+L) or goat anti-mouse IgG (H+L) as secondary antibodies for 1 hour the next day. Signals from blotted membranes were detected with the Odyssey Imaging System after the membranes were washed three times with TBST. Detailed information about the antibodies and their usage in this study is presented in table S3. Epidermis separation Dorsal skin samples were excised from adult mice and incubated with Dispase II (3 mg/ml; catalog no. 17105041, Thermo Fisher Scientific) for 16 hours at 4°C, and then the epidermis were separated from the dermis under a microscope using a glass slide. RNA sequencing For RNA-seq, Ribo-Zero Gold (Illumina) was used for cDNA library construction. To construct the library, the generated cDNAs were amplified by PCR. The libraries were validated on the Agilent 2100 bioanalyzer and sequenced on NovaSeq 6000 (Illumina). MeRIP sequencing MeRIP-seq was performed based on a previously described protocol ([248]60). In brief, total RNA from the skin tissue of patients or the epidermis of mice together with m^6A+ control RNA (GLuc) and m^6A− control RNA (CLuc) was randomly fragmented to 200 to 500 nt through sonication (Covaris S220). Subsequently, 1/20 volume of the fragmented RNA was retained as input RNA for total RNA-seq library construction. The rest was incubated with Protein A beads (catalog no. 10001D, Thermo Fisher Scientific) that were coupled with the polyclonal anti-m^6A antibody (catalog no. E1610S, NEB) in an immunoprecipitation (IP) buffer [10 mM tris-HCl, 150 mM NaCl, RNasin (4 U/μl) (pH 7.4), and 0.1% NP-40] for 4 hours. After IP, the RNA reaction mixture was washed twice in the IP buffer, twice in a low-salt IP buffer [50 mM NaCl, 10 mM tris-HCl (pH 7.4), and 0.1% IGEPAL CA-630 (catalog no. I8896, Sigma-Aldrich) in nuclease-free H[2]O], and twice in a high-salt IP buffer [500 mM NaCl, 10 mM tris-HCl (pH 7.4), and 0.1% IGEPAL CA-630 in nuclease-free H[2]O) for 5 min each time. RNA extraction followed by phenol-chloroform extraction and ethanol precipitation was performed to purify the m^6A-enriched RNA fragments. Last, the purified RNA from IP and input RNA was subjected to RT-qPCR or library construction through the SMARTer Stranded Total RNA-Seq Kit version 2 (Takara, 634411). The libraries were sequenced on an Illumina NovaSeq 6000 platform (Novogene and Nanjing Jiangbei New Area Biophamaceutical Public Service Platform). SN ratio calculation for MeRIP-qPCR The following equation was used to calculate the Signal-to-Noise (SN) ratio: SN ratio = 2^(−Ct[m6A-positive or target gene in IP] + Ct[m6A-positive or target gene in input])/2^(−Ct[m6A-negative gene in IP] + Ct[m6A-negative gene in input]). IMQ-induced psoriasis mouse model and MC903-induced AD mouse model For the IMQ-induced psoriasis mouse model, mice aged 7 weeks were treated topically with a daily dose of 62.5 mg of IMQ cream (5%) (Shichuan MedShine Pharmaceuticals Co.) that was applied to the shaved back for the indicated time. For the MC903-induced AD mouse model, 1 nmol of MC903 (catalog no. HY-10001, MedChemExpress), dissolved in 10 μl of ethanol, was applied daily to the ears of 7-week-old mice. For dorsal application, 4 nmol of MC903, dissolved in 40 μl of ethanol, was applied daily to the backs of the mice for the specified duration. TEWL test TEWL was measured on the dorsal skin of mice using the GPSkin Barrier device at the indicated time points prior to IMQ or MC903 treatment. Measurements were performed in triplicate for each time point, and mean values were calculated for further analysis. Skin cell preparation and flow cytometry To obtain whole skin cell suspension, the skin of mice was minced with fine scissors after removal of subcutaneous fat. Cells were digested with 10 ml of RPMI 1640 medium (catalog no. 11875093, Gibco) containing 0.25% collagenase (catalog no. C9091, Sigma-Aldrich), 0.01 M Hepes (catalog no. BP310, Thermo Fisher Scientific), 0.001 M sodium pyruvate (catalog no. BP356, Thermo Fisher Scientific), and DNase (deoxyribonuclease) (0.1 mg/ml; catalog no. DN25, Sigma-Aldrich) for 1 to 1.5 hours at 37°C with rotation, and 1 ml of fetal bovine serum (FBS) was added per sample to terminate the digestion. Cells were then filtered through a 40-μm filter and were spun down and resuspended in a fluorescence-activated cell sorting buffer (2% FBS in PBS). The skin cells were incubated with TruStain FcX antibody to block Fc receptors for 10 min at room temperature. Isolated cells were stained with different cell surface markers (CD45 for leukocytes; CD45^+ and CD3^+ for T cells; CD45^+, CD11b^+, and Ly6G^+ for neutrophils; CD45^+, CD11b^+, and F4/80^+ for macrophages; CD45^+ and CD11c^+ for DCs; and CD45^+, CD90.2^+, and γδT cell receptor [TCR^+] for dermal γδT cells or DETCs) for 30 min at 4°C. The cells were then incubated with the SYTOX Blue Dead Cell Stain (catalog no. S34857, Invitrogen) for 5 min, and 10,000 counting beads (catalog no. [249]C36950, Invitrogen) were added per sample before analysis. Skin cells were analyzed using the FACSAria Fusion Sorter (BD Biosciences). Data were analyzed using the FlowJo v.10 software (TreeStar). Detailed information about on the antibodies and their usage in this study is provided in table S3. In vivo neutrophil depletion For the neutrophil depletion assay, 7-week-old Mettl3 cKO mice were treated with 400 μg per 20 g body weight of a rat anti-mouse Ly6G antibody (catalog no. BP0075-1, BioXCell) or rat IgG (catalog no. BP0089, BioXCell) a day before IMQ treatment, and this was followed by treatment with 100 μg per 20 g body weight of a rat anti-mouse Ly6G antibody or rat IgG every other day starting from 1 day after IMQ treatment until the end of the experiment. The effectiveness of neutrophil depletion was assessed by flow cytometry in the mouse skin tissue. RIP assay RIP assay was performed using the Magna RIP Kit (Catalog no. 17-700, Merck) according to the manufacturer’s instructions. Briefly, 1.5 μg each of an anti-IGF2BP3 antibody and anti-rabbit IgG was incubated with 50 μl of magnetic beads before adding cell lysates were added (~1 × 10^7 cells per sample). Then, the RNA-protein IP complexes were washed six times and incubated with proteinase K digestion buffer to remove proteins. Last, RNA was extracted by phenol-chloroform RNA extraction and purified for RT-qPCR analysis. Normalization of the relative enrichment was performed using the following formula: %Input = 1/10 × 2^Ct [IP] − Ct [input]. Detailed information about the IGF2BP3 antibodies and their usage in this study is presented in table S3. Human keratinocyte culture, M5 stimulation, and STM2457 treatment In this study, adult HEKa were isolated from human foreskin and cultured in EpiLife medium (Gibco) containing EpiLife Defined Growth Supplement (Gibco) and 0.06 mM Ca^2+ (Gibco). Cells at passages 3 to 5 were used for subsequent experiments. The immortalized human keratinocyte cell line HaCaT cells (catalog no. CBP6033, Cobioer) were used for human keratinocyte-related research. Cells were cultured in RPMI 1640 medium (Gibco) containing 10% FBS (Gibco), penicillin (50 U ml^−1), and streptomycin (50 μg ml^−1) (Gibco) under standard culture conditions. Cells were cultured at 37°C and 5% CO[2]. The cytokine cocktail used to stimulate the cells in this study is M5, which is a cocktail of recombinant human IL-1α (catalog no. 200-01A, PeproTech), IL-17A (catalog no. 200-17, PeproTech), IL-22 (catalog no. 200-22, PeproTech), oncostatin M (catalog no. 300-10, PeproTech), and TNF-α (catalog no. 300-01A, PeproTech), each at 10 ng ml^−1. For STM2457 treatment, 10 μM STM2457 (catalog no. HY-134836, MedChemExpress) was used to treat HaCaT cells for 48 hours before collection. m^6A ELISA RNAs of HaCaT cells treated with STM2457 or DMSO for 48 hours was isolated as described above. m^6A modification on RNAs was measured with the EpiQuik m^6A RNA Methylation Quantification Kit (catalog no. P-9005, EpigenTek) according to the manufacturer’s instructions. ELISA on all samples was performed at the same time. Topical application of MP3C or the ELOVL6 inhibitor For topical treatment with MP3C (catalog no. M193627, Aladdin Biochem), it was applied at 10 mg/kg daily for 5 days. C57BL/6J mice (7 weeks old at the start of the study) were treated topically with MP3C or its vehicle (ethanol, EtOH) daily for 5 days daily after treatment with a single application of IMQ or MC903 until the end of the study (6 to 8 hours before each application of IMQ or MC903). For ELOVL6 inhibitor (ELOVL6-IN) treatment, 2 μg of ELOVL6-IN-2 (catalog no. HY-12146, MedChemExpress) (diluted with DMSO) was mixed with 2 mg of oil-in-water cream per gram of mouse body weight, as required for immediate application. C57BL/6J mice (7 weeks old at the start of the study) were treated topically with mixed ELOVL6-IN cream or vehicle for 3 days as a pretreatment procedure, and then it was applied daily until the end of the study (6 to 8 hours before each application of IMQ or MC903). Human skin biopsy cultures Skin biopsy samples were processed according to a previously published study ([250]61). Briefly, human skin biopsy samples from healthy donors were incubated with or without 1 mM MP3C and M5/PBS for 6 hours after 2 hours of starvation. For culture of psoriatic lesions, each skin biopsy specimen was equally divided into two equal portions, each containing epidermal and dermal structures, and then treated with or without 1 mM MP3C for 24 hours. The specimens were then cultured in RPMI 1640 medium (Gibco) containing 10% FBS (Gibco), penicillin (50 U ml^−1), and streptomycin (50 μg ml^−1) (Gibco). After the treatment, the specimens were snap frozen in liquid nitrogen and then subjected to RNA isolation. Gene silencing experiments For ELOVL6 and IGF2BP1/2/3 silencing in HaCaT cells, small interfering RNAs (siRNAs) targeting human ELOVL6, IGF2BP1, IGF2BP2, or IGF2BP3 were synthesized by Hippo Biotechnology Co. Ltd. (Huzhou, China). Using the Lipofectamine RNAiMAX Reagent (Invitrogen), siRNAs targeting ELOVL6 and IGF2BP1-3 (table S2) were transfected into HaCaT cells according to the manufacturer’s instructions. Silencing efficiency was analyzed by RT-qPCR and Western blotting. RNA stability assay Cells were seeded in 12-well plates overnight. After being pretreatment with STM2457 or DMSO for 48 hours, the cells were then treated with actinomycin D (5 μg/ml; S8964, Selleckchem) for 0, 3, or 6 hours. Total RNA was isolated using TRIzol and quantified by RT-qPCR. Fatty acid composition Lipids from the mouse epidermis were extracted according to the method of Bligh and Dyer as previously described ([251]62). Briefly, chloroform/methanol (1:2, v/v) was used to extract the epidermis. To break the monophase, 1 M each of NaCl solution and chloroform was added. The samples were incubated on ice for a 10 min and centrifuged at 300g for 5 min. Then, the aqueous solution was discarded, and the chloroform phase was evaporated with nitrogen gas. Samples were incubated at 100°C for 45 min after addition of acetonitrile/6 N HCl (90:10, v/v). Liquid-liquid extraction ([252]63) with ethyl acetate was lastly performed, and the reconstituted samples were injected into an optimized gas chromatography–mass spectrometry (GC-MS) system. RNA-seq and MeRIP-seq analysis RNA reads with adaptors and of low quality were removed using Trim_galore. Clean reads were aligned to the reference genome (UCSC hg38) using the STAR software. As described previously ([253]60), m^6A-modified spike-in (Gluc) was used to normalize mouse input and IP signals. The normalization of the genome coverage BigWig files of input and IP signals was performed as previously described ([254]60). Peak calling was first performed using MetPeak, and the peaks obtained were further filtered by calculating the IP/input signal ratio using scaled genome coverage BigWig files. Peaks with IP/input ratios > 1.5 were retained for further analysis. Differential gene expression analysis in this study was performed using the R package DESeq2 DiffBind v3 and was used to identify differential m^6A peaks. In our RNA-seq analysis, we determined DEGs using cutoffs: an absolute log[2] FC (|log[2]FC|) ≥ 1 and a significance level of adjusted P value < 0.05. In our MeRIP-seq analysis, we determined differentially methylated peaks (DMPs) using cutoffs: an absolute log[2] FC (|log[2]FC|) ≥ 1 and a significance level of FDR (false discovery rate) < 0.05. Statistics and reproducibility The in vivo experiments were repeated three times independently. The symbols in the figures indicate the exact n for each experimental group/condition. Each symbol represents an individual mouse. The in vitro experiments were performed in triplicate and were repeated two or three times independently of each other. To avoid technical bias, each sample was processed identically in each experiment, and internal controls and normalization methods were included. All data are presented as means ± SEM. The significance of differences between two groups was determined using unpaired, two-tailed Student’s t test, and one-way or two-way analysis of variance (ANOVA) was used for multiple group analyses using GraphPad Prism v.9. ACKNOWLEDGMENTS