Abstract Sexual dimorphism is a fundamental characteristic of various physiological and pathological processes in humans, including immune responses, senescence, and metabolism. Most studies on the sex bias have focused on sex hormones or female‐biased genes, whereas male‐biased genetic factors remain understudied. Here, we show that the Y‐linked noncoding RNA, RP11‐424G14.1, is expressed in human male keratinocytes. Microarray study suggests the NF‐κB pathway as the top biological pathway affected by RP11‐424G14.1 knockdown, consistent with known sex differences in inflammation. Additionally, IGFBP3 is identified as the top gene supported by RP11‐424G14.1 in male keratinocytes. Conversely, in female keratinocytes, IGFBP3 is the top gene repressed by the X‐linked long noncoding RNA XIST, suggesting a central role of IGFBP3 in mediating sexual dimorphism. Knockdown of RP11‐424G14.1 or IGFBP3 in male keratinocytes inhibits cellular senescence, consistent with increased longevity in females. IGFBP3 expression is dependent on insulin, and metabolomics analysis suggests that RP11‐424G14.1 and IGFBP3 regulate acrylcarnitine metabolism. Our study identifies the role of the RP11‐424G14.1‐IGFBP3 pathway in coordinating sex differences in immunity, senescence, and metabolism. With RP11‐424G14.1 being a human‐specific genetic element, our study suggests the evolving feature of sexual dimorphisms in biological processes. Keywords: inflammation, metabolism, noncoding RNA, senescence, sexual dimorphism __________________________________________________________________ Sexual dimorphism is a fundamental characteristic of various physiological and pathological processes in humans. Here we identify a human‐specific, Y‐linked noncoding RNA, RP11‐424G14.1, in male keratinocytes. Transcriptomic analysis shows that RP11‐424G14.1 regulates IGFBP3 and the NF‐κB pathway, affecting male‐biased cellular senescence and immunosuppression. Metabolomic study identifies acrylcarnitine metabolism as a specific pathway downstream of RP11‐424G14.1‐IGFBP3, suggesting coordinated regulation of cellular aging, immune activity, and lipid metabolism. graphic file with name FBA2-7-e1479-g007.jpg 1. INTRODUCTION Sexual dimorphism is a fundamental characteristic of various physiological and pathological processes in humans. Generally, it is accepted that females mount stronger innate and adaptive immune responses than males, which is supported by the clinical observation of strikingly increased prevalence of autoimmune diseases in females.[26] ^1 , [27]^2 , [28]^3 In human populations, women are known to live longer than men, consistent with lower biological ages as assessed by molecular markers.[29] ^4 , [30]^5 Furthermore, sex differences have been identified in many aspects of energy balance regulation including glucose metabolism and insulin sensitivity as well as lipid storage and utilization patterns, which may influence the pathogenesis of diseases including obesity and diabetes.[31] ^6 , [32]^7 Studies on the molecular mechanisms underlying sex differences in human physiology have been focused on sex hormones.[33] ^8 , [34]^9 , [35]^10 Additionally, gene‐dosage effects of the X‐ and Y‐chromosomes have been described and on the single‐gene level, sex‐biased, protein‐coding genes have been identified with roles in regulating sexually dimorphic processes.[36] ^11 , [37]^12 , [38]^13 , [39]^14 The map of sexually dimorphic genetic elements is just starting to unfold, as protein‐coding regions only make up approximately 1 percent of the human genome and the rest 99 percent is noncoding.[40] ^15 The active transcription of non‐protein coding genes gives rise mainly to long non‐coding RNA (lncRNAs, 80%–90%).[41] ^16 Long non‐coding RNA (lncRNA) are typically transcripts longer than 200 nt that do not code for proteins but may still serve a diverse array of functions.[42] ^17 They may regulate expression epigenetically by coordinating with DNA or histone‐modifying enzymes or their products and can also directly affect transcription by interacting with or acting as transcription factors. Post‐transcriptionally, lncRNA can inhibit or coordinate with small interfering (siRNA) or microRNA (miRNA).[43] ^18 Though not as common, they are also capable of post‐translational modification.[44] ^19 The importance of lncRNA in sex‐biased expression has only started to gain traction in the last decade. One of the most studied in this regard is X‐inactive specific transcript, XIST, known for its role in female development through X‐chromosome inactivation (XCI).[45] ^20 During XCI, XIST RNA triggers gene silencing and drives a major structural reorganization to shut down one copy of the X‐chromosome, which encodes for various immune‐associated genes, in females. XIST overexpression has been associated with systemic autoimmune disease including systemic lupus erythematosus (SLE) and rheumatoid arthritis, both of which are female‐biased.[46] ^21 Beyond immune regulation, lncRNA is implicated in both glucose and lipid metabolism, with the focus being XIST as well.[47] ^22 To the best of our knowledge, little is known about the function of other lncRNAs in mediating sexual dimorphism. To this end, in this study we have address the expression and function of RP11‐424G14.1, a lncRNA of 2666 bp length encoded by the Y chromosome. Intriguingly, it is also a human‐specific genetic elements and does not show homology in alignment with the rhesus monkey, mouse, or other sequenced vertebrates. We confirm the male‐specific expression of RP11‐424G14.1 in human keratinocytes and identify its multifaceted function in mediating sex differences in inflammation, senescence, and metabolism. 2. MATERIALS AND METHODS 2.1. Keratinocyte culture and transfection ATCC primary epidermal keratinocyte cultures were grown in Keratinocyte Growth Medium, or KGM, (Lifeline Cell Technology) on labeled 10 cm cell culture‐treated dishes. KGM was refreshed after seeding and every other day after during culture. Upon sufficient cell growth and 70%–90% cell confluency, keratinocytes were collected via trypsinization, counted by hemocytometer, and transfected on the Lonza 4‐D X unit nucleofector according to P3 Lonza keratinocyte nucleofection protocols (V4XP‐3024). If the cells were treated with immunostimulants, they (and their controls) were given 24 h post‐nucleofection to recover. 24 h post‐treatment, cells were either harvested for RNA or protein, or were collected and used for experiments. siRNA used was purchased through Thermofisher: Silencer Select negative control siRNA (referred to as scrambled siRNA), pre‐designed siRNAs for XIST (assay ID: n272634) or IGFBP3 (assay ID: s7228), or custom designed for RP11‐424G14.1 (GGGAAGUGUUUGUACAGUATT and UACUGUACAAACACUUCCCGA). 2.2. qPCR RNA isolated from keratinocyte culture either by Trizol extraction or Miniprep was quantified and used to make cDNA with reverse transcriptase. Standardized amounts of cDNA template were pipetted into 96‐well plates with respective primers, Power SYBR Green master mix, and nuclease‐free water. Optical adhesive film was used to cover the plates, then centrifuged and run on a QuantStudio 6 at standard speed for 40 cycles. Differential gene expression was calculated using the ΔΔct method. 2.3. Microarray mRNA was isolated from cultured male and female keratinocytes using Trizol following manufacturer instructions. mRNA was hybridized to the Thermofisher human Clariom S array. Raw signals were obtained by the GeneChip Scanner and analyzed by the Transcriptome Analysis Console Software according to standard procedures. Differentially expressed genes were identified using a false discovery rate of 0.05 and a fold change of 2. 2.4. Metabolomics Keratinocyte lysates were profiled using Biocrates MxP Quant500 plates with LC‐ and FIA‐MS following manufacturer instructures. Metabolites were identified, quantified, and normalized by the MetIDQ workflow following standard analysis workflows. Differentially expressed metabolites were identified using an adjusted p‐value of 0.05. 2.5. Immunofluorescence staining and imaging Keratinocytes were transferred to 8‐well chamber slides after transfection at a seeding density of 70,000–100,000 per well. After control or target gene knockdown such as XIST, RP11‐424G14.1, or IGFBP3 knockdown (oligo info specified in the ‘keratinocyte culture and transfection’ section), cells were washed with PBS, fixed with 4% paraformaldehyde (PFA) for 15 min, and permeabilized with 0.1% Triton X‐100 for 5 min. Cells were then washed in wash buffer (PBS with 0.1% Triton X‐100) for 5 times and stained with the respective primary antibodies for 1 h at room temperature at manufacturer‐recommended dilutions (IGFBP3, Thermofisher, 10,189‐2‐AP; NFkB, Cell Signaling Technologies, 3033; pIkBa, Cell Signaling Technologies, 2859; p21, Cell Signaling Technologies, 2947; THBS2, Thermofisher, MA5‐46953; TSPAN1, Thermofisher, PA5‐119458; WNK1, Abcam, ab174854), followed by 5 washes. Cells were subsequently stained the respective secondary antibodies at manufacturer‐recommended dilutions (goat anti‐rabbit IgG (H + L) Alexa Fluor 488, Thermofisher, A11008; donkey anti‐mouse IgG (H + L) Alexa Fluor 488, Thermofisher, A21202) for 1 h. Cells were then washed with milliQ water, stained with 1 μg/mL DAPI for 5 min, and washed again. Upon completion of staining, the cells were then covered with Vectashield antifade mounting medium and imaged on a Nikon TI‐2 E inverted fluorescent microscope. The same acquisition parameters, including laser intensity and exposure settings, were used for all conditions in comparison. 2.6. Western blot Keratinocytes were transferred to 6‐well tissue culture‐treated plates after transfection. After treatments, cells were washed with ice‐cold PBS and placed on ice. Cells were then treated with RIPA buffer and scraped to isolate protein. Cell‐lysate protein concentration was measured for each sample using a bicinchoninic acid assay with bovine serum albumin for a standard on an Agilent BioTek Epoch plate reader and Take3 plate. For each sample, 35 μL were loaded into wells on a precast mini‐PROTEAN gel, containing 20 μg protein, 8.75 μL 4× Laemmli buffer, and milliQ water. 4 μL of BioRad Protein Precision Plus standard ladder were loaded into surrounding wells. Gels were run in a BioRad mini‐PROTEAN tetra cell with Tris‐glycine‐SDS running buffer at 100 V for 2 h. Upon separation, gels were then equilibrated and transferred to an activated PVDF membrane for blotting, according to the BioRad transfer manual. Blots were then cut based on ladder‐indicated size ranges to capture protein targets for efficient antibody probing. Blots were rinsed in Tris buffered saline with 0.1% Tween‐20 (TBS‐T) and then blocked in 5% non‐fat dry milk in TBS‐T for 1 hour at RT. Primary and HRP‐linked secondary antibodies were diluted to indicated concentrations. Blots were incubated with indicated primary antibodies for 1 h at RT and then washed 3x in TBS‐T. Blots were then stained with respective secondary antibodies for 1 h at RT and then washed 5x in TBS‐T and 1x in TBS. Stained blots were marked with WesternSure pen to identify marker lanes upon imaging. Stained blots were incubated with LI‐COR WesternSure chemiluminescent substrate for 5 min and placed in a laminated pouch, aspirating any excess solution. Blots were imaged on a C‐Digit, after 12‐min exposure. 3. RESULTS 3.1. Sex‐biased expression of lncRNAs in human keratinocytes To understand the potential roles of lncRNAs in regulating sexual dimorphism in human male keratinocytes, we used qPCR to examine the expression levels of top male‐biased lncRNAs suggested by previous RNA‐sequencing studies using whole skin biopsies.[48] ^14 Of the three candidates tested, including RP11‐372M18.2, RP11‐424G14.1, and RP11‐331F4.1, we were able to detect male‐specific expression of RP11‐424G14.1 in keratinocytes (Figure [49]1A). It is possible that the other two candidates, RP11‐372M18.2 and RP11‐331F4.1, are expressed in other types of skin cells such as fibroblasts instead. Consistent with its male‐specific expression, RP11‐424G14.1 maps to chromosome Y: 21,853,827–21, 856, 492 with no overlap to known protein‐coding genes. The expression level of RP11‐424G14.1, but not the control gene GAPDH, was significantly reduced by knockdown using its sequence‐specific siRNA, suggesting specificity of expression detection (Figure [50]1B, Figure [51]S1). We did not detect a significant association between RP11‐424G14.1 and the age of donors (Figure [52]1A). FIGURE 1. FIGURE 1 [53]Open in a new tab Sex‐biased expression of RP11‐424G14.1 and XIST as well as their downstream targets. (A) Expression levels RP11‐424G14.1 and XIST in female (F) and male (M) keratinocytes (left) by qPCR, showing lack of association with age (right). (B) Specificity of RP11‐424G14.1 detection and knockdown, shown by expression levels of RP11‐424G14.1 and GAPDH upon scrambled RNAi (Scr Ri, as control) or RP11‐424G14.1 sequence‐specific RNAi by qPCR. (C) Venn diagram of genes supported or repressed by RP11‐424G14.1 knockdown in male keratinocytes and XIST knockdown in female keratinocytes compared to respective controls from microarray studies (FDR <0.05 for differentially expressed genes). (D) Expression levels of indicated genes upon scrambled (control) or RP11‐424G14.1 knockdown (KD) in male keratinocytes (top) and upon scrambled (control) or XIST knockdown (KD) in female keratinocytes (bottom) by qPCR. (E) Pathway enrichment analysis of microarray data showing the top 5 pathways impacted by the loss of RP11‐424G14.1 or XIST. FDR, false discovery rate. Mean ± stdev, *p < 0.05, Student's t‐test. As expected, as a lncRNA known to mediate X‐chromosome inactivation in females, XIST RNA is expressed in human keratinocytes in a female‐based manner (Figure [54]1A). The expression level of XIST, but not the control gene GAPDH, was reduced by its sequence‐specific knockdown, suggesting specificity of expression detection (Figure [55]S1). While XIST has been shown to decrease in expression in aged hematopoietic stem cells (HSCs),[56] ^23 we have found that in keratinocytes, the expression level of XIST did not associate with the age of donors (Figure [57]1A). Taken together, these data suggest tissue specificity in the expression change of XIST during aging. Based on these results, we focused our subsequent study on RP11‐424G14.1 in male keratinocytes and XIST in female keratinocytes. 3.2. Genes and pathways regulated by sex‐biased lncRNAs Because little is known about the roles of RP11‐424G14.1 and XIST on a genome‐wide level in human keratinocytes, we performed microarray to identify their downstream genes in an unbiased manner. We performed RP11‐424G14.1 and scrambled control knockdown in human male keratinocytes as well as XIST and scrambled control knockdown in human female keratinocytes (age‐ and race‐matched), followed by microarray analyses. We identified four groups of differentially expressed genes (FDR <0.05), namely genes that were downregulated by RP11‐424G14.1 knockdown in male keratinocytes (i.e., RP11‐424G14.1‐supported), genes that were upregulated by XIST knockdown in female keratinocytes (i.e., XIST‐repressed), genes that were upregulated by RP11‐424G14.1 knockdown in male keratinocytes (i.e., RP11‐424G14.1‐repressed), and genes that were downregulated by XIST knockdown in female keratinocytes (i.e., XIST‐supported). Compared to XIST, RP11‐424G14.1 regulated a restricted set of genes, supporting the expression of 20 genes and repressing the expression of 4 genes (Figure [58]1C). Of the 20 RP11‐424G14.1‐supported genes in male keratinocytes, 4 genes (20%) were repressed by XIST in female keratinocytes, suggesting that these four genes are male‐biased genes that are coregulated by the two lncRNAs (i.e. activated by RP11‐424G14.1 in males and repressed by XIST in females). These four genes are IGFBP3, CXCL14, SEMA5A, and WNK1. Of the 4 RP11‐424G14.1‐repressed genes in male keratinocytes, 2 genes (50%) were supported by XIST in female keratinocytes, suggesting that these two genes are female‐biased genes that are coregulated by the two lncRNAs (i.e., repressed by RP11‐424G14.1 in males and activated by XIST in females). These two genes are TSPAN1 and THBS2. We further validated the co‐regulation of these six genes by RP11‐424G14.1 and XIST by independent qPCR experiments (Figure [59]1D) and summarized their known functions (Table [60]1). TABLE 1. Known functions for co‐targets of RP11‐424G14.1 and XIST as identified by microarray. Gene Known roles Expression IGFBP3 * Negative regulator of IGF signaling.[61] ^39 * Antagonist of TGF‐βII receptor, arresting growth[62] ^34 , [63]^38 , [64]^40 * Facilitate NHEJ.[65] ^41 * Inhibit keratinocyte proliferation.[66] ^42 * Modulate carbohydrate and lipid metabolism.[67] ^43 Male‐biased CXCL14 * Important for differentiation of dendritic cells into Langerhans cells.[68] ^44 * Immune surveillance through chemotaxis.[69] ^45 * Neutrophil, DC, and uterine NK cell chemoattractant.[70] ^46 Male‐biased SEMA5A * Inhibits synaptogenesis during neurogenesis.[71] ^47 * Promotes angiogenesis and inhibits apoptosis.[72] ^48 Male‐biased WNK1 * Ion homeostasis in kidney and nervous system.[73] ^49 * Autophagy inhibition.[74] ^50 * Suppresses NLRP3 inflammasome activation, and therefore inflammation and pyroptosis.[75] ^51 Male‐biased TSPAN1 * Promotes cell migration.[76] ^52 * Suppresses NFκB signaling.[77] ^53 Female‐biased THBS2 * Inhibits angiogenesis.[78] ^54 * Promotes cell motility and migration.[79] ^55 Female‐biased [80]Open in a new tab Furthermore, gene ontology analyses suggested that the top pathways affected by RP11‐424G14.1 knockdown included the NF‐κB pathway, consistent with known sex differences in inflammation. Intriguingly, both RP11‐424G14.1 and XIST knockdown significantly affected senescence‐related pathways, in line with sexual dimorphism in aging (Figure [81]1E). With available antibodies, we were able to confirm the regulation of IGFBP3 (Insulin‐like growth factor binding protein 3), among other potential leads, by RP11‐424G14.1 (Figure [82]2A,B, Figure [83]S2). Because IGFBP3 was the top candidate from the microarray experiment, we focused subsequent study on IGFBP3 (insulin‐like growth factor binding protein) on its potential role in cellular senescence, the co‐regulated biological pathway. FIGURE 2. FIGURE 2 [84]Open in a new tab Regulation of senescence by male‐biased molecules RP11‐424G14.1 and IGFBP3. (A) Expression level of IGFBP3 in female or male keratinocytes by qPCR, with RNAi‐mediated knockdown (Ri) of indicated genes. Scr, scrambled control. (B, C) Immunofluorescence staining (B) and quantification (C) of IGFBP3 with DAPI counterstain in male (M) or female (F) keratinocytes, with indicated knockdown or scrambled control. (D, E) Immunofluorescence staining (D) and quantification (E) of p21 with DAPI counterstain in male (M) or female (F) keratinocytes, with indicated knockdown or scrambled control. (F) Quantification of BrdU assay in male or female keratinocytes, with indicated knockdown or scrambled control. Average fluorescence intensity per cell is used in quantification. Scale bar, 10 μm. Mean ± stdev, *p < 0.05, Student's t‐test. 3.3. IGFBP3 as a lncRNA‐regulated male‐biased gene as well as their modulation of senescence As an RP11‐424G14.1‐supported and XIST‐repressed gene from microarray results, we predicted that IGFBP3 was a male‐biased gene. Indeed, in independent experiments we confirmed the male‐biased expression of IGFBP3 in human keratinocytes on the mRNA level and in subcellular compartments (Figure [85]2A,B). Additionally, we confirmed that in male keratinocytes, RP11‐424G14.1 knockdown reduced IGFBP3 observed within the cell (Figure [86]2A–C). On the contrary, in female keratinocytes, XIST knockdown increased IGFBP 3 observed within the cell (Figure [87]2A–C). Using p21 as a marker for cell senescence,[88] ^24 we showed that age‐ and race‐matched male keratinocytes expressed increased levels of p21 compared to female (Figure [89]2D,E), consistent with previous reports that females exhibit lower biological ages as assessed by molecular markers.[90] ^4 , [91]^5 Knockdown of RP11‐424G14.1 or IGFBP3 reduced p21 levels in male keratinocytes, while knockdown of XIST increased p21 levels in female keratinocytes (Figure [92]2D,E). Similar results were obtained using the bromodeoxyuridine (BrdU) assay,[93] ^25 which showed limited proliferative capability of male keratinocytes compared to females (Figure [94]2F, Figure [95]S3). The knockdown of RP11‐424G14.1 or IGFBP3 restored proliferation of male keratinocytes (Figure [96]2F). Our results suggest that the RP11‐424G14.1 – IGFBP3 axis promotes senescence in male keratinocytes. 3.4. Regulation of IGFBP3 expression by insulin and nutrient levels IGFBP3 has been historically used as a marker for growth hormone levels and is known to associate with high body mass index (BMI).[97] ^26 , [98]^27 In addition to its role in binding IGF (insulin‐like growth factor), IGFBP3 can signal through the IGF receptor, which can also relay signal from insulin, to regulate cell growth and survival.[99] ^27 Based on the known link of IGFBP3 to insulin signaling, we tested the hypothesis that IGFBP3 itself is regulated by insulin, a component of the keratinocyte growth media. Indeed, removal of insulin from the keratinocyte growth media reduced IGFBP3 levels, and similar effects were achieved by removing all growth supplements from the media (Figure [100]S4). Removal of insulin or nutrients and the reduction in IGFBP3 were concurrent with the decrease in p21 (Figure [101]S4). The decrease in p21 and cell number with insulin treatment is consistent with previous reports that increased insulin levels promote senescence of various human cell types and that nutrient restriction reduces senescence.[102] ^28 , [103]^29 Insulin is known to signal through AKT signaling,[104] ^30 and in male keratinocytes we have found that AKT phosphorylation was dependent on RP11‐424G14.1 and IGFBP3, which suggests that IGFBP3 can both respond to and regulate insulin signaling (Figure [105]S4). Our ancestors have been dubbed “fat hunters”, and to accommodate the large brains, the need for fat is thought to propel the evolution of H. erectus.[106] ^31 Given the upregulation of IGFBP3 by insulin and nutrient levels, we were curious to examine its expression regulation in other organisms. Intriguingly, we found that RP11‐424G14.1, the promoter of IGFBP3 expression in human keratinocytes, was human‐specific and not found in the rhesus monkey, mouse, or other lower organisms (Figure [107]S5A). In the mouse skin, without RP11‐424G14.1, there was no longer a significant male bias in Igfbp3 expression (Figure [108]S5B). Therefore, the male‐biased RP11‐424G14.1 – IGFBP3 axis is human‐specific. 3.5. Metabolism regulation by RP11‐424G14.1 and IGFBP3 Given the observed link between IGFBP3 and nutrition, we directly tested whether RP11‐424G14.1 and IGFBP3 regulated cell metabolism by metabolomic studies. Over 600 metabolites from 26 biochemical classes were profiled and quantified in keratinocytes with control or knockdown of RP11‐424G14.1 or IGFBP3. Consistent with a putative role of RP11‐424G14.1 – IGFBP3 in lipid metabolism, the most significant effect upon knockdown of RP11‐424G14.1 or IGFBP3 was observed in the acylcarnitine group. RP11‐424G14.1 or IGFBP3 knockdown resulted in increases in various long‐chain acylcarnitines such as octadecenoylcarnitine, hydroxyhexadecenoylcarnitine, hydroxyhexadecanoylcarnitine, hydroxytetradecadienoylcarnitine, and tetradecanoylcarnitine, suggesting impairment of beta‐oxidation of fatty acids (Figure [109]3A). In addition, RP11‐424G14.1 or IGFBP3 knockdown led to decreases in medium‐chain acycarnitines including dodecenoylcarnitine, dodecanedioylcarnitine, decenoylcarnitine, and nonaylcarnitine (Figure [110]3A). Meanwhile, limited changes were detected for other lipid groups including diglycerides, triglycerides, phosphatidylcholines, lysophosphatidylcholines, or free or non‐covalently bound fatty acids (Figures [111]4 and [112]5). Small molecules including nucleobases, amino acid, vitamins, and other metabolites remain unchanged upon the knockdowns (Figure [113]3C, Figure [114]5). The pattern of metabolite changes with knockdown in RP11‐424G14.1 and IGFBP3 is consistent with them working in the same pathway. Therefore, the male‐biased factors RP11‐424G14.1 and IGFBP3 regulate lipid metabolism in keratinocytes. FIGURE 3. FIGURE 3 [115]Open in a new tab Regulation of metabolism by male‐biased molecules RP11‐424G14.1 and IGFBP3. Metabolic profiling and quantification of acylcarnitine (A), amino acid (B) and nucleobases and vitamin and cofactors (C) in male keratinocytes with scrambled RNAi (scr Ri), RP11‐424G14.1‐specific RNAi (RP11‐424G14.1 Ri), and IGFBP3‐specific RNAi (IGFBP3 Ri). *, FDR <0.05. FIGURE 4. FIGURE 4 [116]Open in a new tab Regulation of diglycerides and triglycerides by male‐biased molecules RP11‐424G14.1 and IGFBP3. Metabolic profiling and quantification of diglycerides (A) and triglycerides (B) in male keratinocytes with scrambled RNAi (scr Ri), RP11‐424G14.1‐specific RNAi (RP11‐424G14.1 Ri), and IGFBP3‐specific RNAi (IGFBP3 Ri). *, FDR <0.05. FIGURE 5. FIGURE 5 [117]Open in a new tab Regulation of phosphoglycerolipids and additional metabolites by male‐biased molecules RP11‐424G14.1 and IGFBP3. Metabolic profiling and quantification of phosphoglycerolipids (A) and free fatty acids (B) in male keratinocytes with scrambled RNAi (scr Ri), RP11‐424G14.1‐specific RNAi (RP11‐424G14.1 Ri), and IGFBP3‐specific RNAi (IGFBP3 Ri). *, FDR <0.05. 3.6. Regulation of NF‐κB pathway by RP11‐424G14.1 Enrichment of the NFκB pathway upon RP11‐424G14.1 knockdown from the microarray study prompted us to investigate the role of RP11‐424G14.1 in regulating inflammation, a female‐biased response. During inflammation, activated NFκB translocates from cytosol to nucleus to coordinate with cofactors and regulate expression of inflammatory genes.[118] ^32 We simulated inflammation using TNFα, LPS, and IL‐1β and found that the combination of RP11‐424G14.1 knockdown and TNFα stimulation promoted NFκB nuclear translocation and activation (Figure [119]6, Figure [120]S6). Mechanistically, RP11‐424G14.1 inhibition of NFκB activation does not appear to function through canonical means, as TNFα induced IκBα degradation regardless of RP11 knockdown (Figure [121]6, Figures [122]S6 and [123]S7). Therefore, the lack of RP11‐424G14.1 in female keratinocytes potentiates TNFα‐induced NFκB activation. FIGURE 6. FIGURE 6 [124]Open in a new tab Regulation of NFκB nuclear translocation by RP11‐424G14.1. (A) Immunofluorescence staining of NFκB and pIκBα in male keratinocytes with RP11‐424G14.1 knockdown (RP11‐424G14.1 Ri) or scrambled knockdown (Scr Ri), with or without stimulation with TNFa, LPS, or IL1B. Scale bar, 10 μm. (B) Keratinocytes stained with DAPI to delineate their nuclei were analyzed for nuclear NFkB localization. Regions of interest (ROI) were drawn around nuclei using DAPI counterstain, and then anti‐NFkB‐GFP probe mean fluorescence intensity (MFI) within the ROI was measured with standardized laser intensity and exposure settings between conditions. Whisker‐plot of nuclear NFκB signal calculated with mean fluorescence intensity (MFI) of n ≥ 199. Kruskal–Wallis test was performed due to half‐normal distribution and differences in group size (α = 0.05). **, P < 0.01. ****, P < 0.0001. 4. DISCUSSION With protein‐coding genes making up only approximately 1 percent of the human genome, it is important to understand how the non‐coding genetic elements may contribute to sexual dimorphism. Our study uncovers the multifaceted roles of a Y‐linked noncoding RNA, RP11‐424G14.1, in mediating sex differences in inflammation, senescence, and metabolism. We have found that in male keratinocytes, RP11‐424G14.1 signals through IGFBP3, which is regulated by XIST in an opposing manner in female cells. Through IGFBP3, we show that RP11‐424G14.1 and XIST modulate p21‐mediated senescence in a manner consistent with the increased longevity of females. The roles of the Y‐linked RP11‐424G14.1 and X‐linked XIST in regulation of inflammation, senescence, and metabolism suggest the contribution of sex chromosomes to these processes. While we have so far obtained no direct evidence for the regulation of RP11‐424G14.1 and XIST by sex hormones in cultured keratinocytes, future studies are warranted to test for a potential genetic and hormonal interaction. Indeed, studies show that XIST promotes insulin resistance in gestational diabetes,[125] ^33 suggesting a regulation of noncoding RNAs by dynamic changes in sex hormone levels. RP11‐424G14.1 is a human‐specific lncRNA. Consistently, we have found that RP11‐424G14.1‐supported male bias of IGFBP3 is not conserved in the mouse. Intriguingly, the evolution of humans is tightly linked to the need to feed on and metabolize fat.[126] ^31 Our metabolomic data suggests that RP11‐424G14.1 and IGFBP3 work in a coordinated manner to regulate fatty acid metabolism, with a decrease in mobilized medium‐chain acylcarnitines and increase in mobilized long‐chain acylcarnitines with their knockdown. The lack of change in diglycerides or triglycerides implies that the cells are not simply meeting the demands of increased proliferation on their membranes. Furthermore, RP11‐424G14.1 and IGFBP3 may regulate lipid metabolism through mitochondria homeostasis, as has been reported for IGFBP3.[127] ^34 Further study on the mechanism by which RP11‐424G14.1 and IGFBP3 regulate various classes of lipids is necessary to address these possibilities. RP11‐424G14.1 also has a role in protecting male keratinocytes from TNFα‐induced inflammation. Knockdown of RP11‐424G14.1, mimicking the female scenario, promotes the nuclear translocation of NFκB. Given the degradation of IκBα, we speculate that RP11‐424G14.1 functions on other regulatory components for NFκB subunits such as HDAC3 and IRFD themselves or the importins/exportins and modulates the nuclear translocation process.[128] ^35 , [129]^36 , [130]^37 lncRNAs are capable themselves of modifying proteins through ubiquitination or adenylation.[131] ^19 Otherwise, RP11‐424G14.1 is located on the Y chromosome upstream of KDM5D, a histone demethylase known for sexually dimorphic epigenetic regulation and interaction with HDACs.[132] ^38 In conclusion, XIST and RP11‐424G14.1 are sex‐chromosome‐encoded lncRNAs that modulate sexual dimorphic pathways in females and males, respectively. While XIST has previously been described as an important factor in female bias towards autoimmunity, we have found that RP11‐424G14.1 plays an opposing role in males. In male keratinocytes, RP11‐424G14.1 promotes senescence, modulate fatty acid metabolism, and prevent TNFα‐induced NFκB activation. Our data establishes RP11‐424G14.1 as a Y‐linked, human‐specific lncRNA that plays a multifaceted role in regulating sexual dimorphism. AUTHOR CONTRIBUTIONS Conceptualization, K.K. and Y.L.; Methodology, K.K. and Y.L.; Data collection and analysis, K.K. and Y.L.; Writing, reviewing and editing, K.K. and Y.L. All authors have read and agreed to the published version of the manuscript. FUNDING INFORMATION Y.L. has received research support from the NIH National Institute of Arthritis and Musculoskeletal and Skin Diseases grant K01 AR073340, R01 AR078781 and the National Institute of Dental and Craniofacial Research grant R21 DE031765. CONFLICT OF INTEREST STATEMENT The authors have stated explicitly that there are no conflicts of interest in connection with this article. Supporting information Figure S1. [133]FBA2-7-e1479-s001.zip^ (22.6MB, zip) ACKNOWLEDGMENTS