Abstract
Previous studies showed that noncalcemic 20(OH)D3, a product of CYP11A1 action on vitamin D3, has antifibrotic activity in human dermal fibroblasts and in a bleomycin mouse model of scleroderma. In this study, we tested the role of retinoic acid-related orphan receptor γ (RORγ), which is expressed in skin, in the action of CYP11A1-derived secosteroids using murine fibroblasts isolated from the skin of wild-type (RORγ +/+), knockout (RORγ -/-), and heterozygote (RORγ +/-) mice. CYP11A1-derived 20(OH)D3, 20,23(OH)2D3, 1,20(OH)2D3, and 1,20,23(OH)3D3 inhibited proliferation of RORγ +/+ fibroblasts in a dose-dependent manner with a similar potency to 1,25(OH)2D3. Surprisingly, this effect was reversed in RORγ +/- and RORγ -/- fibroblasts, with the most pronounced stimulatory effect seen in RORγ -/- fibroblasts. All analogs tested inhibited TGF-β1-induced collagen synthesis in RORγ +/+ fibroblasts and the expression of other fibrosis-related genes. This effect was curtailed or reversed in RORγ -/- fibroblasts. These results show that the antiproliferative and antifibrotic activities of the vitamin D hydroxy derivatives are dependent on a functional RORγ. The dramatic changes in the transcriptomes of fibroblasts of RORγ -/- versus wild-type mice following treatment with 20(OH)D3 or 1,20(OH)2D3 provide a molecular basis to explain, at least in part, the observed phenotypic differences.
Keywords: fibroblasts, vitamin D3, hydroxyderivatives, RORγ
The retinoic acid-related orphan receptors (RORs), RORα, RORβ, and RORγ, constitute a subfamily of nuclear receptors (1). They regulate the transcription of target genes by binding to ROR-response elements (2). ROR receptors play a critical role in the control of various physiological and developmental processes and are implicated in the development of many diseases, including several malignancies, inflammatory and metabolic diseases, and psoriasis (2, 3). The RORγ gene (RORC) gives rise to 2 different receptor isoforms: RORγ1 and RORγt (RORγ2). RORγ1 is expressed in several metabolic tissues such as liver, kidney, skeletal muscles, small intestine, pancreas, and brown fat cells (4), and plays a role in insulin sensitivity and obesity (5, 6). RORγt expression is restricted to several immune cells (1). Mice lacking the RORγ gene lack lymph node organogenesis as well as an impaired allergic response (1) and increased susceptibility to thymic lymphoma (7). Treatment with RORγ inverse agonists diminishes the severity of autoimmune diseases such as diabetes and multiple sclerosis, and inflammatory lung and skin disease, in experimental mouse models (8-13). Thus, RORγ inverse agonists might potentially be useful in the treatment of collagen-induced arthritis, rheumatoid arthritis, asthma, and other autoimmune diseases, as well as obesity (14, 15) and psoriasis (11). Novel vitamin D3 derivatives used in this study are among the many endogenous ligands for RORs (16, 17).
New pathways of vitamin D activation through the hydroxylation of its side chain by CYP11A1 have recently been established (18-29). The products of these pathways, including 20(OH)D3, 20,23(OH)2D3, 1,20(OH)2D3, and 1,20,23(OH)3D3, exhibit potent biological activity in different types of cells, including skin-derived cells (18-30). In addition, 20(OH)D3 and 20,23(OH)2D3 are noncalcemic, whereas 1,20(OH)2D3 shows low calcemic activity, in contrast to calcitriol (1,25(OH)2D3) (24-26, 31). As with 1,25(OH)2D3, which is the major hormonally active form of vitamin D3, the CYP11A1-derived hydroxymetabolites exhibit anticancer activities, antiproliferative, and pro-differentiation effects, and have immunomodulatory functions (32-34).
Vitamin D derivatives act mainly through the vitamin D receptor (VDR) (35). Our previous studies pertaining to CYP11A1-derived vitamin D3 hydroxy-analogs show that in addition to acting on the VDR (17, 25, 33, 36-38), they also act as inverse agonists of RORα and RORγ receptors (16, 17), and as agonists on the aryl hydrocarbon receptor (AhR) (39). Both RORα and RORγ receptors are expressed in murine and human skin (16, 40-45).
As outlined previously, vitamin D and its derivatives exhibit antifibrotic properties (46, 47) including antifibrogenic activities by 20(OH)D3 and 20,23(OH)2D3 in human dermal fibroblasts, and in mice when delivered IP (25). In this study, we explored the role of RORγ in response to CYP11A1-derived vitamin D derivatives in dermal fibroblasts isolated from RORγ -/- (knockout [KO]), RORγ -/+ (heterozygotes), and RORγ +/+ (wild-type) mice. Our aim was to identify a link between fibrosis, CYP11A1-derived vitamin D3 analogs and the RORγ receptor in relation to their therapeutic potential, and to expand our understanding of the actions of novel vitamin D3 derivatives. We also tested whether orally delivered 20(OH)D3 exhibits antifibrogenic effects in mice.
Material and Methods
Antifibrogenic effects of 20(OH)D3 in a mouse model
Six-week-old female C57/BL6 mice, purchased from Jackson Laboratory, were treated as previously described (24) with minor differences as detailed in the legend to Table 1. Briefly, mice were injected subcutaneously, 6 days per week, with 50 µL sterile saline or an equal amount of saline containing 50 µg bleomycin (Fresenius Kabi USA, Lake Zurich, IL). Mice were further gavaged with 100 µL propylene glycol (1:5 dilution in saline) or with propylene glycol containing 20S-hydroxyvitamin D3 (20(OH)D3) at 3 different doses: 5, 15, or 30 µg/kg. Mice were sacrificed after 22 days. Skin was used for histological analyses. Briefly, skin samples were formalin-fixed paraffin embedded, sectioned, and stained with hematoxylin and eosin (24, 25). The Institutional Animal Care and Use Committee at the University of Tennessee Health Science Center approved experiments in mice under protocol number 15-111 (Dr. A. Postlethwaite, principal investigator).
Table 1.
20S(OH)D3 Given via Oral Gavage Suppresses Bleomycin-induced Dermal Fibrosis
| Condition | Dermal Thickness | P Value | |
|---|---|---|---|
| Subcutaneous | Gavage | µM mean ± SEM | |
| Saline | Vehicle | 238 ± 9 | |
| Bleomycin | Vehicle | 476 ± 33 | <0.001 |
| Bleomycin | 20S(OH)D3 (5 µg/kg) |
278 ± 15 | 0.004 |
| Bleomycin | 20S(OH)D3 (15 µg/kg) |
314 ± 18 | 0.005 |
| Bleomycin | 20S(OH)D3 (30 µg/kg) |
234 ± 31 | 0.004 |
Six-week-old C57BL/6 female mice were injected SC in a 1.5 cm × 1.5 cm area in the back in the lower thoracic region 6 days/wk with 50 µL sterile intravenous grade normal saline (“saline”) or 50 µL saline containing 50 µg bleomycin (Fresenius Kabi USA). Mice were gavaged 6 days/wk with either 100 µL propylene glycol diluted 1:5 with saline or propylene glycol diluted 1:5 with saline containing 20(OH)D3 at a dose of 5 µg/kg, 15 µg/kg, or 30 µg/kg. On day 22, mice were euthanized and skin from the injection site was processed for histology and stained with Masson Trichrome. The degree of dermal thickening was measured by light microscopy by 2 examiners.
Experiments with fibroblasts from newborn mice
Skin tissue from newborn mice, RORγ wild-type (RORγ +/+), heterozygotes (RORγ -/+), or knock out (RORγ -/-) was collected was collected at the National Institute of Environmental Health Sciences. The protocols for the breeding of mice, genotyping, and organ collections are described elsewhere (48, 49). All animal studies followed guidelines outlined by the National Institutes of Health Guide for the Care and Use of Laboratory Animals and protocols were approved by the Institutional Animal Care and Use Committee at the National Institute of Environmental Health Sciences (approval date: April 26, 2018; animal welfare assurance number is A4149-1, Dr. A. Jetten, principal investigator). Fibroblasts were isolated from the skin of 1- to 2-day-old mice. Briefly, skin was rinsed in PBS (Gibco, Life Technologies, Carlsbad, CA), cut into smaller pieces and incubated in 2.4 U/mL of dispase II (Roche, Indianapolis, IN) overnight at 4°C. The dermis was first separated from the epidermis using a razor blade and further incubated in 0.1% collagenase (Roche) overnight at 4°C. The following day, the dermis was cut into smaller pieces and incubated in 0.25% Trypsin (CellGrow, Corning, NY) for 1 hour at 37°C. A suspension of single cells was prepared by pipetting and draining through a cell strainer. After centrifuging to remove trypsin, the cell pellet was resuspended in complete DMEM media (CellGrow) containing 10% serum (Sigma-Aldrich, St. Louis, MO), mouse fibroblast growth factor (Sigma-Aldrich), and 1% antibiotic (Penicillin-Streptomycin, CellGrow), and plated onto tissue culture-treated dishes (TPP, Midwest Scientific, Valley Park, MO) for expansion. Cells in their early passage were used for experiments and were treated with compounds diluted in DMEM and 10% charcoal-stripped fetal bovine serum (FBS; Sigma-Aldrich). In addition, cells isolated from different mice were pooled to provide more representative samples.
Cell proliferation assay (MTS assay)
Murine fibroblasts were plated onto 96-well plates at a confluence of about 70%. Cells were treated with the following vitamin D3 derivatives: 1,25(OH)2D3 (from Sigma) or 20(OH)D3 synthesized chemically (50), or 20,23(OH)2D3, 1,20(OH)2D3, or 1,20,23(OH)3D3 synthesized enzymatically (22, 51, 52), diluted in ethanol in DMEM containing charcoal-treated serum. Final concentrations of the vitamin D hydroxyderivatives were 0.1, 10, or 100 nM, with the final ethanol concentration being 0.1%, which was also used as a vehicle control. After 44 hours of incubation, MTS (Promega, Madison, WI) solution (20 μL) was added to the media, which was further incubated for 3 hours. The absorbance (490 nm) was recorded using an ELISA plate reader. The number of viable cells was measured in 6 replicates.
Collagen assay
Murine fibroblasts were grown to 90% confluence. Cells were treated with 1,25(OH)2D3, 20(OH)D3, 20,23(OH)2D3, 1,20(OH)2D3, or 1,20,23(OH)3D3 in 10% charcoal-treated FBS plus supplements, for 6 or 24 hours, in 60-mm Petri dishes in a total volume of 10 mL. The hydroxyvitamin D3 derivatives were used at a concentration of 100 nM. Supernatant (1 mL) was collected and used for collagen quantification by the Sircol collagen assay protocol (Biocolor, S1000). Total collagen concentration was calculated based on the absorbance of the samples in comparison to the standard curve absorbance.
RNA sequencing preparation and analyses
Murine fibroblasts were grown as described previously. After supernatant was collected for the assay of collagen, cells were harvested for RNA isolation (RNAeasy Micro kit, Qiagen) and cDNA synthesis (High Capacity cDNA Reverse Transcription Kit with RNase Inhibitor, Applied Biosystems) following the manufacturers’ protocols. At least 200 ng of RNA for each sample (24-hour treatment) was used to create libraries for RNA sequencing by BGI Americas Corporation Service (Cambridge, MA). Sample quality and concentration were determined using Agilent 2100.
The raw sequence FASTQ files were aligned to Gencode’s GRCm38 p4 Release M11 genome using STAR, version 2.5.4b (parameters used --outReadsUnmapped Fastx; --outSAMtype BAM SortedByCoordinate; --outSAMattributes All; --outFilterIntronMotifs RemoveNoncanonicalUnannotated). Transcript abundances were then calculated from the alignments using Cufflinks, version 2.2.1 (parameters used -G; -L; --library-type fr-firststrand). Samples were then merged using default parameters with Cuffmerge. Finally, differential expression was calculated using Cuffdiff’s default parameters. The raw data are deposited at the NCBI GEO (GSE145818).
Pathway analysis using Ingenuity
Data were analyzed with Ingenuity Pathway Analysis (Ingenuity Systems, www.ingenuity.com). For generating networks, a data set containing gene identifiers and corresponding expression values was uploaded into the application. Each identifier was mapped to its corresponding object in Ingenuity’s Knowledge Base. A fold change cutoff of ±2 was set to identify molecules whose expression was significantly differentially regulated. These molecules, called Network Eligible molecules, were overlaid onto a global molecular network developed from information contained in Ingenuity’s Knowledge Base. Networks of Network Eligible Molecules were then algorithmically generated based on their connectivity. The Functional Analysis identified the biological functions and/or diseases that were most significant to the entire data set. Molecules from the dataset that met the fold change cutoff of ±2 and were associated with biological functions and/or diseases in Ingenuity’s Knowledge Base were considered for the analysis. Right-tailed Fisher’s exact test was used to calculate the P value determining the probability that each biological function and/or disease assigned to that data set is due to chance alone. The RNA sequencing (RNA-seq) raw data are deposited at the NCBI GEO (GSE145818).
Gene set enrichment analysis (GSEA (53)) was performed as follows. The fragments per kilobase of exon model per million reads mapped values were log2 transformed and the differentially expressing genes with an additional criterion of fold-change ±1.5 were selected for the enrichment analysis. GSEA was performed through METASCAPE (http://metascape.org) (54).
Quantitative RT-PCR
RT-PCR was carried out using Sybr green Master Mix (Thermo Scientific, Waltham, MA), in triplicates; as previously described (55). Approximately 750 ng of cDNA was used for PCR reactions. GAPDH and β-actin were used as internal controls. Primer sequences are listed in Table 2. The source of primers is as follows: Bmp7 (56), Mmp1b, Tgfb1, Col1A2, Col3A1 (57), Hgf (58), Cyp1b1 (59).
Table 2.
Primers and Sequences Used for RT-PCR
| Gene name | Sequences |
|---|---|
| Bmp7 | GATT- TCAGCCTGGACAACGAG GGGCAAC- CCTAAGATGGACAG |
| Mmp1b | AAGGCGATATTGTGCTCTCC CCTCATTGTTGTCGGTCCAC |
| Tgfb1 | GCGGACTACTATGCTAAAGAGG TCAAAAGACAGCCACTCAGG |
| Col1A2 | CCGTTGGCAAAGATGGTAG GTCAGCCCTGTAGAAGTATCC |
| Col3A1 | AAAAACCCTGCTCGGAACT CAGCTGCACATCAACGACA |
| Hgf |
CATTGGTAAAGGAGGCAGCTATAAA GGATTTCGACAGTAGTTTTCCTGTAGG |
| Cyp1b1 | AATGAGGAGTTCGGGCGCACA GGCGTGTGGAATGGTGACAGG |
| Gapdh | CCATTTTGTCTACGGGACGA AGTATGACTCCACTCACGGCAAAT |
| B-actin | CTAAGGCCAACCGTGAAAAG ACCAGAGGCATACAGGGACA |
Data were analyzed using GraphPad Prism 8 (San Diego, CA) statistical software 1-way ANOVA or t-test, where appropriate; *P < 0.05; **P < 0.01; ***P < 0.001.
Results with Discussion
We previously reported that 20(OH)D3 suppressed fibrogenesis in mice after IP treatment and that 20(OH)D3, 20,23(OH)2D3, and 17,20,23(OH)3D3 inhibited TGFβ-1-induced fibrinogenic effects in cultured dermal fibroblasts (24). In the current study, we analyzed the effect of 20(OH)D3 on bleomycin-induced dermal fibrosis in mice after oral delivery by gavage to examine its usefulness for (pre)clinical studies on scleroderma. We focused on 20(OH)D3, the first and major product of CYP11A1-mediated vitamin D3 metabolism (20-22). Oral administration of 20(OH)D3 resulted in significantly less bleomycin-induced dermal fibrosis and its thickening than in control mice, as shown by the representative sections in Fig. 1 and the quantification in Table 1. The effect was seen at doses of 5 µg/kg, 15 µg/kg, and 30 µg/kg with the maximum effect seen at 30 µg/kg (Table 1). These data demonstrated that oral treatment with 20(OH)D3 attenuated the bleomycin-induced dermal fibrosis in mice. Hence, this route of administration can be considered for future clinical trials with noncalcemic vitamin D3 derivatives for the treatment of scleroderma, morphea, and other diseases involving fibrosis.
Figure 1.
H&E stained sections of murine skin. Bleomycin (BM) was administered, 50 µg/mouse SC, for 22 days. 20(OH)D3 or its solvent, propylene glycol (PG) was given as an oral gavage as described in the legend to Table 1. (A) Vehicle control (saline SC and propylene glycol gavage); (B) bleomycin SC and propylene glycol gavage; (C) bleomycin SC and 5 µg/kg 20(OH)D3 gavage; (D) bleomycin SC and 15 µg/kg 20(OH)D3 gavage; and (E) bleomycin SC and 30 µg/kg 20(OH)D3 gavage.
Previous studies have shown that 1,25(OH)2D3 inhibits fibroblast proliferation and decreases the synthesis of collagen (60); effects also seen with CYP11A1-derived vitamin D3 hydroxyderivatives (24, 61, 62). To ascertain the role of RORγ in these responses, we examined the effect of these derivatives on cells isolated from the skin of wild type (RORγ +/+), RORγ -/-, and RORγ -/+ mice. The proliferation of fibroblasts treated for 48 hours with 1,25(OH)2D3 or with the novel vitamin D3 derivatives 20(OH)D3, 20,23(OH)2D3, 1,20(OH)2D3, and 1,20,23(OH)3D3, was significantly reduced in wild-type fibroblasts (P < 0.01 or P < 0.001) compared with ethanol-treated control cells. This inhibition was observed with both 0.1 and 100 nM secosteroid, concentrations selected based on a previous study (33). In contrast, this inhibitory effect on proliferation was reversed in both homozygous KO (RORγ -/-) and heterozygous (RORγ -/+) fibroblasts (Fig. 2) and instead a significant increase in proliferation was observed in response to every secosteroid tested. The effects were generally more pronounced in homozygous fibroblasts than heterozygous fibroblasts indicating a dosing effect (Fig. 2) and were observed at 24 and 72 hours posttreatment (data not shown).
Figure 2.
The effect of secosteroids on the proliferation of fibroblast cells from RORγ +/+, RORγ -/+, and RORγ -/- mice. Fibroblasts were isolated from murine skin and grown until the third passage. After they reached 70% confluence, cells were treated with secosteroids at the indicated concentrations for 48 hours. MTS was added and absorbance read after 3 h of incubation. Data were analyzed using the t test and represent means ± SD (n ≥ 6), **P < 0.01, ***P < 0.001, ****P < 0.0001 by t test. ROR, retinoic acid-related orphan receptor.
It is known that VDR is required for proliferation and differentiation of epidermal cells and the action of 1,25(OH)2D3 (63), as well as epidermal stem cells (64) and plays a role in inflammation and injury. Dermal fibroblast function was altered in VDR KO mice (65). The effect of 1,25(OH)2D3 has been tested in mouse models of tissue fibrosis, showing that this vitamin D3 derivative can prevent fibrosis independently of VDR-mediated transcriptional activity (66). Because vitamin D3 derivatives can act on fibrosis independently of VDR, we proposed that that action might be RORγ receptor dependent. There is a shortage of information in the literature to support the role of RORγ receptor in the action of vitamin D in murine skin. The presented results support an important role of RORγ in the inhibition of dermal fibroblast proliferation by secosteroids.
Next, we examined the role of RORγ in the inhibition of collagen synthesis by vitamin D3 derivatives in dermal fibroblasts. Fibroblasts, isolated from the skin of RORγ wild type or KO mice, were treated with 100 nM 1,25(OH)2D3, 20(OH)D3, 20,23(OH)2D3, 1,20(OH)2D3, or 1,20,23(OH)3D3 and the amount of collagen in the supernatant measured after 6 hours (data not shown) or 24 hours of treatment (Fig. 3). There was a significant decrease in the collagen concentration in the supernatants from wild-type fibroblasts after treatment with all of the secosteroids compared with the ethanol-treated control cells. As observed for proliferation, the effect was reversed for the RORγ -/- fibroblasts, which showed a significant increase in the collagen concentration in response to all the secosteroids tested (Fig. 3).
Figure 3.
The effect of secosteroids on collagen production by fibroblast cells from RORγ +/+ and RORγ -/- mice. Fibroblasts were isolated from RORγ +/+ (wild-type [WT]) and RORγ -/- (knockout [KO]) mouse skin and were grown in DMEM supplemented with 10% FBS, testosterone, and mouse fibroblast growth factor to 90% confluence. Cells were then treated with secosteroids (100 nM) in 10% charcoal-treated FBS plus supplements for 24 hours in 60-mm Petri dishes in total volume of 10 mL. The supernatant was collected and used for collagen quantification using the Sircol collagen assay Biocolor, S1000. Data were analyzed using the t test and represent means ± SD, *P < 0.05, **P < 0.01. FBS, fetal bovine serum; ROR, retinoic acid-related orphan receptor.
To relate these effects to global transcriptomic activity, we compared the gene expression profiles between RORγ +/+ and RORγ -/- fibroblasts by RNA-seq analysis. Pathway analysis showed that several proliferation-related functions, DNA damage responses, and hepatic fibrosis were among the top control pathways (Table 3, Supplemental Table 1, located in a digital research materials repository (67)). In addition, the main regulators for which expression was inhibited in RORγ -/- fibroblasts compared with the RORγ +/+ included Tgfb1, Vegf, TNnfa, dextran sulfate, and Erbb2 (Table 3, Supplemental Table 1) (67). A dramatic change in the expression of upstream genes, with predominant inhibition in RORγ -/- fibroblasts compared to RORγ +/+ fibroblasts, was observed. This included genes for many cytokines, chemokines, growth factors, enzymes, and transcriptional regulators with only a few being upregulated (Table 3, Supplemental Table 2, located in a digital research materials repository (67)).
Table 3.
Gene Set Enrichment Analysis for Signaling, Proliferation, Differentiation, or Healing Processes Based on RNA-Seq Data Obtained After 24 Hours of in vitro Incubation of Murine Fibroblasts, RORγ+/+ (WT), or RORγ-/- (KO) with Ethanol (vehicle), 1,20(OH)2D3, or 20(OH)D3
| Pathway Name | Sample Sets | |||||||||
|---|---|---|---|---|---|---|---|---|---|---|
| KO_EtOH vs WT-EtOH | KO_1,20(OH)2D3 vs KO_EtOH | KO_20(OH)D3 vs KO_EtOH | WT_1,202D3 vs WT_EtOH | WT_20(OH)D3 vs WT_EtOH | ||||||
| logp | Expression | |||||||||
| Positive regulation of collagen biosynthetic process | -3.4 | UP | ||||||||
| Vascular endothelial growth factor receptor signaling pathway | -1.8 | UP | ||||||||
| Wound healing | -1.8 | UP | ||||||||
| Epithelial cell development | -1.7 | UP | ||||||||
| Regulation of epidermal cell differentiation | -1.6 | UP | ||||||||
| Cellular response to vascular endothelial growth factor stimulus | -1.5 | UP | ||||||||
| Regulation of wound healing | -1.3 | UP | ||||||||
| Epidermal cell differentiation | -1.3 | UP | -1.5 | DOWN | ||||||
| Cell cycle | -31 | DOWN | ||||||||
| Cell division | -30 | DOWN | -1.4 | UP | ||||||
| DNA replication | -27 | DOWN | ||||||||
| Cell cycle, mitotic | -26 | DOWN | ||||||||
| Regulation of cell-cycle process | -22 | DOWN | ||||||||
| Regulation of cell-cycle phase transition | -16 | DOWN | ||||||||
| DNA repair | -15 | DOWN | ||||||||
| Cell cycle | -15 | DOWN | ||||||||
| Epithelial cell proliferation | -14 | DOWN | -2 | UP | ||||||
| Inflammatory response | -12 | DOWN | -2.1 | UP | ||||||
| Positive regulation of cell cycle | -10 | DOWN | ||||||||
| DNA replication | -9.5 | DOWN | ||||||||
| Regulation of reactive oxygen species metabolic process | -6.7 | DOWN | ||||||||
| Regulation of vascular endothelial growth factor production | -6.3 | DOWN | ||||||||
| IL-17 signaling pathway | -5.9 | DOWN | -2 | DOWN | ||||||
| Response to growth factor | -5.8 | DOWN | ||||||||
| Nitric oxide metabolic process | -5.2 | DOWN | ||||||||
| Regulation of DNA repair | -4.6 | DOWN | ||||||||
| TP53 regulates transcription of genes involved in G1 cell-cycle arrest | -4.6 | DOWN | ||||||||
| TNF signaling pathway | -4.4 | DOWN | ||||||||
| Regulation of TNF superfamily cytokine production | -4.4 | DOWN | ||||||||
| Response to oxidative stress | -4.2 | DOWN | ||||||||
| Endothelial cell proliferation | -4.2 | DOWN | ||||||||
| Cellular response to reactive oxygen species | -4.2 | DOWN | -1.6 | UP | ||||||
| Response to UV | -3.6 | DOWN | ||||||||
| Positive regulation of inflammatory response | -3.6 | DOWN | ||||||||
| Response to ionizing radiation | -3.6 | DOWN | ||||||||
| Keratinocyte proliferation | -3.2 | DOWN | ||||||||
| Epithelial cell differentiation | -3 | DOWN | -1.5 | DOWN | -1.5 | DOWN | ||||
| Cellular response to ionizing radiation | -2.9 | DOWN | ||||||||
| NF-kappa B signaling pathway | -2.9 | DOWN | ||||||||
| Collagen degradation | -2.7 | DOWN | ||||||||
| Positive regulation of fibroblast proliferation | -2.6 | DOWN | ||||||||
| Wound healing | -2.6 | DOWN | ||||||||
| Stem cell differentiation | -2.6 | DOWN | -1.5 | DOWN | -2 | DOWN | ||||
| Response to X-ray | -2.5 | DOWN | ||||||||
| Cellular response to hydrogen peroxide | -2.4 | DOWN | ||||||||
| Positive regulation of nitric oxide metabolic process | -2.4 | DOWN | ||||||||
| Collagen formation | -2.3 | DOWN | ||||||||
| Regulation of insulin-like growth factor (IGF) transport and uptake by IGF binding proteins | -2.3 | DOWN | ||||||||
| TGF-beta signaling pathway | -2 | DOWN | ||||||||
| Regulation of response to wounding | -2 | DOWN | ||||||||
| Response to epidermal growth factor | -2 | DOWN | ||||||||
| BMP signaling pathway | -1.9 | DOWN | ||||||||
| Nucleotide excision repair | -1.9 | DOWN | ||||||||
| Base excision repair | -1.8 | DOWN | -2.5 | UP | ||||||
| Response to gamma radiation | -1.8 | DOWN | ||||||||
| Fibroblast proliferation | -1.8 | DOWN | -2 | UP | ||||||
| Negative regulation of NF-kappaB transcription factor activity | -1.8 | DOWN | ||||||||
| Positive regulation of BMP signaling pathway | -1.6 | DOWN | ||||||||
| Vascular wound healing | -1.6 | DOWN | ||||||||
| Response to oxygen radical | -1.6 | DOWN | ||||||||
Abbreviations: BMP, bone morphogenetic protein; KO, knockout; RNA-seq, RNA sequencing; ROR, ROR, retinoic acid-related orphan receptor; WT, wild-type.
Among ingenuity’s predicted associations with biological functions and diseases, cancer, gastrointestinal diseases, organismal injury and abnormalities, reproductive system diseases, and hematological diseases were significantly affected in RORγ -/- fibroblasts compared with the RORγ +/+ (Supplemental Table 1 (67)). Molecular and cellular functions, such as cell death and survival, cellular movement, cell cycle, cellular growth and proliferation, cellular development, as well as organismal survival, development and tissue morphology also appear to be affected in RORγ -/- fibroblasts compared to the RORγ +/+ (Supplemental Tables 1, 2, 3) (67). The main cellular functions that were affected are pertinent to liver, kidney, and heart and included hepatotoxicity, renal toxicity, and cardiotoxicity (Supplemental Tables 1, 2, 3, 4, located in a digital research materials repository (67)). The analysis of the canonical (Supplemental Table 3) (67) and toxicity pathway (Supplemental Table 4, located in a digital research materials repository (67)) also reveals dramatic changes in a number of genes and signaling pathways with predominant inhibition in RORγ -/- fibroblasts as compared with the RORγ +/+. Among the pathways/genes affected are nuclear receptor signaling including VDR, retinoid X receptor (RXR), AhR, peroxisome proliferator-activated receptor, inflammatory signaling including nuclear factor-ƙB and interleukins, growth factors including vascular endothelial growth factor (VEGF), TGF-B1, platelet-derived growth factor, and others such as NRF2 and bone morphogenetic protein (BMP). Thus, deletion of RORγ leads to dramatic changes not only in the gene expression pattern, but also in signaling pathways and their regulators, which would affect the fibroblast phenotype and cell responsiveness to secosteroids (see the following section).
Data obtained from RNA-seq Ingenuity pathway analysis are confirmed with GSEA. Knocking out RORγ receptor affected differentiation and proliferation of a variety of cells, including lymphocytes, T and B cells, mononuclear cells, erythrocytes, stem cells, osteoblasts, mesenchymal, chondrocytes, cardiomyocytes, hepatocytes, muscle, fat cells, epidermal and endodermal cells, etc. (data not shown). Proliferation of fibroblasts, as well as endothelial cell development is impaired in cells lacking RORγ receptor (Table 3). Collagen synthesis and degradation and wound healing processes are downregulated by the lack of RORγ in murine fibroblasts (Table 3). In addition, processes that are known to be affected by vitamin D3 derivatives are altered by the lack of RORγ in murine fibroblasts, including cell cycle and cell division, DNA replication and repair, nucleotide excision repair, reaction to radical oxygen species, response to radiation (X-ray, UV, ionizing radiation, and gamma radiation) (Table 3). Conversely, inflammatory responses, including interleukin-17 pathway, p53, TNFα, and nuclear factor-ƙB, along with the BMPB and VEGF signaling are downregulated in cells lacking RORγ receptor in comparison to wt (Table 3).
In RORγ +/+ fibroblasts, expression of many genes was altered by 20(OH)D3 compared with ethanol-treated control cells, of which 337 were up-regulated and 238 down-regulated (Fig. 4). Biologically relevant target genes were determined based on a fold change (FC) threshold of 1.5. Ingenuity pathway analyses shows that 20(OH)D3 affects many immune, bone/cartilage, atherosclerosis, antioxidative and growth factor related, and other signaling pathways with significant (P < 0.05) VDR/RXR activation (Supplemental Tables 5, 6, located in a digital research materials repository (67)). This is consistent with previously published data in human keratinocytes (28, 30, 34, 39, 68).
Figure 4.
Venn diagrams. Genes with a fold change of +1.5 or -1.5 were compared between fibroblasts treated with 1,20(OH)2D3 and 20(OH)D3 to identify the genes that were common/unique.
In wild-type fibroblasts treated with 1,20(OH)2D3 many target genes were affected by the secosteroid compared with ethanol-treated control cells, with the expression of 376 genes up-regulated (FC ≥1.5) including Fbxw7, Nup210, Trim5, Cr2, Rpl, Rps, Tdrd, and Stub, and 215 and genes down-regulated (FC ≤-1.5) including Pde4dip, Tnrc6b, Ptpn6, Rpl21, Chn2, Ehf, and Erbb4 (Fig. 4). Identified pathways regulated by 1,20(OH)2D3 were similar to those regulated by 20(OH)D3, and included interleukins, VDR/RXR, nuclear factor kB, VEGF, and NRF2 (Supplemental Tables 7, 8, located in a digital research materials repository (67)). Identification of these pathways is consistent with our functional data indicating their immunomodulatory functions, involvement of VDR/RXR, and NRF2 signaling (23, 30, 34, 36, 37, 39, 68).
Further analysis revealed that the expression of 215 genes was downregulated by only 1,20(OH)2D3 and 238 genes were downregulated only by 20(OH)D3, with a further 235 genes downregulated by both vitamin D derivatives for the RORγ +/+ fibroblasts, as presented in the Venn diagram (Fig. 4A). The major functions and the relevance of these genes are described in Table 4. In addition, the expression of 376 genes was up-regulated by only 1,20(OH)2D3 and 337 genes by only 20(OH)D3, with 147 genes up regulated by both vitamin D derivatives (Fig. 4B). For the RORγ -/- fibroblasts, similar pathways were affected by treatment with 20(OH)D3 compared with ethanol treated RORγ -/-cells (Supplemental Tables 9, 10, located in a digital research materials repository (67)) or with 1,20(OH)2D3 (Supplemental Tables 11, 12, located in a digital research materials repository (67)) with some differences. Importantly, VDR/RXR signaling was not detected among pathways activated by 1,20(OH)2D3 in RORγ -/- fibroblasts. In conclusion, knocking out the RORγ significantly affected the responsiveness of cells to 20(OH)D3 and 1,20(OH)2D3.
Table 4.
Gene Expression in Murine Fibroblasts: Comparisons Between RORγ-/- and Wild-Type
| Expression (Fold Change) | Gene Name | |
|---|---|---|
| -1.5 | MMP1b | Interstitial collagenase b |
| -1.23782 | MMP2 | Matrix metallopeptidase 2 or type IV collagenase |
| -3 | MMP8 | Matrix metallopeptidase 8 or neutrophil collagenase |
| -2.86067 | MMP13 | Matrix metallopeptidase 13 or collagenase 3 |
| -2.89474 | BMP6 | Bone morphogenic protein |
| 1.738077 | TGFb1 | Transforming growth factor beta |
| -6.14074 | TGFbr3 | Transforming growth factor beta receptor |
| -1.43419 | Col1A1 | Collagen type I alpha 1 chain or collagen of skin, tendon, and bone, Alpha-1 chain |
| -6.07225 | Col1A2 | Collagen type I alpha 2 chain or collagen of skin, tendon, and bone, alpha-2 chain |
| -4.09706 | Col3A1 | Collagen type III alpha 1 chain or Ehlers-Danlos syndrome type IV |
| 24.464 | Col7A1 | Collagen type VII alpha 1 chain |
| -61.0003 | Col28A1 | Collagen type XXVIII alpha 1 chain |
| -21.1603 | HGF | Hepatocyte growth factor (HGF) or scatter factor (SF) |
| -9.12445 | Cyp1B1 | Cytochrome P450 family 1 subfamily B member 1 |
RNA-sequencing analysis was performed using RNA from murine fibroblasts. Gene expression is shown as fold change.
Comparing the effects of 20(OH)D3 and 1,20(OH)2D3 on RORγ -/- fibroblasts, analysis revealed that the expression of 262 genes was downregulated by only 1,20(OH)2D3 and 379 genes by only 20(OH)D3, with the expression of 273 genes being down regulated by both secosteroids (Fig. 4C). There were 498 genes whose expression was up-regulated by only 1,20(OH)2D3 and 412 genes by only 20(OH)D3, with the expression of 158 genes being up-regulated by both secosteroids (Fig. 4D). The genes whose expression was regulated by both 1,20(OH)2D3 and 20(OH)D3 are presented in Table 3. These genes are primarily involved in regulation of cell proliferation, differentiation, and tumorigenesis, cell signaling, and metal transport. Thus, treatment with 20(OH)D3 and 1,20(OH)2D3 activate distinct and overlapping patterns of gene expression in RORγ -/- fibroblasts.
We performed further RNA-seq analyses to determine whether RORγ is important for transcriptional regulation of key genes involved in fibrosis. We selectively checked the expression of different collagenases and genes from the family of TGF-β-related growth factors. Collagenases/matrix metallopeptidases (MMPs) play roles in degradation of extracellular matrix proteins, as well as differentiation, apoptosis, angiogenesis, and host defense (69). MMPs were expressed at only low levels in cells lacking RORγ (Table 4). MMP8 is involved in skin tumors, arthritis, and lung fibrosis (69), whereas MMP13 (collagenase 3), is the main collagenase in mouse tissue involved in regulation of interstitial collagen and growth plate cartilage defects (70). MMP8 and MMP13 expression changed, respectively 3- and 2.86-fold, in RORγ -/- fibroblasts compared with wild-type, whereas MMP2 and MMP1b expression was not significantly different (-1.5 and -1.2, respectively) (Table 4).
Collagen is the most abundant protein in connective tissue. In skin, the collagen is produced by fibroblasts, which play an important role in wound healing. Collagen type I alpha 1 chain (71) and collagen type I alpha 2 chain (Col1A2) have a ubiquitous distribution, including bones, dermis of the skin, ligaments, and tendons (71). Mutations in these genes are associated with various diseases in humans, including osteogenesis imperfecta, Ehlers-Danlos syndrome, and osteoporosis (71). Collagen type 3 alpha 1 chain (Col3A1) is found in skin, blood vessels, and intestine, and mutations in this type of collagen are involved in aneurysms in arteries, as well as in Ehlers-Danlos syndrome (71). Collagen type XXVIII alpha 1 chain (Col28A1) is found in skin dermis and the sciatic nerve (71). Collagen type VII alpha 1 chain (Col7A1) is commonly found in the dermis of the skin and the bladder, and is associated with epidermolysis bullosa acquisita (71). The expression of the collagen genes, Col1A2 and Col3A1, in RORγ -/- fibroblasts was reduced, respectively, 6- and 4-fold compared with wild-type fibroblasts, whereas Col1A1 showed only a -1.4-fold change (Table 4). Interestingly, Col28A1 expression was decreased 61-fold in RORγ -/- fibroblasts compared with wild-type cells. Of all the collagen genes analyzed, only Col7A1 showed an increase in expression (24-fold) in RORγ -/- fibroblasts compared with wild-type.
BMP6 belongs to the family of TGF-β-related growth factors, plays an important role in the proliferation and development of the epidermis, and mutations have been implicated in the development of psoriasis in humans (72). BMP6 is strongly expressed in mouse skin under pathological conditions, such as inflammation (72). BMP6 mRNA was expressed at a 2.9-fold lower level in RORγ -/- fibroblasts compared with wild-type cells. TGF-β receptor 3 mRNA expression decreased 6.1-fold, whereas TGFβ1 expression was not significantly changed (1.7-fold) (Table 4).
Hepatocyte growth factor (HGF) or scatter factor plays an anti-inflammatory and anti-apoptotic role (73). Transgenic HGF/scatter factor mice are vulnerable to UV light and serve as a model for pigmentation and skin pathology (73). HGF was expressed at 21-fold lower levels in RORγ -/- cells than wild-type cells (Table 4). Cyp1b1 expression was also down-regulated in RORγ -/- fibroblasts (9-fold) when compared with wild-type cells (Table 4). The expression of Cyb1b1 was stimulated by secosteroids tested in wild-type fibroblasts, but not in cells lacking RORγ (Fig. 5). Cytochrome P450 CYP1b1 (CYP1b1) belongs to the cytochrome P450 family of heme-containing monooxygenases and plays a key role the metabolic activation of polycyclic aromatic hydrocarbons (74). Cyp1b1 is found in skin keratinocytes, and it can produce sterols that affect tumorigenesis (74).
Figure 5.
mRNA levels of genes involved in fibrosis after treatment of fibroblasts from RORγ +/+ and RORγ -/- mice. Fibroblasts from RORγ +/+ (WT) and RORγ -/- (KO) mice were treated with 100 nM of the secosteroids indicated, or ethanol vehicle (control). Data are presented as mean ± SD, n = 3. Analysis was done using the t-test: *P < 0.05, **P < 0.01, ***P < 0.001 or ****P < 0.0001 versus control (ethanol). KO, knockout; ROR, retinoic acid-related orphan receptor; WT, wild-type.
In summary, the RNA-seq analysis shows dramatic differences in the gene transcriptome between wild-type and RORγ -/- fibroblasts in response to several secosteroids. The differences at least in part correlate with some of the phenotypic differences described in this paper. However, it is possible that this effect is not solely related to the RORγ receptor because different factors may also contribute to the observed differences, which represents a challenge for future investigation.
Quantitative RT-PCR was used to determine whether the presence of RORγ in murine fibroblasts is important for the expression of genes involved in fibrotic processes upon treatment with the CYP11A1-derived vitamin D3 metabolites. The expression of genes encoding collagen (Col1A2 and Col3A1) was decreased in wild-type fibroblasts treated with 1,25(OH)2D3 or 20(OH)D3 (and also by 1,20(OH)2D3 in the case of Col3A1) (Fig. 5). This effect was reversed in RORγ -/- cells, in which an increase in the expression of the collagen genes was seen after treatment with all the secosteroids tested, 11- to 13-fold with 20(OH)D3 and 20,23(OH)2D3) (Fig. 5).
The expression of TGF-β and its receptor (Tgfbr) is upregulated in tumors of the skin (72). TGFβ1 plays an important role in skin cell proliferation, inflammation, and apoptosis, specifically in the epidermis (72) and Tgfβ11 KO mice exhibit delayed wound healing (75). The expression of theTgfb1 gene was not significantly altered by the treatment of wild-type fibroblasts with any of the secosteroids, whereas all secosteroids stimulated its expression in the RORγ -/- fibroblasts, 11- and 20-fold, respectively, for 20(OH)D3 and 20,23(OH)2D3 (Fig. 5). Our data suggest that besides the VDR (76), RORγ may also be involved in the action of vitamin D3 derivatives in the wound healing process.
MMP1b or fibroblast collagenase is an interstitial collagenase that is involved in tissue repair, remodeling of skin and wound healing, tumor invasion, and atherosclerosis (70). The expression of Mmp1b, as well as Hgf and Bmp7, were significantly higher in wild-type fibroblasts after treatment with secosteroids with only 1 exception (1,20,23(OH)3D3 on Hgf) (Fig. 5). In contrast, Hgf 1 expression was not significantly altered in RORγ -/- fibroblasts treated with the secosteroids, whereas the expression of Mmp1b and Bmp7 was significantly reduced. These results suggest a possible role for RORγ in the anti-inflammatory action of vitamin D3 derivatives.
Concluding Discussion
This study shows that oral delivery of 20(OH)D3, a product of CYP11A1 action on vitamin D3, can attenuate bleomycin-induced dermal fibrosis in mice, similar to what we reported before for IP administration (24). CYP11A-derived secosteroids, like 1,25(OH)2D3, inhibited the proliferation of wild-type dermal fibroblasts, but increased the proliferation of RORγ -/- and RORγ +/- fibroblasts. Similarly, collagen production was decreased by the secosteroids in the wild-type fibroblasts, but increased in RORγ -/- cells. Thus, the inhibition of fibroblast proliferation and collagen synthesis by secosteroids is dependent on an adequate level of RORγ gene expression as one copy of RORγ (as in heterozygotes) is not sufficient to generate the same effects as in wild-type fibroblasts.
Comparison of gene expression profiles by RNA-seq identified numerous genes that were differentially expressed in RORγ -/- fibroblasts compared with wild-type cells. Treatment of wild-type fibroblasts with secosteroids revealed large differences in gene expression between 20(OH)D3- and 1,20(OH)2D3-treated cells, with a subset of 382 genes regulated by both secosteroids. This was also seen with RORγ -/- fibroblasts, with both 20(OH)D3 and 1,20(OH)2D3 regulating the expression of 431 genes. These observations suggested different mechanisms of action and may be related to differences in the ability of these 2 secosteroids to activate receptors and in the case of wild-type, fibroblasts likely include RORγ, VDR, and AhR as reported for keratinocytes (68). The involvement of some of these receptors in RORγ -/- fibroblasts requires further study.
We also identified a correlation between fibroblasts lacking RORγ expression and the expression of genes related to fibrosis. We further demonstrated that the effects of CYP11A1-derived hydroxyvitamin D3 derivatives were different between RORγ -/- and wild-type dermal fibroblasts, with reduced expression of fibrosis-related genes generally only occurring in fibroblasts with functional RORγ. Our findings indicate that RORγ facilitates the inhibition of the profibrotic activities by secosteroids in murine fibroblasts. In summary, our study identifies a role for RORγ in the regulation of skin fibrosis.
Acknowledgments
Financial Support: The study was supported by National Institutes of Health (NIH) grants 1RO1 AR052190 and 1R21 AR0665051 to A.T.S. and A.P., and 1R01AR073004-01A1 and R01AR071189-01A1 and by a VA merit grant (no. 1I01BX004293-01A1) to A.T.S., and R21 AI149267-01A1 to C.R. and A.T.S. and by the Intramural Research Program of the National Institute of Environmental Health Sciences, NIH Z01-ES-101585 to A.M.J.
Glossary
Abbreviations
- AhR
aryl hydrocarbon receptor
- BMP
bone morphogenetic protein
- FBS
fetal bovine serum
- FC
fold change
- GSEA
gene set enrichment analysis
- HGF
hepatocyte growth factor
- KO
knockout
- MMP
matrix metallopeptidase
- RNA-seq
RNA sequencing
- ROR
retinoic acid-related orphan receptor
- RXR
retinoid X receptor
- VDR
vitamin D receptor
- VEGF
vascular endothelial growth factor
Additional Information
Disclosure Summary: The authors have declared that no conflict of interest exists.
Data Availability
All data generated or analyzed during this study are included in this published article or in the data repositories listed in References.
References
- 1. Jetten AM. Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nucl Recept Signal. 2009;7:e003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Jetten AM, Takeda Y, Slominski A, Kang HS. Retinoic acid-related orphan receptor γ (RORγ): connecting sterol metabolism to regulation of the immune system and autoimmune disease. Curr Opin Toxicol. 2018;8:66-80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Jetten AM, Kang HS, Takeda Y. Retinoic acid-related orphan receptors α and γ: key regulators of lipid/glucose metabolism, inflammation, and insulin sensitivity. Front Endocrinol (Lausanne). 2013;4:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Takeda Y, Kang HS, Freudenberg J, DeGraff LM, Jothi R, Jetten AM. Retinoic acid-related orphan receptor γ (RORγ): a novel participant in the diurnal regulation of hepatic gluconeogenesis and insulin sensitivity. Plos Genet. 2014;10(5):e1004331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Meissburger B, Ukropec J, Roeder E, et al. Adipogenesis and insulin sensitivity in obesity are regulated by retinoid-related orphan receptor gamma. EMBO Mol Med. 2011;3(11):637-651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Tinahones FJ, Moreno-Santos I, Vendrell J, et al. The retinoic acid receptor-related orphan nuclear receptor γ1 (RORγ1): a novel player determinant of insulin sensitivity in morbid obesity. Obesity (Silver Spring). 2012;20(3):488-497. [DOI] [PubMed] [Google Scholar]
- 7. Ueda E, Kurebayashi S, Sakaue M, Backlund M, Koller B, Jetten AM. High incidence of T-cell lymphomas in mice deficient in the retinoid-related orphan receptor RORgamma. Cancer Res. 2002;62(3):901-909. [PubMed] [Google Scholar]
- 8. Solt LA, Banerjee S, Campbell S, Kamenecka TM, Burris TP. ROR inverse agonist suppresses insulitis and prevents hyperglycemia in a mouse model of type 1 diabetes. Endocrinology. 2015;156(3):869-881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Dai J, Choo MK, Park JM, Fisher DE. Topical ROR inverse agonists suppress inflammation in mouse models of atopic dermatitis and acute irritant dermatitis. J Invest Dermatol. 2017;137(12):2523-2531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Jetten AM, Cook DN. (Inverse) agonists of retinoic acid-related orphan receptor γ: regulation of immune responses, inflammation, and autoimmune disease. Annu Rev Pharmacol Toxicol. 2020;60:371-390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Smith SH, Peredo CE, Takeda Y, et al. Development of a topical treatment for psoriasis targeting RORγ: from bench to skin. Plos One. 2016;11(2):e0147979. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Whitehead GS, Kang HS, Thomas SY, et al. Therapeutic suppression of pulmonary neutrophilia and allergic airway hyperresponsiveness by a RORgammat inverse agonist. JCI Insight. 2019;5(14):e125528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Xu T, Wang X, Zhong B, Nurieva RI, Ding S, Dong C. Ursolic acid suppresses interleukin-17 (IL-17) production by selectively antagonizing the function of RORgamma t protein. J Biol Chem. 2011;286(26):22707-22710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Huh JR, Littman DR. Small molecule inhibitors of RORγt: targeting Th17 cells and other applications. Eur J Immunol. 2012;42(9):2232-2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Solt LA, Griffin PR, Burris TP. Ligand regulation of retinoic acid receptor-related orphan receptors: implications for development of novel therapeutics. Curr Opin Lipidol. 2010;21(3):204-211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Slominski AT, Kim TK, Takeda Y, et al. RORα and ROR γ are expressed in human skin and serve as receptors for endogenously produced noncalcemic 20-hydroxy- and 20,23-dihydroxyvitamin D. Faseb J. 2014;28(7):2775-2789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Slominski AT, Kim TK, Hobrath JV, et al. Endogenously produced nonclassical vitamin D hydroxy-metabolites act as “biased” agonists on VDR and inverse agonists on RORα and RORγ. J Steroid Biochem Mol Biol. 2017;173:42-56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Slominski A, Semak I, Zjawiony J, et al. The cytochrome P450scc system opens an alternate pathway of vitamin D3 metabolism. Febs J. 2005;272(16):4080-4090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Slominski A, Semak I, Wortsman J, et al. An alternative pathway of vitamin D metabolism. Cytochrome P450scc (CYP11A1)-mediated conversion to 20-hydroxyvitamin D2 and 17,20-dihydroxyvitamin D2. Febs J. 2006;273(13):2891-2901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Tuckey RC, Li W, Zjawiony JK, et al. Pathways and products for the metabolism of vitamin D3 by cytochrome P450scc. Febs J. 2008;275(10):2585-2596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Slominski AT, Kim TK, Shehabi HZ, et al. In vivo evidence for a novel pathway of vitamin D3 metabolism initiated by P450scc and modified by CYP27B1. Faseb J. 2012;26(9):3901-3915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Tuckey RC, Li W, Shehabi HZ, et al. Production of 22-hydroxy metabolites of vitamin d3 by cytochrome p450scc (CYP11A1) and analysis of their biological activities on skin cells. Drug Metab Dispos. 2011;39(9):1577-1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Slominski AT, Kim TK, Li W, Yi AK, Postlethwaite A, Tuckey RC. The role of CYP11A1 in the production of vitamin D metabolites and their role in the regulation of epidermal functions. J Steroid Biochem Mol Biol. 2014;144 Pt A:28-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Slominski A, Janjetovic Z, Tuckey RC, et al. 20S-hydroxyvitamin D3, noncalcemic product of CYP11A1 action on vitamin D3, exhibits potent antifibrogenic activity in vivo. J Clin Endocrinol Metab. 2013;98(2):E298-E303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Slominski AT, Kim TK, Janjetovic Z, et al. 20-Hydroxyvitamin D2 is a noncalcemic analog of vitamin D with potent antiproliferative and prodifferentiation activities in normal and malignant cells. Am J Physiol Cell Physiol. 2011;300(3):C526-C541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Slominski AT, Janjetovic Z, Fuller BE, et al. Products of vitamin D3 or 7-dehydrocholesterol metabolism by cytochrome P450scc show anti-leukemia effects, having low or absent calcemic activity. Plos One. 2010;5(3):e9907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Janjetovic Z, Tuckey RC, Nguyen MN, Thorpe EM Jr, Slominski AT. 20,23-dihydroxyvitamin D3, novel P450scc product, stimulates differentiation and inhibits proliferation and NF-kappaB activity in human keratinocytes. J Cell Physiol. 2010;223(1):36-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Janjetovic Z, Zmijewski MA, Tuckey RC, et al. 20-Hydroxycholecalciferol, product of vitamin D3 hydroxylation by P450scc, decreases NF-kappaB activity by increasing IkappaB alpha levels in human keratinocytes. Plos One. 2009;4(6):e5988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Janjetovic Z, Brozyna AA, Tuckey RC, et al. High basal NF-κB activity in nonpigmented melanoma cells is associated with an enhanced sensitivity to vitamin D3 derivatives. Br J Cancer. 2011;105(12):1874-1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Chaiprasongsuk A, Janjetovic Z, Kim TK, et al. CYP11A1-derived vitamin D3 products protect against UVB-induced inflammation and promote keratinocytes differentiation. Free Radic Biol Med. 2020;155:87-98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Wang J, Slominski A, Tuckey RC, et al. 20-hydroxyvitamin D3 inhibits proliferation of cancer cells with high efficacy while being non-toxic. Anticancer Res. 2012;32(3):739-746. [PMC free article] [PubMed] [Google Scholar]
- 32. Skobowiat C, Oak AS, Kim TK, et al. Noncalcemic 20-hydroxyvitamin D3 inhibits human melanoma growth in in vitro and in vivo models. Oncotarget. 2017;8(6):9823-9834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Kim TK, Wang J, Janjetovic Z, et al. Correlation between secosteroid-induced vitamin D receptor activity in melanoma cells and computer-modeled receptor binding strength. Mol Cell Endocrinol. 2012;361(1-2):143-152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Chaiprasongsuk A, Janjetovic Z, Kim TK, et al. Protective effects of novel derivatives of vitamin D3 and lumisterol against UVB-induced damage in human keratinocytes involve activation of Nrf2 and p53 defense mechanisms. Redox Biol. 2019;24:101206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Bikle DD. The vitamin D receptor: a tumor suppressor in skin. Discov Med. 2011;11(56):7-17. [PMC free article] [PubMed] [Google Scholar]
- 36. Lin Z, Marepally SR, Goh ESY, et al. Investigation of 20S-hydroxyvitamin D3 analogs and their 1α-OH derivatives as potent vitamin D receptor agonists with anti-inflammatory activities. Sci Rep. 2018;8(1):1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Lin Z, Chen H, Belorusova AY, et al. 1α,20S-dihydroxyvitamin D3 interacts with vitamin D receptor: crystal structure and route of chemical synthesis. Sci Rep. 2017;7(1):10193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Zbytek B, Janjetovic Z, Tuckey RC, et al. 20-Hydroxyvitamin D3, a product of vitamin D3 hydroxylation by cytochrome P450scc, stimulates keratinocyte differentiation. J Invest Dermatol. 2008;128(9):2271-2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Slominski AT, Kim TK, Janjetovic Z, et al. Differential and overlapping effects of 20,23(OH)(2)D3 and 1,25(OH)(2)D3 on gene expression in human epidermal keratinocytes: identification of AhR as an alternative receptor for 20,23(OH)(2)D3. Int J Mol Sci. 2018;19(10):3072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Steinmayr M, André E, Conquet F, et al. staggerer phenotype in retinoid-related orphan receptor alpha-deficient mice. Proc Natl Acad Sci U S A. 1998;95(7):3960-3965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Kobayashi H, Kromminga A, Dunlop TW, et al. A role of melatonin in neuroectodermal-mesodermal interactions: the hair follicle synthesizes melatonin and expresses functional melatonin receptors. Faseb J. 2005;19(12):1710-1712. [DOI] [PubMed] [Google Scholar]
- 42. Slominski A, Fischer TW, Zmijewski MA, et al. On the role of melatonin in skin physiology and pathology. Endocrine. 2005;27(2):137-148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Markiewicz A, Brożyna AA, Podgórska E, et al. Vitamin D receptors (VDR), hydroxylases CYP27B1 and CYP24A1 and retinoid-related orphan receptors (ROR) level in human uveal tract and ocular melanoma with different melanization levels. Sci Rep. 2019;9(1):9142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Brożyna AA, Jóźwicki W, Roszkowski K, Filipiak J, Slominski AT. Melanin content in melanoma metastases affects the outcome of radiotherapy. Oncotarget. 2016;7(14):17844-17853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Ecoeur F, Weiss J, Kaupmann K, Hintermann S, Orain D, Guntermann C. Antagonizing retinoic acid-related-orphan receptor gamma activity blocks the T helper 17/interleukin-17 pathway leading to attenuated pro-inflammatory human keratinocyte and skin responses. Front Immunol. 2019;10:577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Pattanaik D, Brown M, Postlethwaite BC, Postlethwaite AE. Pathogenesis of systemic sclerosis. Front Immunol. 2015;6:272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Tao Q, Wang B, Zheng Y, Jiang X, Pan Z, Ren J. Vitamin D prevents the intestinal fibrosis via induction of vitamin D receptor and inhibition of transforming growth factor-beta1/Smad3 pathway. Dig Dis Sci. 2015;60(4):868-875. [DOI] [PubMed] [Google Scholar]
- 48. Takeda Y, Jothi R, Birault V, Jetten AM. RORγ directly regulates the circadian expression of clock genes and downstream targets in vivo. Nucleic Acids Res. 2012;40(17):8519-8535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Kurebayashi S, Ueda E, Sakaue M, et al. Retinoid-related orphan receptor gamma (RORgamma) is essential for lymphoid organogenesis and controls apoptosis during thymopoiesis. Proc Natl Acad Sci U S A. 2000;97(18):10132-10137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Li W, Chen J, Janjetovic Z, et al. Chemical synthesis of 20S-hydroxyvitamin D3, which shows antiproliferative activity. Steroids. 2010;75(12):926-935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Tang EK, Li W, Janjetovic Z, et al. Purified mouse CYP27B1 can hydroxylate 20,23-dihydroxyvitamin D3, producing 1alpha,20,23-trihydroxyvitamin D3, which has altered biological activity. Drug Metab Dispos. 2010;38(9):1553-1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Tuckey RC, Janjetovic Z, Li W, et al. Metabolism of 1alpha-hydroxyvitamin D3 by cytochrome P450scc to biologically active 1alpha,20-dihydroxyvitamin D3. J Steroid Biochem Mol Biol. 2008;112(4-5):213-219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Subramanian A, Tamayo P, Mootha VK, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102(43):15545-15550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Zhou Y, Zhou B, Pache L, et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10(1):1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Janjetovic Z, Nahmias ZP, Hanna S, et al. Melatonin and its metabolites ameliorate ultraviolet B-induced damage in human epidermal keratinocytes. J Pineal Res. 2014;57(1):90-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Ross A, Munger S, Capel B. Bmp7 regulates germ cell proliferation in mouse fetal gonads. Sex Dev. 2007;1(2):127-137. [DOI] [PubMed] [Google Scholar]
- 57. Arai KY, Hara T, Nagatsuka T, et al. Postnatal changes and sexual dimorphism in collagen expression in mouse skin. Plos One. 2017;12(5):e0177534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Nejak-Bowen K, Orr A, Bowen WC Jr, Michalopoulos GK. Conditional genetic elimination of hepatocyte growth factor in mice compromises liver regeneration after partial hepatectomy. PLoS One. 2013;8(3):e59836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Reiners JJ Jr, Jones CL, Hong N, Clift RE, Elferink C. Downregulation of aryl hydrocarbon receptor function and cytochrome P450 1A1 induction by expression of Ha-ras oncogenes. Mol Carcinog. 1997;19(2):91-100. [DOI] [PubMed] [Google Scholar]
- 60. Greiling D, Thieroff-Ekerdt R. 1alpha,25-dihydroxyvitamin D3 rapidly inhibits fibroblast-induced collagen gel contraction. J Invest Dermatol. 1996;106(6):1236-1241. [DOI] [PubMed] [Google Scholar]
- 61. Slominski A, Kim TK, Zmijewski MA, et al. Novel vitamin D photoproducts and their precursors in the skin. Dermatoendocrinol. 2013;5(1):7-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Slominski AT, Li W, Bhattacharya SK, et al. Vitamin D analogs 17,20S(OH)2pD and 17,20R(OH)2pD are noncalcemic and exhibit antifibrotic activity. J Invest Dermatol. 2011;131(5):1167-1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Bikle DD. Vitamin D metabolism and function in the skin. Mol Cell Endocrinol. 2011;347(1-2):80-89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Oda Y, Hu L, Nguyen T, et al. Vitamin D receptor is required for proliferation, migration, and differentiation of epidermal stem cells and progeny during cutaneous wound repair. J Invest Dermatol. 2018;138(11):2423-2431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Luderer HF, Nazarian RM, Zhu ED, Demay MB. Ligand-dependent actions of the vitamin D receptor are required for activation of TGF-β signaling during the inflammatory response to cutaneous injury. Endocrinology. 2013;154(1): 16-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Ito I, Waku T, Aoki M, et al. A nonclassical vitamin D receptor pathway suppresses renal fibrosis. J Clin Invest. 2013;123(11):4579-4594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Janjetovic Z, Postlethwaite A, Kang HS, et al. Supplemental Tables from: Antifibrogenic activities of CYP11A1-derived vitamin D3-hydroxyderivatives are dependent on RORγ. Dryad Digital Repository Deposited 17 July. 2020; doi: 10.5061/dryad.41ns1rnbp. [DOI]
- 68. Slominski AT, Chaiprasongsuk A, Janjetovic Z, et al. Photoprotective properties of vitamin D and lumisterol hydroxyderivatives. Cell Biochem Biophys. 2020;78(2):165-180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Iyer RP, Patterson NL, Fields GB, Lindsey ML. The history of matrix metalloproteinases: milestones, myths, and misperceptions. Am J Physiol Heart Circ Physiol. 2012;303(8):H919-H930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Balbín M, Fueyo A, Knäuper V, et al. Identification and enzymatic characterization of two diverging murine counterparts of human interstitial collagenase (MMP-1) expressed at sites of embryo implantation. J Biol Chem. 2001;276(13):10253-10262. [DOI] [PubMed] [Google Scholar]
- 71. Shoulders MD, Raines RT. Collagen structure and stability. Annu Rev Biochem. 2009;78:929-958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Wach S, Schirmacher P, Protschka M, Blessing M. Overexpression of bone morphogenetic protein-6 (BMP-6) in murine epidermis suppresses skin tumor formation by induction of apoptosis and downregulation of fos/jun family members. Oncogene. 2001;20(53):7761-7769. [DOI] [PubMed] [Google Scholar]
- 73. Wolnicka-Glubisz A, Pecio A, Podkowa D, Plonka PM, Grabacka M. HGF/SF increases number of skin melanocytes but does not alter quality or quantity of follicular melanogenesis. Plos One. 2013;8(11):e74883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Slominski AT, Zmijewski MA, Semak I, et al. Cytochromes p450 and skin cancer: role of local endocrine pathways. Anticancer Agents Med Chem. 2014;14(1):77-96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Crowe MJ, Doetschman T, Greenhalgh DG. Delayed wound healing in immunodeficient TGF-beta 1 knockout mice. J Invest Dermatol. 2000;115(1):3-11. [DOI] [PubMed] [Google Scholar]
- 76. Ding J, Kwan P, Ma Z, et al. Synergistic effect of vitamin D and low concentration of transforming growth factor beta 1, a potential role in dermal wound healing. Burns. 2016;42(6):1277-1286. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
All data generated or analyzed during this study are included in this published article or in the data repositories listed in References.





