Abstract
Background:
Previously, we demonstrated that the ketone body, β-hydroxybutyrate, is a potent antihypertensive and reno-protective metabolite in Dahl Salt-Sensitive rats. However, the mechanism by which β-hydroxybutyrate confers these beneficial effects is understudied. Here we focused on determining whether the reno-protective effect of β-hydroxybutyrate is due to its known ability to epigenetically remodel chromatin via histone β-hydroxybutyrylation.
Methods:
We used the same animal protocol previously used for the discovery of the renoprotective effect of β-hydroxybutyrate. Briefly, post-weaning, male and female Dahl Salt-Sensitive rats were split into two groups and supplemented with or without 1,3-butanediol for 6 weeks. At euthanasia, circulating β-hydroxybutyrate was quantitated. Renal homogenates were examined for histone 3 lysine 9 β-hydroxybutyrylation, chromatin occupancy, transcriptomic and proteomic profiles with validations.
Results:
Rats supplemented with 1,3-butanediol had higher circulating β-hydroxybutyrate, renal histone β-hydroxybutyrylation and significant remodeling of chromatin. Notably, regions of the genome associated with lipid catabolism were predominantly in an open chromatin configuration, leading to active transcription and translation. The most highly upregulated gene actively transcribed and translated was 3-hydroxy-3-methyglutaryl CoA Synthase 2 (Hmgcs2), a gene responsible for the biosynthesis of β-hydroxybutyrate in mitochondria. In contrast, regions with more compact chromatin structures contained immune function genes, protein tyrosine phosphatase receptor type C (Ptprc) and lymphocyte cytosolic protein 1 (Lcp1), which were suppressed.
Conclusions:
These results reveal that renal epigenetic histone β-hydroxybutyrylation is a novel mechanism by which transcriptional regulation of both energy metabolism and immune function occur concomitantly and contribute to renoprotection in the hypertensive Dahl rat.
Keywords: Blood pressure, histone, energy metabolism, ketone body, epigenetics, chromatin remodeling, kidney
Graphical Abstract
Introduction
β-hydroxybutyrate is a ketone body produced in the liver during the breakdown of fatty acids during low carbohydrate intake, fasting, or exercise1. It is a crucial alternate energy substrate which makes β-hydroxybutyrate essential during states of ketosis, where the body shifts from using carbohydrates to using fats for energy1,2. β-hydroxybutyrate is not only a fuel; it also functions as a signaling molecule that can influence gene expression2. It modulates pathways related to energy metabolism, inflammation, and oxidative stress3–5. β-hydroxybutyrate is shown to inhibit histone deacetylases, which affect gene transcription, potentially promoting longevity and reducing inflammation4,6,7. Particularly related to inflammation, β-hydroxybutyrate is reported to downregulate the expression of the NLRP3 inflammasome7–9. Due to all of these known beneficial effects, there is a growing interest in leveraging β-hydroxybutyrate to help manage chronic metabolic diseases, such as type 2 diabetes, obesity, and cardiovascular diseases3,9–13.
Metabolism plays a key role in hypertension14. Previously, we and others reported metabolic perturbations in salt-sensitive hypertension14–16. Previously, we examined the effect of reconstituting β-hydroxybutyrate in the Dahl S rat and discovered that it has a profound antihypertensive effect, which was associated with a betterment of kidney damage and renal function9. However, the molecular mechanism by which β-hydroxybutyrate protects kidneys remains largely unknown.
Here we focused on the kidney and specifically examined whether the mechanism underlying the beneficial effect of β-hydroxybutyrate was due to β-hydroxybutyrylation, which is a newer epigenetic histone modification caused by β-hydroxybutyrate17. Data in support of histone β-hydroxybutyrylation mediated epi-transcriptional regulation of energy metabolism and immune function as a novel reno-protective mechanism are presented.
Methods
Please see the Major Resources Table in the Supplemental Materials (Table S1).
Data Availability Statement:
All data and materials have been made publicly available at the GEO database (GSE298451 and GSE298452).
Experimental Model and Study Details:
Male and female Dahl Salt-Sensitive rats (SS/Jr) from the original colony maintained at the University of Toledo College of Medicine and Life Sciences were used for this study. Rats weaned at 28 days were placed on a high salt diet (2% NaCl) with or without 1,3-butanediol (20% v/v with drinking water). After 6 weeks of supplementation, rats were sacrificed, and tissues were harvested.
Blood Pressure Measurements
Rats were surgically implanted with radiotelemetry transmitters as described previously 18. Post-surgery, rats were housed individually and allowed to recover prior to recording their BP using the DSI software and equipment (https://www.datasci.com/). Systolic blood pressure was collected at 5-minute intervals and analyzed using the Dataquest A.R.T 4.2 Software.
Monitoring Energy Metabolism and Activity
Metabolic parameters were measured in 12 to 13-week-old male rats after 6 weeks of 1,3-butanediol supplementation. Rats were housed individually for 24 hours in the Comprehensive Laboratory Animal Monitoring System (CLAMS, Columbus Instruments). Oxygen consumption (VO2) and carbon dioxide production (VCO2) were sampled sequentially for 5 seconds in a 10-minute interval and motor activity was recorded every second in X and Z dimensions. Respiratory exchange ratio (RER) was calculated as VCO2/VO2.
Food Intake
Food given to rats housed in CLAMS was measured before and after 24 hours. The difference was recorded as food intake.
Measurement of Serum β-hydroxybutyrate
Serum β-hydroxybutyrate level was measured using a colorimetric assay kit from Cayman Chemicals as previously described9,10.
Histone Extraction
Snap-frozen kidney samples were homogenized for histone extraction using the EpiQuik histone extraction kit and quantitated using the Bicinchoninic Acid method19. Proteins were aliquoted and stored at −80°C until further use.
Western Blotting
Histones were resolved by gel electrophoresis on a 4-20% gradient SDS PAGE gel and probed using primary antibody against histone H3 lysine 9 (H3K9) β-hydroxybutyrylation, H3, ribosomal protein S6 and phospho-S6 ribosomal protein. from PTM Biolabs. Please refer to the major resources table in Table S1 for details.
Molecular Analyses
Detailed methods for Molecular Analyses are under Supplementary Methods. Briefly, kidneys were examined for chromatin accessibility via ATAC-seq20–31 and Chromatin immunoprecipitation (ChIP) assays32–36, gene expression via RNA-seq37,38, and real-time PCR39, and protein abundance by mass spectrometry-based quantitative proteomic analysis40. Primer sequences are in Tables S2 and S3.
Pathway Analyses
KEGG based pathway enrichment analyses were conducted using ShinyGOv0.77 (FDR cut off <0.05)41. Reactome pathway knowledgebase was used to generate pathways42.
Transmission Electron Microscopy and Analysis of Mitochondrial Morphology
Transmission electron microscopy was used to examine the impact of β-hydroxybutyrylation on mitochondrial morphology in the proximal tubule epithelial cells as detailed under Supplementary Methods.
Quantification of Peripheral T cells Proliferation
Peripheral lymphocytes were isolated from blood using the Histopaque method43. Briefly, lymphocytes (1x106 cells/well) were stained with carboxyfluorescein diacetate succinimidyl ester (CFSE, Sigma, 5 μM), washed 3 times in cold IX PBS and cultured in 1% heat inactivated fetal bovine serum in DMEM media for 5 days followed by staining with anti-rat CD3-APC antibody for getting CD3+ T cells population. The proliferation of CFSE-stained cells was analyzed by quantifying the degree of CFSE dilution in proliferated cells via flow cytometry (BD AccuriTM C6 Plus, BD Biosciences). BD Accuri software was used to analyze the proliferation. Data presented in the histogram indicate CFSE-labeled CD3+ T cells.
Histological Analyses
Kidneys were fixed in 10% neutral buffered formalin for 24 hours and transferred to 70% ethanol. Then, kidney tissues were embedded in paraffin and 5-μm-thick-sections were prepared. Rabbit anti-PMP70 antibody (ThermoFisher Scientific, Cat# PA1-650, 1:100 dilution) was used and detected by using Vectastain Elite ABC kit and SigmaFast 3,3-Diamino-benzidine tablets. The images were captured by VS120 Virtual Slide Microscope (Olympus) and analyzed using Olyvia Ver.2.9.
Quantification and Statistical Analyses
GraphPad Prism version 9.3.1 was used for data analysis. Statistical analyses were performed using the students’ T-test. For comparison with two groups across multiple time intervals, data were analyzed by two-way repeated measures ANOVA. Data with a p-value < 0.05 was considered significant. All data: Mean ± SEM. Statistical analyses for multi-omics data are presented in the supplementary methods.
Results
1,3-Butanediol is a Precursor of β-hydroxybutyrate and Lowers Blood Pressure in Both Sexes
Oral administration of 1,3-butanediol to male rats increased their circulating β-hydroxybutyrate and lowered blood pressure9. Here, we first examined whether the ability of 1,3-butanediol to reconstitute serum β-hydroxybutyrate and lower blood pressure was sex-independent. As seen in Figures 1A, 1B, 1C, and 1D, both male and female rats responded to 1,3-butanediol treatment with significant elevation of circulating β-hydroxybutyrate and reduced systolic blood pressure. However, the magnitude of the 1,3-butanediol to β-hydroxybutyrate conversion as well as the BP lowering effect was higher in males than in females. This indicates that the conversion of 1,3-butanediol to β-hydroxybutyrate and the BP lowering effect occurred independently of sex, but to a much greater extent in males. Unfortunately, while we did not collect renal parameters from the current study, our previous study has demonstrated that the observed reduction in blood pressure was strongly associated with decreased urinary protein excretion, lower renal fibrosis, and lower protein casts9.
Figure 1: Histone β-hydroxybutyrylation is elevated with 1,3-butanediol treatment.
Groups of S rats on a high salt diet (2% Nacl) were supplemented with or without 1,3-butanediol (20% v/v) in their drinking water as described in the methods section. (A-B) Systolic blood pressure of males (n= 5-6/group) and females (n= 5/group). (C-D) Serum levels of β-hydroxybutyrate in males (n=4-6/group) and females (n=6-8/group). (E-F) Immunoblotting for β-hydroxybutyrylation. (G-H) Quantification of the blots. (n=4-7/group), BHB: β-hydroxybutyrate, H3K9BHB: Histone 3 lysine 9 β-hydroxybutyrylation, H3: Histone 3. All data are mean ±SEM, *p<0.05, **p<0.01, ****p<0.0001.
Enhanced Histone β-hydroxybutyrylation in 1,3-Butanediol Treated Rats
Next, we examined histone 3 lysine-9 β-hydroxybutyrylation, which is the modification reported to be affected during starvation17. As seen in Figures 1C and 1D, both males and females treated with 1,3-butanediol, exhibited a significant increase in renal histone 3 lysine-9 β-hydroxybutyrylation. These data are presented with equal protein loading of isolated histones. Despite this, for reasons unknown, we noticed that the levels of H3 were also elevated in samples from the rats treated with 1,3-butanediol. However, to maintain rigor, β-hydroxybutyrylation was quantified using H3 as the normalizing factor. As seen in Figures 1E and 1F, β-hydroxybutyrylation was more prominent in males than in females. Subsequent multi-omics studies were conducted in male rats and validated in both sexes. Henceforth, the group of rats with increased renal histone 3 lysine 9 β-hydroxybutyrylation will be referred to as the BHB group.
Histone β-hydroxybutyrylation Promoted Large-Scale Chromatin Remodeling
To assess the extent of differential chromatin accessibility attributed to histone β-hydroxybutyrylation, we performed ATAC-sequencing. In the BHB group, 3494 genomic regions or loci (regardless of whether they were protein coding or not) were detected in the open configuration implying enhanced accessibility for transcriptional regulation (Figure 2A). Similarly, 7404 loci were ‘open’ in the control group indicating that in the BHB group these regions were ‘closed’ or compacted regions of the genome with decreased accessibility for transcription (Figure 2A). In Figure 2B, the x-axis denotes the genomic annotations, and the Y-axis denotes proportions of loci (genomic regions) in each category falling into the annotation type. The proportion is calculated using the total number of loci in each category with a particular annotation (number at the top of each bar divided by the total number of annotations in the respective category). Most of the remodeling occurred in the interCGI regions (regions of the genome that lie between CpG islands) followed by intergenic regions (parts of the genome that lie between genes) and introns. Interestingly, 249 and 196 promoter regions were open in the BHB and control groups respectively (Figure 2B).
Figure 2: Chromatin accessibility, gene expression, and protein levels are altered with 1,3-butanediol treatment.
(A) Volcano plot for BHB vs. control groups. Each data point is a tested locus. The vertical lines correspond to the logFC cutoff. (B) Annotation summary of loci tested in BHB vs control groups. X-axis: genomic annotations; Y-axis proportion of loci in each category within each type of annotation. All Tested loci (red bars), serve as a background by which to compare the significant loci (blue and green bars). Loci or genomic regions were calculated based on differentially open if FDR < 0.05 and |logFC| > 0.585. (C) Pathways associated with the upregulated genes of BHB group. (D) Pathways associated with upregulated proteins in the BHB group. (E) Venn diagram showing common upregulated genes. Different shades of blue circles depict ATACseq, RNAseq, and Proteomics data and numbers within the circles are numbers of genes/proteins significantly upregulated. (F) Chromatin accessibility for genes of interest. The y-axis represents chromatin cut sites and thus open chromatin, and x-axis represents the chromatin location of genes of interest. Black peaks represent the control group, and green peaks represent BHB group. (n=3/group).
Histone β-hydroxybutyrylation-mediated Remodeling of the Renal Transcriptome and Proteome
To uncover the extent to which histone β-hydroxybutyrylation regulated the transcriptome, we performed renal RNA-sequencing. In the BHB group compared to the control, a total of 186 genes were upregulated, and 490 genes were downregulated (Table S4). Pathway analysis using these upregulated genes revealed that the top 2 upregulated pathways were pantothenate and CoA biosynthesis and fatty acid degradation (Figure 2C).
Next, to examine the impact of the chromatin-remodeling responsive transcriptome on the proteome, we conducted a mass spectrometry-based quantitative proteomic analysis. A total of 119 and 79 proteins were significantly upregulated and downregulated, respectively, in the BHB group compared to control (Table S5). Pathway analysis of the proteomics data using these upregulated proteins indicated that butanoate metabolism, fatty acid biosynthesis, and fatty acid metabolism were the upregulated pathways based on the differentially expressed proteins (Figure 2D). Interestingly, pathways identified in the transcriptome which were also upregulated in the proteome were fatty acid metabolism, tryptophan metabolism, PPAR signaling and metabolic pathways (Figures 2C, 2D).
In contrast, the downregulated transcriptome (Table S4) and downregulated proteome signatures (Table S5) were highly related to the overall theme of immunity and infection. The common pathways identified in both downregulated transcriptome and proteome were ECM-receptor interaction, focal adhesion, phagosome, toxoplasmosis, viral myocarditis, amoebiasis, ECM receptor interaction, and leishmaniasis (Figures S1A, S1B).
Chromatin Remodeling by Histone β-hydroxybutyrylation Upregulated Lipid Catabolism
Next, we explored if the observed alterations in the transcriptome and proteome of the BHB group was specifically due to chromatin remodeling via histone β-hydroxybutyrylation. We prioritized common differentially regulated outputs between chromatin states identified by ATAC-Seq, RNA-seq, and proteomics datasets. Such a combinatorial analysis revealed that there were 10 upregulated proteins aligned with enhanced transcription caused by histone β-hydroxybutyrylation (Figure 2E). Intriguingly, four of the top genes share a common function, lipid metabolism. These were 3-Hydroxy-3-methylglutaryl-Coenzyme A synthase 2 (Hmgcs2), Acetyl-Coenzyme A acetyltransferase 1B (Acca1b), Cytochrome P450, family 2, subfamily d, polypeptide 4 (Cyp2d4), Cytochrome P450, family 2, subfamily e, polypeptide 1 (Cyp2e1) (Figure 2E). Chromatin accessibility of the promoter regions of Hmgcs2, Acaa1b, Cyp2d4, and Cyp2e1 were higher in the BHB group compared to control (Figure 2F). These data indicated that histone β-hydroxybutyrylation caused open chromatin within the promoter regions of Hmgcs2, Acaa1b, Cyp2d4, and Cyp2e1. Reactome pathway analysis using the 10 genes identified metabolism as the top upregulated pathway along with metabolism of lipids, synthesis and metabolism of ketone bodies, and β-oxidation of very long chain fatty acids (Figure S2E).
Collectively, these results led us to further prioritize β-hydroxybutyrylation as a key mechanism promoting lipid catabolism as a primary source for energy.
Metabolic Reprogramming in Response to Elevated Histone β-hydroxybutyrylation
To determine if rats treated with 1,3-butanediol were preferring lipids for energy, we housed both groups of control and BHB rats in the Comprehensive Lab Animal Monitor System (CLAMS) and monitored their metabolic parameters. Overall, both oxygen consumption (VO2) and carbon dioxide production (VCO2) were lower in the BHB group compared to control (Figures 3A, 3B). Importantly, respiratory exchange ratio (RER) was dramatically decreased in the BHB group compared to the control group (Figure 3C). A lower RER points to lipids, but not carbohydrates as the primary source of energy44,45. This data provided further evidence for histone β-hydroxybutyrylation enhanced transcription of key metabolic genes in the lipid mobilizing pathways. Those key genes, Hmgcs2, Acaa1b, Cyp2d4 and Cyp2e1 likely contributed to a preferential switch in bioenergetic fuel source to lipids.
Figure 3: 1,3-butanediol treatment reduces respiratory exchange ratio (RER) and inhibits mammalian target of rapamycin complex 1 (mTORC1).
(A-C) Metabolic CLAMS data showed reduced VO2, VCO2, and RER level in the BHB group compared to controls. Black line: control group and green line: BHB group. (n=5/group) (D-E) Reduced phospho-S6 ribosomal protein levels were found in male rats treated with BHB compared to control. See also Figure S5 for data from female rats. BHB: β-hydroxybutyrate. (n=5-6/group) All data are mean±SEM; *p ≤ 0.05, **p ≤ 0.01, and ***p ≤ 0.001.
Hmgcs2, Acaa1b, Cyp2d4, and Cyp2e1 are Bonafide Targets Linking Epigenetic Renal Histone-3 β-hydroxybutyrylation to Energy Metabolism
In addition to β-hydroxybutyrylation, β-hydroxybutyrate is a histone deacetylase (HDAC) inhibitor and is also known to epigenetically modify histones by acetylation7,46. Therefore, we tested and confirmed that H3K9Acetylation, and H3K23Acetylation were significantly upregulated with 1,3-butanediol treatment (Figures S3A, S3B). Hence, it was important to determine whether the 4 upregulated energy metabolism genes, Hmgcs2, Acaa1b, Cyp2d4, and Cyp2e1, which were prioritized in our study were specific targets of β-hydroxybutyrylation. We therefore performed Chromatin immunoprecipitation (ChIP) assays using an antibody specific to β-hydroxybutyryl-histone H3 (Lys 9) β-hydroxybutyrate. ChIP assay using this antibody revealed that promoter regions of Hmgcs2, Acaa1b, Cyp2d4, and Cyp2e1 were significantly enriched in the BHB group (Figure 4A), thus confirming that these genes were bonafide targets of β-hydroxybutyrylation.
Figure 4: Validation of common upregulated genes through CHIP-qPCR, Real time PCR, and proteomics.
(A) CHIP-qPCR data demonstrating enrichment of Hmgcs2, Acaa1b, Cyp2d4, and Cyp2e1 in the BHB group compared to control (n=3 replicates/group). Black bar: Control, Green bar: BHB. (B) Real time PCR showed higher expression of Hmgcs2, Acaa1b, Cyp2d4, and Cyp2e1 in BHB group compared to control (housekeeping gene is L36a) (n=5-6/group) (C) Normalized relative abundance of Hmgcs2, Acaa1b, Cyp2d4, Cyp2e1 proteins (n=3-6/group). See also Figure S2 for female data. Black open circle- control, green closed circle- BHB. All data are mean±SEM; *p< 0.05, **p< 0.01, ***p< 0.001, ****p <0.0001 and #p=0.0567.
Next, transcripts of these genes were quantitated by real time qPCR analysis. The abundance of Hmgcs2 was prominently upregulated in the BHB group compared to control (Figure 4B, Figure S2A). Similarly, the abundances of Acaa1b, Cyp2d4 and Cyp2e1 were also upregulated in the BHB group compared to controls (Figure 4B, Figures S2B–S2D). Aligned with these data, quantitative mass-spectrometry using high-quality MS3 spectra indicated that all 4 protein products of Hmgcs2, Acaa1b, Cyp2d4 and Cyp2e1 were significantly upregulated in the BHB group compared to control (Figure 4C). Collectively, these data demonstrate that β-hydroxybutyrate, via conferring epigenetic histone β-hydroxybutyrylation, upregulated enzymes which mobilize lipids for energy metabolism.
Upregulation of Hmgcs2-mediated Fatty Acid Oxidation Increased Renal Proximal Tubule Epithelial Cell Mitochondrial Circulatory Index
One of the prominent targets upregulated due to histone β-hydroxybutyrylation was the mitochondrial protein Hmgcs2. Hmgcs2 is a cytosolic and rate-limiting enzyme regulating mitochondrial fatty acid oxidation to promote ketogenesis47,48. Ketogenesis is largely attributed to the liver, but recent work demonstrates that Hmgcs2 in the kidneys also participates in this pathway49,50. Because ketogenesis in the liver increases mitochondrial circularity, we asked if such alterations in the ultrastructure of mitochondria also occur in the kidney. Transmission electron microscopy revealed more circular mitochondria in the proximal tubule epithelial cells from the BHB group compared to the elongated mitochondria in the controls (Figure 5A). This observation was confirmed by morphometric analyses of the measurement of average mitochondrial area, area/perimeter, and circularity index of these mitochondria. As seen in Figures 5B, 5C and 5D, each of these morphometric measures were elevated in the BHB supplemented group compared to the controls. These data support our conclusion that histone β-hydroxybutyrylation mediated upregulation of Hmgcs2 to promote ketosis contributed to enhanced renal mitochondrial circularity index.
Figure 5: Kidney proximal tubule epithelial cell mitochondrial circulatory index was found to be higher in BHB group compared to control.
(A) Representative transmission electron microscopy images of renal mitochondria. Orange arrows point to mitochondria. (B-D) Bar graph shows quantification (B) Average mitochondrial area (C) Area/Perimeter, and (D) Circularity index. BHB: β-hydroxybutyrate. n=30 images per group. A total of 3 rats per group were used. All data are mean±SEM, ***p< 0.001, and ****p<0.0001.
Upregulation of Acaa1b-mediated Fatty Acid Oxidation Increased Peroxisomal Biogenesis
Fatty acid oxidation occurs both in mitochondria and peroxisomes51–56. Whereas mitochondria are the main site of oxidation of medium-and long-chain fatty acids, peroxisomes catalyze the β-oxidation of a distinct set of fatty acids, including very-long-chain fatty acids54–56. While Hmgcs2 is located within mitochondria, interestingly, another target of histone β-hydroxybutyrylation, Acaa1b, which is responsible for β-oxidation of fatty acids, is located within peroxisomes57,58. In response to metabolic stress, peroxisomes proliferate by upregulating their biogenesis59. One of the factors required for peroxisomal biogenesis is peroxisomal biogenesis factor 11 γ60. Interestingly, proteomic data revealed that Pex11γ was increased in the BHB group, which indicated that peroxisomal biogenesis was promoted in the BHB group (Figure S4A). To further confirm this observation, we examined the renal sections for the abundance of peroxisomes using the classical marker, peroxisomal membrane protein 70 (Pmp70). Peroxisome staining was more prominent in the BHB group compared to control (Figures S4B, S4C) suggesting that the upregulation of the peroxisomal target of histone-β-hydroxybutyrylation, Acaa1b, placed an increased demand for fatty acid oxidation, which in turn promoted peroxisome biogenesis.
β-hydroxybutyrylation-mediated Enhanced Fatty Acid Oxidation Inhibits Mammalian Target of Rapamycin Complex 1 (mTORC1).
Stimulation of peroxisome biogenesis by drugs such as rapamycin are known to inhibit mammalian target of rapamycin complex 1 (mTORC1) activity61,62. Hyperactivation of mTORC1 signaling is associated with several human diseases whereas suppression of mTORC1 is known to curb senescence, extend lifespan of yeast, C. elegans, Drosophila and mice and promote autophagy63. Importantly, mTORC1 inhibits fatty acid β-oxidation, whereas inhibition of mTORC1 by rapamycin promotes fatty acid oxidation64. Rapamycin suppresses mTORC1 signaling to ameliorate kidney injury and hypertension in S rats16. Based on the similarity in function between rapamycin and the targets of β-hydroxybutyrylation to promote fatty acid oxidation, we hypothesized that mTORC1 is inhibited in the BHB group to protect kidneys. In support, mTORC1 was significantly inhibited in the BHB group compared to control (Figures 3D, 3E, and Figures S5A, S5B).
Chromatin Remodeling by Histone β-hydroxybutyrylation Resulted in Downregulation of Immune Function Genes
Next, we examined the downregulated loci, transcripts and proteins in the BHB group (Figure 2A). These are ‘open’ chromatin regions in the control group and ‘closed’ chromatin region in the BHB group. Similar to the shared upregulated genes and proteins, the combinatorial analysis using ATAC-seq, transcriptomic, and proteomic datasets showed that 9 proteins were downregulated in all the 3 analyses (Figure 6A). Among these, the top 2 were protein tyrosine phosphatase receptor type C (Ptprc) and Lymphocyte cytosolic protein 1 (Lcp1). Chromatin accessibility of the promoter regions of Ptprc and Lcp1 were lower in the BHB group compared to control (Figure 6B). These results indicate that histone β-hydroxybutyrylation mediated chromatin compaction of the promoter regions of Ptprc and Lcp1 contributed to the observed lower transcription (Figure 6C) and translation (Figure 6D) of these loci.
Figure 6: Chromatin accessibility, gene expression, protein levels, and CD3+ T cells proliferation were altered with 1,3-butanediol treatment.
(A) Venn diagrams showing common downregulated genes and proteins leading to downregulation of immune function pathways. Different shades of blue circles are used to indicate ATACseq, RNAseq, and Proteomics data. The gray box lists the names of common downregulated genes and proteins, beneath which are shown the associated downregulated pathways. (B) Chromatin accessibility at the promoter region of Ptprc and Lcp1 promoter. The y-axis represents chromatin cut sites and thus open chromatin, and x-axis represents the chromatin location of genes of interest. Black peaks represent the control group, and green peaks represent BHB group. (C) Real time PCR data showing reduced expression with Ptprc and Lcp1 in the 1,3-butanediol supplementation. (n=5-6/group) (D) Normalized relative abundances of Ptprc (Cd45) and Lcp1 detected in the quantitative proteomics study. Black open circle– control, green closed circle- BHB. (n=3/group). (E) Peripheral white blood cells and Lymphocytes in the control and BHB treated male S rats. (F) Representative histogram for percent CFSE-positive CD3+ T cells andquantification for percent CFSE-positive CD3+ T cells in control and BHB groups after 5 days. Data represented as Mean ± SEM and N was plotted for control and BHB group. N=7-8/group. WBC: white blood cells, Lym: Lymphocytes, CFSE: carboxyfluorescein diacetate succinimidyl ester, CD3: Cluster of differentiation 3, P= Parent population, D1= Daughter 1, and D2= Daughter 2 population All data are mean±SEM, *p<0.05, **p< 0.01, ***p< 0.001 and ****p<0.0001.
Unlike the upregulated pathways of energy metabolism, reactome pathway analysis of the downregulated genes showed that the overall downregulated pathways due to histone β-hydroxybutyrylation were related to immune function. These were phosphorylation of CD3 and TCR zeta chains, downstream TCR signaling, translocation of ZAP-70 to immunological synapse, PD-1 signaling, immune system, interferon gamma signaling, interleukin-12 family signaling, and TCR signaling (Figure 6A).
Histone β-hydroxybutyrylation Promoted Downregulation of CD45 and LCP-1
Next, we focused on the two top genes which were prominently downregulated in our ATAC-seq study, Ptprc and Lcp-1. Lower expression of both Ptprc and Lcp1 in the BHB group was confirmed by qPCR (Figure 6C). These data correlated with lower abundance of the protein products of these genes (CD45 and LCP-1, respectively) in the BHB group compared to the controls (Figure 6D). CD45 and LCP-1 are both important for the function of T cells and dysfunctional regulation of T cells are implicated in renal disease and hypertension. Therefore, we focused on examining T cells using CD3+ as a pan marker reporting for all T cells in our experimental groups. However, as expected, the CHIP assay showed comparable enrichment of promoter regions of Ptprc and Lcp1 in both the groups (Figures S6A, S6B), indicating that these are not direct targets of β-hydroxybutyrylation but consequential due to chromatin compaction.
Lower Expression of CD45 and LCP-1 Enhanced CD3+ T cell Proliferation
Both CD45 and LCP-1 are present in all hematopoietic cells65–68. Therefore, we examined if the downregulation of these two genes in the BHB group was facilitated by the epigenetic action of β-hydroxybutyrate reflected in immunomodulatory effects. Analysis of complete blood count (CBC) showed a trend toward reduced circulating monocyte and neutrophil counts with BHB treatment; but these differences did not reach statistical significance. However, a significant decrease in the level of circulating white blood cells (WBCs), specifically lymphocytes, was noted in the BHB group (Figure 6E). Similarly, mean corpuscular volume, mean corpuscular hemoglobin, mean platelet volume, plateletcrit and platelet distribution width were also lower in the BHB group (Figure S7). Taken together, these data affirm an immunomodulatory function of β-hydroxybutyrate (Figure 6A, Figures S7A–S7F). In alignment, the parent population of CD3+ T cells was lower in the BHB group compared to controls (Figure 6F). Intriguingly, CD3+ T cells from the BHB group exhibited increased proliferation as indicated by a higher population of dividing cells (D2) while maintaining a similar population of D1 cells (Figure 6F). These findings suggest that the lower expression of CD45 and LCP-1 in the BHB group likely contributed to a rapid turnover of T lymphocytes by depleting existing parental cells and promoting the generation of a sufficient number of new effector T cells to maintain immune homeostasis. However, a key limitation is that we were unable to quantify T cells in the kidney tissues. Notably, in our previously published data, we have demonstrated that BHB inhibits the infiltration of CD68+ macrophages into the kidneys9.
Discussion
This study was designed to delineate the molecular mechanism underlying the previously documented protective effect of BHB9. Specifically, we examined the known function of β-hydroxybutyrate to epigenetically remodel chromatin by histone β-hydroxybutyrylation17. Chromatin remodeling by histone β-hydroxybutyrylation exerted dynamic effects of distinctly modifying the renal transcriptome and proteome to promote both mitochondrial and peroxisomal regulation of fatty acid catabolism while parallelly dampening immune cell function. We have methodically dissected and characterized the major loci, transcripts and proteins contributing to the specific consequence of renal histone β-hydroxybutyrylation to conclude that epigenetic chromatin remodeling by histone β-hydroxybutyrylation simultaneously regulates the dual processes of upregulation of fatty acid catabolism and dampening of immune cells. These results constitute a new mechanism underlying the dynamic beneficial renoprotective effect of β-hydroxybutyrate.
Histone β-hydroxybutyrylation was first identified as a new epigenetic modification occurring in the mouse liver in response to starvation17, which is a condition simultaneously promoting ketosis and production of β-hydroxybutyrate11,69,70. Its epigenetic action to post translationally modify histones by β-hydroxybutyrylation has been subsequently studied in the context of a variety of pathologies including cardiomyopathy71, depression72, glomerulosclerosis73, and lung adenocarcinoma74, but none in any renal or hemodynamic studies. While our previous study on the benefit of β-hydroxybutyrate9 has been reproduced by others in different contexts such as preeclampsia and diabetic kidney disease, the underlying molecular mechanism has remained elusive75–77. In this context, our results presented in this study are the first to reveal that post translational modification of histones by β-hydroxybutyrylation is a novel mechanism protecting kidneys.
BHB is a known epigenetic modifier46. Along with its direct modification of histones, it can also indirectly modify histones by histone deacetylase (HDAC) inhibition. Our results here showed an increase of β-hydroxybutyrylation as well as acetylation (Figure 1, Figures S3A, S3B). This raised the question of which modification was responsible for the upregulation of lipid catabolism. To address this, we performed ChIP-PCR using a specific anti-H3K9 β-hydroxybutyrylation antibody and identified Hmgcs2, Acaa1b, Cyp2d4, and Cyp2e1 as bonafide targets of histone β-hydroxybutyrylation.
The food intake was significantly lowered in the BHB group, whereas the 24-hour activity was not different between the BHB and control group (Figures S6C, S6D). Pathway analysis revealed an upregulation of fatty acid metabolism, which could be the reason for lowering of food intake in the BHB group. Mitochondria, a major organelle involved in fat metabolism, undergoes morphological changes based on nutrient availability and stress conditions. mTORC1 is a known regulator of numerous cellular processes, such as cell proliferation, metabolism, and cell growth78. mTORC1 regulates mitochondrial metabolism and controls mitochondrial biogenesis79,80. Mitochondrial fission and apoptosis are controlled by mechanistic/mammalian target of rapamycin complex 1 (mTORC1) mediated stimulated translation of mitochondrial fission process 1 (MTFP1)80. Our study showed a distinct mitochondrial morphological alteration occurring in response to β-hydroxybutyrate, whereby mitochondria were more circular. Reduced circularity of mitochondria is reported to increase fatty acid oxidation, which concur with our findings81. Further, our data demonstrate that increased fatty acid oxidation was linked to a significant inhibition of mTORC1. In support, rapamycin, which is a uniquely specific mTOR inhibitor, increases fatty acid oxidation64. Similarly, fenofibrate, which is reported to have beneficial cardiovascular and renal effects, increased fatty acid oxidation and inhibited mTOR82. Importantly, in the context of salt-sensitive hypertension and kidney injury, Kumar et al., have shown that the inhibition of mammalian target of rapamycin 1 (mTORC1) ameliorates both hypertension and renal injury83. These data taken together with our findings reported in the current study leads us to propose that epigenetic chromatin remodeling by histone β-hydroxybutyrylation promotes the renal fatty acid oxidation-mTORC1 axis to protect kidneys and lower hypertension.
An interesting finding of our study is that Hmgcs2, which is the main enzyme catalyzing the production of β-hydroxybutyrate from Acetyl CoA, is also the target locus impacted by β-hydroxybutyrylation. This indicates that β-hydroxybutyrate autoregulates its own production by epigenetically remodeling chromatin to promote the transcription of Hmgcs2. If this process is perpetual, systemic β-hydroxybutyrate levels would remain consistently elevated after a single intervention to raise systemic β-hydroxybutyrate. Such a dysregulated continuous production of β-hydroxybutyrate would be detrimental as it would lead to ketoacidosis. Besides, β-hydroxybutyrate is elevated only during fasting and is lowered during the fed state9. Similarly, β-hydroxybutyrylation was discovered to be elevated only during fasting17. Collectively, these data support our conclusion that histone β-hydroxybutyrylation by β-hydroxybutyryrate is not perpetual but turned ‘off’ when β-hydroxybutyryrate is limiting by yet undiscovered mechanisms.
A very low-carbohydrate diet is reported to enhance human T-cell immunity through immunometabolic reprogramming84. Such a low carbohydrate diet could promote ketosis and inhibit mTOR. mTOR inhibition by Rapamycin, which also lowers hypertension, is shown to promote T cell hypo-responsiveness or anergy83. In alignment with these observations, the downregulated pathways generated from downregulated genes/proteins led to lowering of T cell signaling, which implies that the mechanism by which T cell signaling is affected by β-hydroxybutyrate is a result of epigenetic remodeling of chromatin to a condensed state in regions harboring Ptprc and Lcp1.
The ChIP assays performed in this study conclusively demonstrate that H3K9 β-hydroxybutyrylation contributes to the transcriptional upregulation of the genes Hmgcs2, Acaa1b, Cyp2d4 Cyp2e1. These are the only genes that are validated and thereby directly related to increased histone 3 lysine-9 βhydroxybutyrylation. All other genes enlisted as identified via ATACseq and RNAseq require additional confirmations.
The combinatorial ATACseq, RNAseq and proteomic datasets allow for the interpretation that the mechanistic effect of this remodeling is traced to impact renal energy metabolism and dampen the transcription of key genes in immune cell pathways. While our data are convincing to demonstrate that H3K9 β-hydroxybutyrylation cannot be ruled out as a mechanistic contributor, it may not be the sole mechanism. Further studies will be needed to clarify whether other factors could also contribute to the observed differential gene and protein expressions reported in our study.
Although our study reports a new epigenetic mechanism for lowering blood pressure and kidney damage, it has some additional limitations. First, the study is conducted in Dahl S rats, which needs further validation from other experimental models of hypertension and kidney damage. Whether this operates in humans remains unknown. Secondly, the observations presented are largely in males. Future studies are required to establish whether BHB mediated chromatin modification is a primary driver of the observed beneficial effect on BP and kidney damage. In addition, further in-depth studies are required to rule out the involvement of other known histone modifications affecting the genes identified as targets in our study. Lastly, the mechanism converging on mTOR requires additional studies to understand how mTOR inhibition lowers blood pressure and protects kidneys.
In summary, our findings reveal that renal histone β-hydroxybutyrylation causes a dynamic increase in chromatin accessibility to the promoter regions harboring lipid catabolizing genes while simultaneously decreasing accessibility to the promoter regions of genes contributing to immune-related functions. Together, these newly discovered mechanisms contribute to the beneficial effects of β-hydroxybutyrate on renal-hemodynamic health.
Perspectives
This is the first study which demonstrates that in a ketogenic state, histone β-hydroxybutyrylation indeed occurs in the kidney to remodel renal chromatin. Key target genes impacted for their transcription via this remodeling of chromatin have been identified. The mechanistic effect of this remodeling is traced to impact renal energy metabolism and dampen the transcription of key genes in immune cell pathways. Our study serves as the foundation for further research into intricate details on which cell types are affected by this epigenetic effect of β-hydroxybutyrate. Nevertheless, it establishes that histone β-hydroxybutyrylation is at least in part contributing to the dynamic effects of a ketone body on the kidney and hypertension.
Supplementary Material
Novelty and Relevance.
What is New?
Discovery of the epigenetic modification, H3K9 β-hydroxybutyrylation, as a novel mechanism underlying the beneficial effect of the ketone body β-hydroxybutyrate in curtailing kidney damage and lowering hypertension.
What is Relevant?
Our study expands current limited mechanistic knowledge on how a relatively new histone modification by the ketone body, β-hydroxybutyrate, can specifically upregulate lipid energy metabolism and downregulate harmful overactive immune cell function to improve kidney health.
Clinical/Pathophysiological Implications?
Starvation, intermittent fasting, keto diet, and exercise, all are lifestyle modifications recommended for patients with hypertension. Precisely how these operate to lower blood pressure is largely unknown. In this context, our finding that a ketone body, β-hydroxybutyrate, which is generated in all these lifestyle modifications can protect kidneys of a hypertensive rat model via histone β-hydroxybutyrylation to protect kidneys, is a notable mechanistic advancement.
Acknowledgements
Grant acknowledgements: NIH: BJ (R01-HL171401, R01-HL143082); MVK (R01DK134053); AHA: SA (25PRE1375711), IM (24PRE1186688), PS (855256), TY (852969); Crohn’s and Colitis Foundation: PS (854385); American Liver Foundation Liver Scholar Award: BSY; University of Toledo Startup funds: TY; Melanoma Research Foundation: MRF; University of Toledo Stimulus and Bridge Awards: IDS.
Non-standard Abbreviations and Acronyms:
- BHB
β-hydroxybutyrate
- H3K9bhb
Histone-3 lysine 9 β-hydroxybutyrylation
- H3K9Ac
Histone-3 lysine 9 acetylation
- H3K23Ac
Histone-3 lysine 23 acetylation
- S rats
Dahl Salt-Sensitive rats
- VO2
Volume of oxygen consumption
- VCO2
Volume of carbon dioxide production
- RER
Respiratory exchange ratio
- CLAMS
Comprehensive laboratory animal monitoring system
- ATAC-seq
Assay for transposase-accessible chromatin with sequencing
- ChIP
Chromatin immunoprecipitation
- Hmgcs2
3-Hydroxy-3-methylglutaryl-Coenzyme A synthase 2
- Acaa1b
Acetyl-Coenzyme A acetyltransferase 1B
- Cyp2d4
Cytochrome P450, family 2, subfamily d, polypeptide 4
- Cyp2e1
Cytochrome P450, family 2, subfamily e, polypeptide 1
- Pex11γ
Peroxisomal biogenesis factor 11 gamma
- Pmp70
Peroxisomal membrane protein 70
- mTORC1
Mammalian target of rapamycin complex 1
- Ptprc
Protein tyrosine phosphatase receptor type C
- Lcp1
Lymphocyte cytosolic protein 1
- MTFP1
Mitochondrial fission process 1
- MCV
Mean corpuscular volume
- MCH
Mean corpuscular hemoglobin
- MPV
Mean platelet volume
- PCT
Plateletcrit
- PDWs
Platelet distribution width-standard deviation
- PDWc
Platelet distribution width-coefficient of variation
Footnotes
Disclosure: All the authors declare no competing interests.
References
- 1.Wang L, Chen P, Xiao W. β-hydroxybutyrate as an Anti-Aging Metabolite. Nutrients. 2021;13(10):3420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Newman JC, Verdin E. β-Hydroxybutyrate: a signaling metabolite. Annual review of nutrition. 2017;37(1):51–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Huang C, Wang J, Liu H, Huang R, Yan X, Song M, Tan G, Zhi F. Ketone body β-hydroxybutyrate ameliorates colitis by promoting M2 macrophage polarization through the STAT6-dependent signaling pathway. BMC medicine. 2022;20(1):148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RD. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013;339(6116):211–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Hwang CY, Choe W, Yoon K-S, Ha J, Kim SS, Yeo E-J, Kang I. Molecular mechanisms for ketone body metabolism, signaling functions, and therapeutic potential in cancer. Nutrients. 2022;14(22):4932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Edwards C, Canfield J, Copes N, Rehan M, Lipps D, Bradshaw PC. D-beta-hydroxybutyrate extends lifespan in C. elegans. Aging (Albany NY). 2014;6(8):621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Deng Y, Xie M, Li Q, Xu X, Ou W, Zhang Y, Xiao H, Yu H, Zheng Y, Liang Y. Targeting mitochondria-inflammation circuit by β-hydroxybutyrate mitigates HFpEF. Circulation research. 2021;128(2):232–245. [DOI] [PubMed] [Google Scholar]
- 8.Youm Y-H, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, D’agostino D, Planavsky N, Lupfer C, Kanneganti TD. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome–mediated inflammatory disease. Nature medicine. 2015;21(3):263–269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chakraborty S, Galla S, Cheng X, Yeo J-Y, Mell B, Singh V, Yeoh B, Saha P, Mathew AV, Vijay-Kumar M. Salt-responsive metabolite, β-hydroxybutyrate, attenuates hypertension. Cell reports. 2018;25(3):677–689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.McCarthy CG, Chakraborty S, Singh G, San Yeoh B, Schreckenberger ZJ, Singh A, Mell B, Bearss NR, Yang T, Cheng X. Ketone body β-hydroxybutyrate is an autophagy-dependent vasodilator. JCI insight. 2021;6(20). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Fang Y, Chen B, Gong AY, Malhotra DK, Gupta R, Dworkin LD, Gong R. The ketone body β-hydroxybutyrate mitigates the senescence response of glomerular podocytes to diabetic insults. Kidney international. 2021;100(5):1037–1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ortí JE de la R, Cuerda-Ballester M, Sanchis-Sanchis CE, Lajara Romance JM, Navarro-Illana E, García Pardo MP. Exploring the impact of ketogenic diet on multiple sclerosis: obesity, anxiety, depression, and the glutamate system. Frontiers in Nutrition. 2023;10:1227431. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Hanners A, Melnyk BM, Volek J, Kelley MM. Ketogenic diet, African American women, and cardiovascular health: A systematic review. Worldviews on Evidence-Based Nursing. 2022;19(1):35–41. [DOI] [PubMed] [Google Scholar]
- 14.Chakraborty S, Mandal J, Yang T, Cheng X, Yeo J-Y, McCarthy CG, Wenceslau CF, Koch LG, Hill JW, Vijay-Kumar M. Metabolites and hypertension: insights into hypertension as a metabolic disorder: 2019 Harriet Dustan Award. Hypertension. 2020;75(6):1386–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Tian Z, Liu Y, Usa K, Mladinov D, Fang Y, Ding X, Greene AS, Cowley AW Jr, Liang M. Novel role of fumarate metabolism in dahl-salt sensitive hypertension. Hypertension. 2009;54(2):255–260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kumar V, Evans LC, Kurth T, Yang C, Wollner C, Nasci V, Zheleznova NN, Bukowy J, Dayton A, Cowley AW Jr. Therapeutic suppression of mTOR (mammalian target of rapamycin) signaling prevents and reverses salt-induced hypertension and kidney injury in Dahl salt-sensitive rats. Hypertension. 2019;73(3):630–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Xie Z, Zhang D, Chung D, Tang Z, Huang H, Dai L, Qi S, Li J, Colak G, Chen Y. Metabolic regulation of gene expression by histone lysine β-hydroxybutyrylation. Molecular cell. 2016;62(2):194–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mell B, Jala VR, Mathew AV, Byun J, Waghulde H, Zhang Y, Haribabu B, Vijay-Kumar M, Pennathur S, Joe B. Evidence for a link between gut microbiota and hypertension in the Dahl rat. Physiological genomics. 2015;47(6):187–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Walker JM. The bicinchoninic acid (BCA) assay for protein quantitation. The protein protocols handbook. 2009:11–15. [DOI] [PubMed] [Google Scholar]
- 20.Corces MR, Trevino AE, Hamilton EG, Greenside PG, Sinnott-Armstrong NA, Vesuna S, Satpathy AT, Rubin AJ, Montine KS, Wu B. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nature methods. 2017;14(10):959–962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Köster J, Rahmann S. Snakemake—a scalable bioinformatics workflow engine. Bioinformatics. 2012;28(19):2520–2522. [DOI] [PubMed] [Google Scholar]
- 22.McCarthy DJ, Chen Y, Smyth GK. Differential expression analysis of multifactor RNA-Seq experiments with respect to biological variation. Nucleic acids research. 2012;40(10):4288–4297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Cavalcante RG, Sartor MA. Annotatr: genomic regions in context. Bioinformatics. 2017;33(15):2381–2383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ou J, Liu H, Yu J, Kelliher MA, Castilla LH, Lawson ND, Zhu LJ. ATACseqQC: a Bioconductor package for post-alignment quality assessment of ATAC-seq data. BMC genomics. 2018;19:1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nature methods. 2012;9(4):357–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, Marth G, Abecasis G, Durbin R. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25(16):2078–2079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26(6):841–842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhang Y, Liu T, Meyer CA, Eeckhoute J, Johnson DS, Bernstein BE, Nusbaum C, Myers RM, Brown M, Li W. Model-based analysis of ChIP-Seq (MACS). Genome biology. 2008;9:1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Neph S, Kuehn MS, Reynolds AP, Haugen E, Thurman RE, Johnson AK, Rynes E, Maurano MT, Vierstra J, Thomas S. BEDOPS: high-performance genomic feature operations. Bioinformatics. 2012;28(14):1919–1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ewels P, Magnusson M, Lundin S, Käller M. MultiQC: summarize analysis results for multiple tools and samples in a single report. Bioinformatics. 2016;32(19):3047–3048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Orchard P, Kyono Y, Hensley J, Kitzman JO, Parker SCJ. Quantification, dynamic visualization, and validation of bias in ATAC-seq data with ataqv. Cell systems. 2020;10(3):298–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mehrotra A, Joe B, de la Serna IL. SWI/SNF chromatin remodeling enzymes are associated with cardiac hypertrophy in a genetic rat model of hypertension. Journal of Cellular Physiology. 2013;228(12):2337–2342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Perna A, Alberi LA. TF-ChIP method for tissue-specific gene targets. Frontiers in cellular neuroscience. 2019;13:95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Basuroy T, Dreier M, Baum C, Blomquist T, Trumbly R, Filipp FV, de la Serna IL. Epigenetic and pharmacological control of pigmentation via Bromodomain Protein 9 (BRD9). Pigment Cell & Melanoma Research. 2023;36(1):19–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Untergasser A, Cutcutache I, Koressaar T, Ye J, Faircloth BC, Remm M, Rozen SG. Primer3—new capabilities and interfaces. Nucleic acids research. 2012;40(15):e115–e115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kõressaar T, Lepamets M, Kaplinski L, Raime K, Andreson R, Remm M. Primer3_masker: integrating masking of template sequence with primer design software. Bioinformatics. 2018;34(11):1937–1938. [DOI] [PubMed] [Google Scholar]
- 37.Thompson O, von Meyenn F, Hewitt Z, Alexander J, Wood A, Weightman R, Gregory S, Krueger F, Andrews S, Barbaric I. Low rates of mutation in clinical grade human pluripotent stem cells under different culture conditions. Nature communications. 2020;11(1):1528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Martin M Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. journal. 2011;17(1):10–12. [Google Scholar]
- 39.Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2- ΔΔCT method. methods. 2001;25(4):402–408. [DOI] [PubMed] [Google Scholar]
- 40.Cheng X, Mell B, Alimadadi A, Galla S, McCarthy CG, Chakraborty S, Basrur V, Joe B. Genetic predisposition for increased red blood cell distribution width is an early risk factor for cardiovascular and renal comorbidities. Disease models & mechanisms. 2020;13(5):dmm044081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ge SX, Jung D, ShinyGO RY. A graphical gene-set enrichment tool for animals and plants, 2020, 36. DOI: 10.1093/bioinformatics/btz931.:2628–2629 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Milacic M, Beavers D, Conley P, Gong C, Gillespie M, Griss J, Haw R, Jassal B, Matthews L, May B. The reactome pathway knowledgebase 2024. Nucleic acids research. 2024;52(D1):D672–D678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Feldman DL, Mogelesky TC. Use of Histopaque for isolating mononuclear cells from rabbit blood. Journal of immunological methods. 1987;102(2):243–249. [DOI] [PubMed] [Google Scholar]
- 44.Pendergast DR, Leddy JJ, Venkatraman JT. A perspective on fat intake in athletes. Journal of the American College of Nutrition. 2000;19(3):345–350. [DOI] [PubMed] [Google Scholar]
- 45.Simonson DC, DeFronzo RA. Indirect calorimetry: methodological and interpretative problems. American Journal of Physiology-Endocrinology And Metabolism. 1990;258(3):E399–E412. [DOI] [PubMed] [Google Scholar]
- 46.He Y, Cheng X, Zhou T, Li D, Peng J, Xu Y, Huang W. β-Hydroxybutyrate as an epigenetic modifier: Underlying mechanisms and implications. Heliyon. 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Geisler CE, Ghimire S, Bogan RL, Renquist BJ. Role of ketone signaling in the hepatic response to fasting. American Journal of Physiology-Gastrointestinal and Liver Physiology. 2019;316(5):G623–G631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bai M, Wu M, Jiang M, He J, Deng X, Xu S, Fan J, Miao M, Wang T, Li Y. LONP1 targets HMGCS2 to protect mitochondrial function and attenuate chronic kidney disease. EMBO Molecular Medicine. 2023;15(2):e16581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Feola K, Venable AH, Broomfield T, Villegas M, Fu X, Burgess S, Huen SC. Hepatic ketogenesis is not required for starvation adaptation in mice. Molecular Metabolism. 2024;86:101967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Venable AH, Lee LE, Feola K, Santoyo J, Broomfield T, Huen SC. Fasting-induced HMGCS2 expression in the kidney does not contribute to circulating ketones. American Journal of Physiology-Renal Physiology. 2022;322(4):F460–F467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mi Y, Qi G, Vitali F, Shang Y, Raikes AC, Wang T, Jin Y, Brinton RD, Gu H, Yin F. Loss of fatty acid degradation by astrocytic mitochondria triggers neuroinflammation and neurodegeneration. Nature metabolism. 2023;5(3):445–465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Talari NK, Mattam U, Meher NK, Paripati AK, Mahadev K, Krishnamoorthy T, Sepuri NB V. Lipid-droplet associated mitochondria promote fatty-acid oxidation through a distinct bioenergetic pattern in male Wistar rats. Nature Communications. 2023;14(1):766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Liu S, Zhang H, Li Y, Zhang Y, Bian Y, Zeng Y, Yao X, Wan J, Chen X, Li J. S100A4 enhances protumor macrophage polarization by control of PPAR-γ-dependent induction of fatty acid oxidation. Journal for immunotherapy of cancer. 2021;9(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Panov AV, Mayorov VI, Dikalova AE, Dikalov SI. Long-chain and medium-chain fatty acids in energy metabolism of murine kidney mitochondria. International Journal of Molecular Sciences. 2022;24(1):379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cappa M, Todisco T, Bizzarri C. X-linked adrenoleukodystrophy and primary adrenal insufficiency. Frontiers in Endocrinology. 2023;14:1309053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Chevillard G, Clémencet M-C, Latruffe N, Nicolas-Francès V. Targeted disruption of the peroxisomal thiolase B gene in mouse: a new model to study disorders related to peroxisomal lipid metabolism. Biochimie. 2004;86(11):849–856. [DOI] [PubMed] [Google Scholar]
- 57.Antonenkov VD, Van Veldhoven PP, Waelkens E, Mannaerts GP. Comparison of the stability and substrate specificity of purified peroxisomal 3-oxoacyl-CoA thiolases A and B from rat liver. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 1999;1437(2):136–141. [DOI] [PubMed] [Google Scholar]
- 58.Liu C, Fu C, Lu Y, Sun J, Liu T, Wang Y, Wang A, Huang Y, Li Y. Integration of metabolomics and transcriptomics to reveal the mechanism of Gerberae piloselloidis herba in alleviating bronchial asthma. Journal of Ethnopharmacology. 2024;325:117852. [DOI] [PubMed] [Google Scholar]
- 59.Lodhi IJ, Semenkovich CF. Peroxisomes: a nexus for lipid metabolism and cellular signaling. Cell metabolism. 2014;19(3):380–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Li X, Gould SJ. PEX11 promotes peroxisome division independently of peroxisome metabolism. The Journal of cell biology. 2002;156(4):643–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Dunlop EA, Tee AR. Mammalian target of rapamycin complex 1: signalling inputs, substrates and feedback mechanisms. Cellular signalling. 2009;21(6):827–835. [DOI] [PubMed] [Google Scholar]
- 62.Eun SY, Lee JN, Nam I-K, Liu Z, So H-S, Choe S-K, Park R. PEX5 regulates autophagy via the mTORC1-TFEB axis during starvation. Experimental & Molecular Medicine. 2018;50(4):1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kucheryavenko O, Nelson G, von Zglinicki T, Korolchuk VI, Carroll B. The mTORC1-autophagy pathway is a target for senescent cell elimination. Biogerontology. 2019;20:331–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Sipula IJ, Brown NF, Perdomo G. Rapamycin-mediated inhibition of mammalian target of rapamycin in skeletal muscle cells reduces glucose utilization and increases fatty acid oxidation. Metabolism. 2006;55(12):1637–1644. [DOI] [PubMed] [Google Scholar]
- 65.Hermiston ML, Xu Z, Weiss A. CD45: a critical regulator of signaling thresholds in immune cells. Annual review of immunology. 2003;21(1):107–137. [DOI] [PubMed] [Google Scholar]
- 66.Rheinländer A, Schraven B, Bommhardt U. CD45 in human physiology and clinical medicine. Immunology letters. 2018;196:22–32. [DOI] [PubMed] [Google Scholar]
- 67.Kell MJ, Riccio RE, Baumgartner EA, Compton ZJ, Pecorin PJ, Mitchell TA, Topczewski J, LeClair EE. Targeted deletion of the zebrafish actin-bundling protein L-plastin (lcp1). PLoS One. 2018;13(1):e0190353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Wabnitz G, Balta E, Samstag Y. L-plastin regulates the stability of the immune synapse of naive and effector T-cells. Advances in biological regulation. 2017;63:107–114. [DOI] [PubMed] [Google Scholar]
- 69.Han Y, Bedarida T, Ding Y, Somba BK, Lu Q, Wang Q, Song P, Zou M-H. β-Hydroxybutyrate prevents vascular senescence through hnRNP A1-mediated upregulation of Oct4. Molecular Cell. 2018;71(6):1064–1078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Xia G, Wen Z, Zhang L, Huang J, Wang X, Liang C, Cui X, Cao X, Wu S. β-Hydroxybutyrate alleviates cartilage senescence through hnRNP A1-mediated up-regulation of PTEN. Experimental Gerontology. 2023;175:112140. [DOI] [PubMed] [Google Scholar]
- 71.Luo W, He M, Luo Q, Li Y. Proteome-wide analysis of lysine β-hydroxybutyrylation in the myocardium of diabetic rat model with cardiomyopathy. Frontiers in Cardiovascular Medicine. 2023;9:1066822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Chen L, Miao Z, Xu X. β-hydroxybutyrate alleviates depressive behaviors in mice possibly by increasing the histone3-lysine9-β-hydroxybutyrylation. Biochemical and biophysical research communications. 2017;490(2):117–122. [DOI] [PubMed] [Google Scholar]
- 73.Luo W, Yu Y, Wang H, Liu K, Wang Y, Huang M, Xuan C, Li Y, Qi J. Up-regulation of MMP-2 by histone H3K9 β-hydroxybutyrylation to antagonize glomerulosclerosis in diabetic rat. Acta Diabetologica. 2020;57:1501–1509. [DOI] [PubMed] [Google Scholar]
- 74.Huang J, Liang L, Jiang S, Liu Y, He H, Sun X, Li Y, Xie L, Tao Y, Cong L. BDH1-mediated LRRC31 regulation dependent on histone lysine βhydroxybutyrylation to promote lung adenocarcinoma progression. MedComm. 2023;4(6):e449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ishimwe JA, Baker MB, Garrett MR, Sasser JM. Periconceptional 1, 3-butanediol supplementation suppresses the superimposed preeclampsia-like phenotype in the Dahl salt-sensitive rat. American Journal of Physiology-Heart and Circulatory Physiology. 2022;322(2):H285–H295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Wan S-R, Teng F-Y, Fan W, Xu B-T, Li X-Y, Tan X-Z, Guo M, Gao C-L, Zhang C-X, Jiang Z-Z. BDH1-mediated βOHB metabolism ameliorates diabetic kidney disease by activation of NRF2-mediated antioxidative pathway. Aging (Albany NY). 2023;15(22):13384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kim MN, Moon JH, Cho YM. Sodium-glucose cotransporter-2 inhibition reduces cellular senescence in the diabetic kidney by promoting ketone body-induced NRF2 activation. Diabetes, Obesity and Metabolism. 2021;23(11):2561–2571. [DOI] [PubMed] [Google Scholar]
- 78.Szwed A, Kim E, Jacinto E. Regulation and metabolic functions of mTORC1 and mTORC2. Physiological reviews. 2021;101(3):1371–1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Liu X, Zhang Y, Ni M, Cao H, Signer RAJ, Li D, Li M, Gu Z, Hu Z, Dickerson KE. Regulation of mitochondrial biogenesis in erythropoiesis by mTORC1-mediated protein translation. Nature cell biology. 2017;19(6):626–638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Morita M, Prudent J, Basu K, Goyon V, Katsumura S, Hulea L, Pearl D, Siddiqui N, Strack S, McGuirk S. mTOR controls mitochondrial dynamics and cell survival via MTFP1. Molecular cell. 2017;67(6):922–935. [DOI] [PubMed] [Google Scholar]
- 81.Ngo J, Choi DW, Stanley IA, Stiles L, Molina AJA, Chen P, Lako A, Sung ICH, Goswami R, Kim M. Mitochondrial morphology controls fatty acid utilization by changing CPT1 sensitivity to malonyl-CoA. The EMBO journal. 2023;42(11):e111901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lian X, Gu J, Gao B, Li Y, Damodaran C, Wei W, Fu Y, Cai L. Fenofibrate inhibits mTOR-p70S6K signaling and simultaneously induces cell death in human prostate cancer cells. Biochemical and biophysical research communications. 2018;496(1):70–75. [DOI] [PubMed] [Google Scholar]
- 83.Kumar V, Wollner C, Kurth T, Bukowy JD, Cowley AW Jr. Inhibition of mammalian target of rapamycin complex 1 attenuates salt-induced hypertension and kidney injury in Dahl salt-sensitive rats. Hypertension. 2017;70(4):813–821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hirschberger S, Strauß G, Effinger D, Marstaller X, Ferstl A, Müller MB, Wu T, Hübner M, Rahmel T, Mascolo H. Very-low-carbohydrate diet enhances human T-cell immunity through immunometabolic reprogramming. EMBO Molecular Medicine. 2021;13(8):e14323. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data and materials have been made publicly available at the GEO database (GSE298451 and GSE298452).