Influence of Butyrate on Impaired Gene Expression in Colon from Patients with High Blood Pressure

Hypertension (HTN) is associated with gut dysbiosis and the depletion of butyrate-producing bacteria in animal models and people. Furthermore, fecal material transfer from donor hypertensive patients increases blood pressure in normotensive recipient animals and ameliorates HTN-associated pathophysiology. These observations have implications in the impaired interactions between the gut and gut microbiota in HTN. Although this concept is supported in animal models, little is known about human HTN. Therefore, our objective for this study was to compare gene expression with transcriptomics and its potential to influence microbiota in subjects with normal and high blood pressure (HBP). Colon samples from reference subjects with normal blood pressure (REF) and HBP were used for RNA-seq to analyze their transcriptomes. We observed the significant downregulation of gene sets governing immune responses (e.g., SGK1 and OASL), gut epithelial function (e.g., KRT20 and SLC9A3R1), gut microbiota (e.g., PPARG and CIDEC) and genes associated with cardiovascular and gut diseases (e.g., PLAUR and NLN) in HBP subjects; the expression of genes within these pathways correlated with blood pressure. Potential drug targets in the gut epithelium were identified using the Drug Gene International Database for possible use in HTN. They include peroxisome proliferator-activated receptor gamma (PPRG), active serum/glucocorticoid regulated kinase 1 (SGK1) and 3 beta-hydroxysteroid isomerase type II inhibitor (HSD3B). Finally, butyrate, a microbiota-derived short-chain fatty acid, restored the disrupted expression of certain functional genes in colonic organoids from HBP subjects. Patients with HBP exhibit a unique transcriptome that could underlie impaired gut–microbiota interactions. Targeting these interactions could provide a promising new therapeutic intervention for hypertension management.

[1]  S. Juraschek,et al.  Increases in Circulating and Fecal Butyrate are Associated With Reduced Blood Pressure and Hypertension: Results From the SPIRIT Trial , 2022, Journal of the American Heart Association.

[2]  Zhongli Wang,et al.  PPARG Silencing Improves Blood Pressure Control and Alleviates Renal Damage by Modulating RAS Circadian Rhythm in Hypertensive Rats. , 2022, Annals of Clinical and Laboratory Science.

[3]  C. Pepine,et al.  Gut Microbiome and Neuroinflammation in Hypertension , 2022, Circulation research.

[4]  K. He,et al.  The miR-182/Myadm axis regulates hypoxia-induced pulmonary hypertension by balancing the BMP- and TGF-β-signalling pathways in an SMC/EC-crosstalk-associated manner , 2021, Basic Research in Cardiology.

[5]  Wei Hu,et al.  Probiotic yogurt blunts the increase of blood pressure in spontaneously hypertensive rats via remodeling of the gut microbiota. , 2021, Food & function.

[6]  C. Pepine,et al.  Distinct Gene Expression Profiles in Colonic Organoids from Normotensive and the Spontaneously Hypertensive Rats , 2021, Cells.

[7]  D. Jin,et al.  Key Enzymes for the Mevalonate Pathway in the Cardiovascular System. , 2021, Journal of cardiovascular pharmacology.

[8]  C. Pepine,et al.  Butyrate Regulates COVID-19-Relevant Genes in Gut Epithelial Organoids From Normotensive Rats. , 2020, Hypertension.

[9]  Joshua F. McMichael,et al.  Integration of the Drug–Gene Interaction Database (DGIdb 4.0) with open crowdsource efforts , 2020, bioRxiv.

[10]  M. Raizada,et al.  Gut–brain–bone marrow axis in hypertension , 2020, Current opinion in nephrology and hypertension.

[11]  I. Kema,et al.  Different tryptophan-kynurenine metabolism profiles in human pulmonary arterial hypertension and animal models of pulmonary hypertension , 2020 .

[12]  J. P. Rigalli,et al.  Acute regulation of apical ABC transporters in the gut. Potential influence on drug bioavailability. , 2020, Pharmacological research.

[13]  M. Raizada,et al.  SARS-CoV-2 Receptor ACE-2 (Angiotensin-Converting Enzyme 2) Is Upregulated in Colonic Organoids From Hypertensive Rats , 2020, Hypertension.

[14]  E. Elinav,et al.  Interaction between microbiota and immunity in health and disease , 2020, Cell Research.

[15]  T. Spector,et al.  Deficiency of Prebiotic Fiber and Insufficient Signaling Through Gut Metabolite-Sensing Receptors Leads to Cardiovascular Disease , 2020, Circulation.

[16]  C. Pepine,et al.  Transcriptomic Signature of Gut Microbiome-Contacting Cells in Colon of Spontaneously Hypertensive Rats. , 2019, Physiological genomics.

[17]  C. Sigmund,et al.  PPARγ and RhoBTB1 in hypertension. , 2019, Current opinion in nephrology and hypertension.

[18]  F. Jouret,et al.  Gut Microbiota and Fecal Levels of Short-Chain Fatty Acids Differ Upon 24-Hour Blood Pressure Levels in Men. , 2019, Hypertension.

[19]  Hong Li,et al.  Calcium-Activated Chloride Channel A4 (CLCA4) Plays Inhibitory Roles in Invasion and Migration Through Suppressing Epithelial-Mesenchymal Transition via PI3K/AKT Signaling in Colorectal Cancer , 2019, Medical science monitor : international medical journal of experimental and clinical research.

[20]  J. Duarte,et al.  Critical Role of the Interaction Gut Microbiota – Sympathetic Nervous System in the Regulation of Blood Pressure , 2019, Front. Physiol..

[21]  M. Grandoch,et al.  Short-Chain Fatty Acid Propionate Protects From Hypertensive Cardiovascular Damage , 2018, Circulation.

[22]  J. Sierra,et al.  Higher Fecal Short-Chain Fatty Acid Levels Are Associated with Gut Microbiome Dysbiosis, Obesity, Hypertension and Cardiometabolic Disease Risk Factors , 2018, Nutrients.

[23]  Eun Woo Son,et al.  iDEP: an integrated web application for differential expression and pathway analysis of RNA-Seq data , 2018, BMC Bioinformatics.

[24]  C. Shively,et al.  Comparative Microbiome Signatures and Short-Chain Fatty Acids in Mouse, Rat, Non-human Primate, and Human Feces , 2018, Front. Microbiol..

[25]  James A. Hutchinson,et al.  Novel molecules mediate specialized functions of human regulatory macrophages , 2018, Current opinion in organ transplantation.

[26]  C. Pepine,et al.  Imbalance of gut microbiome and intestinal epithelial barrier dysfunction in patients with high blood pressure. , 2018, Clinical science.

[27]  C. Pepine,et al.  Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression , 2017, Gut.

[28]  C. Gieger,et al.  Transcriptome-Wide Analysis Identifies Novel Associations With Blood Pressure , 2017, Hypertension.

[29]  F. Perez-Vizcaino,et al.  Antihypertensive Effects of Probiotics , 2017, Current Hypertension Reports.

[30]  Weili Zhang,et al.  Gut microbiota dysbiosis contributes to the development of hypertension , 2017, Microbiome.

[31]  C. Pepine,et al.  Hypertension-Linked Pathophysiological Alterations in the Gut , 2017, Circulation research.

[32]  E. Boerwinkle,et al.  Genome-Wide and Gene-Based Meta-Analyses Identify Novel Loci Influencing Blood Pressure Response to Hydrochlorothiazide , 2017, Hypertension.

[33]  David S. Wishart,et al.  Heatmapper: web-enabled heat mapping for all , 2016, Nucleic Acids Res..

[34]  C. Pepine,et al.  Brain-Gut-Bone Marrow Axis: Implications for Hypertension and Related Therapeutics. , 2016, Circulation research.

[35]  S. Najjar,et al.  High-fat diet amplifies renal renin angiotensin system expression, blood pressure elevation, and renal dysfunction caused by Ceacam1 null deletion. , 2015, American journal of physiology. Endocrinology and metabolism.

[36]  C. Pepine,et al.  Gut Dysbiosis Is Linked to Hypertension , 2015, Hypertension.

[37]  S. Akira,et al.  IPS-1 differentially induces TRAIL, BCL2, BIRC3 and PRKCE in type I interferons-dependent and -independent anticancer activity , 2015, Cell Death and Disease.

[38]  Young-Joon Kim,et al.  Oligoadenylate synthase-like (OASL) proteins: dual functions and associations with diseases , 2015, Experimental & Molecular Medicine.

[39]  Andrew D. Johnson,et al.  A Meta-analysis of Gene Expression Signatures of Blood Pressure and Hypertension , 2015, PLoS genetics.

[40]  Anders K. Haakonsson,et al.  PPARγ and the global map of adipogenesis and beyond , 2014, Trends in Endocrinology & Metabolism.

[41]  W. Xia,et al.  Slc26a3 deficiency is associated with loss of colonic HCO3− secretion, absence of a firm mucus layer and barrier impairment in mice , 2014, Acta physiologica.

[42]  D. Hassett,et al.  Loss of NHE3 alters gut microbiota composition and influences Bacteroides thetaiotaomicron growth. , 2013, American journal of physiology. Gastrointestinal and liver physiology.

[43]  Barbara M. Bakker,et al.  The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism , 2013, Journal of Lipid Research.

[44]  J. M. Suh,et al.  PPARγ signaling and metabolism: the good, the bad and the future , 2013, Nature Medicine.

[45]  A. Ridley,et al.  MYADM controls endothelial barrier function through ERM-dependent regulation of ICAM-1 expression , 2013, Molecular biology of the cell.

[46]  J. Gordon,et al.  Olfactory receptor responding to gut microbiota-derived signals plays a role in renin secretion and blood pressure regulation , 2013, Proceedings of the National Academy of Sciences.

[47]  F. Lang,et al.  SGK, renal function and hypertension. , 2010, Journal of nephrology.

[48]  T. Nakayama,et al.  Association of HSD3B1 and HSD3B2 gene polymorphisms with essential hypertension, aldosterone level, and left ventricular structure. , 2010, European journal of endocrinology.

[49]  T. Hansen,et al.  Circulating soluble urokinase plasminogen activator receptor predicts cancer, cardiovascular disease, diabetes and mortality in the general population , 2010, Journal of internal medicine.

[50]  Ian R. Holzman,et al.  Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. , 2009, The Journal of nutrition.

[51]  W. Cefalu,et al.  Butyrate Improves Insulin Sensitivity and Increases Energy Expenditure in Mice , 2009, Diabetes.

[52]  A. Burlingame,et al.  Keratin 20 Serine 13 Phosphorylation Is a Stress and Intestinal Goblet Cell Marker* , 2006, Journal of Biological Chemistry.

[53]  S. Gray-Owen,et al.  CEACAM1: contact-dependent control of immunity , 2006, Nature Reviews Immunology.

[54]  K. Ranganna,et al.  Butyrate inhibits proliferation-induced Proliferating Cell Nuclear Antigen expression (PCNA) in rat vascular smooth muscle cells , 2000, Molecular and Cellular Biochemistry.

[55]  E. Levy,et al.  Butyrate induced Caco-2 cell apoptosis is mediated via the mitochondrial pathway , 2003, Gut.

[56]  S. Oparil,et al.  Sex hormones and hypertension. , 2002, Cardiovascular research.

[57]  J. L. Napoli 17β-Hydroxysteroid dehydrogenase type 9 and other short-chain dehydrogenases/reductases that catalyze retinoid, 17β- and 3α-hydroxysteroid metabolism , 2001, Molecular and Cellular Endocrinology.

[58]  T. Tai,et al.  Assessment of the function and effect of diabetes education programs in Taiwan. , 1999, Diabetes research and clinical practice.

[59]  U. Smith,et al.  Elevated fibrinogen and plasminogen activator inhibitor (PAI‐1) in hypertension are related to metabolic risk factors for cardiovascular disease , 1990, Journal of internal medicine.