Elsevier

Free Radical Biology and Medicine

Volume 172, 20 August 2021, Pages 90-100
Free Radical Biology and Medicine

Original article
Ketogenesis alleviates TNFα-induced apoptosis and inflammatory responses in intestinal cells

https://doi.org/10.1016/j.freeradbiomed.2021.05.032Get rights and content

Highlights

  • TNFα suppresses intestinal cell ketogenesis by inhibition of HMGCS2 expression.

  • Protein levels of HMGCS2 are decreased in human IBD tissues.

  • HMGCS2 attenuates TNFα-induced apoptosis, CXCL1-3 expression and ROS generation.

  • Treatment of β-hydroxybutyrate or rosiglitazone alleviates TNFα-induced apoptosis.

  • ROS represses ketogenesis in intestinal cells.

Abstract

The disturbance of strictly regulated self-regeneration in mammalian intestinal epithelium is associated with various intestinal disorders, particularly inflammatory bowel diseases (IBDs). TNFα, which plays a critical role in the pathogenesis of IBDs, has been reported to inhibit production of ketone bodies such as β-hydroxybutyrate (βHB). However, the role of ketogenesis in the TNFα-mediated pathological process is not entirely known. Here, we showed the regulation and role of HMGCS2, the rate-limiting enzyme of ketogenesis, in TNFα-induced apoptotic and inflammatory responses in intestinal epithelial cells. Treatment with TNFα dose-dependently decreased protein and mRNA expression of HMGCS2 and its product, βHB production in human colon cancer cell lines HT29 and Caco2 cells and mouse small intestinal organoids. Moreover, the repressed level of HMGCS2 protein was found in intestinal epithelium of IBD patients with Crohn's disease and ulcerative colitis as compared with normal tissues. Furthermore, knockdown of HMGCS2 enhanced and in contrast, HMGCS2 overexpression attenuated, the TNFα-induced apoptosis and expression of pro-inflammatory chemokines (CXCL1-3) in HT29, Caco2 cells and DLD1 cells, respectively. Treatment with βHB or rosiglitazone, an agonist of PPARγ, which increases ketogenesis, attenuated TNFα-induced apoptosis in the intestinal epithelial cells. Finally, HMGCS2 knockdown enhanced TNFα-induced reactive oxygen species (ROS) generation. In addition, hydrogen peroxide, the major ROS contributing to intestine injury, decreased HMGCS2 expression and βHB production in the intestinal cells and mouse organoids. Our findings demonstrate that increased ketogenesis attenuates TNFα-induced apoptosis and inflammation in intestinal cells, suggesting a protective role for ketogenesis in TNFα-induced intestinal pathologies.

Introduction

The mammalian intestinal epithelium undergoes a highly regimented and tightly controlled process of continuous self-renewal, which includes balanced proliferation, apoptosis and differentiation [1]. An imbalance of this orderly process is associated with various intestinal pathologies such as colorectal cancer and inflammatory bowel diseases (IBDs) [2,3].

Patients with IBD, a chronic inflammatory disorder characterized by periods of remission and relapse [4], suffer from a number of debilitating symptoms such as abdominal pain, diarrhea, weight loss and rectal bleeding [5]. These diseases include primarily Crohn's disease and ulcerative colitis, which differ in several features including the location and the nature of the inflammation [5,6]. Recent studies have shown that IBDs are a result of a combination of multi-factorial etiologies such as genetic, microbial, and environmental factors [4,7]. Although the exact pathogenesis for IBDs is still not entirely known, IBDs are characterized by a deregulated production of pro-inflammatory mediators including tumor necrosis factor α (TNFα) [7,8].

TNFα is associated with cellular processes such as cell proliferation, survival and death and is a key regulator of the inflammatory response in a variety of human diseases, including psoriasis, rheumatoid arthritis, and IBDs [[8], [9], [10]]. TNFα has an important role in maintaining the intestinal integrity and in the pathogenesis of intestinal inflammation [8,10]. Increased systemic and intestinal tissue levels of TNFα in IBD patients and IBD risk alleles, associated with TNF signaling components that have been identified by genome wide association studies, suggest an essential function for TNFα in the pathogenesis of IBD [[11], [12], [13]]. In addition, anti-TNFα therapy is considered as a beneficial therapeutic option for some IBD patients [8,10,11,14].

Ketogenesis represents a series of metabolic reactions that produce ketone bodies, which provide a substitute source of energy in the body during fasting and starvation [15,16]. This process primarily occurs in the mitochondria of liver cells but is also carried out in the kidney, brain and colon to a lesser extent [16,17]. Ketogenesis is regulated by transcriptional and posttranslational modifications of some key ketogenic enzymes [15,17]. Mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2) is the rate-limiting enzyme of ketogenesis, which generates ketone bodies such as β-hydroxybutyrate (βHB) [17,18]. We and others have shown that the expression of HMGCS2 is regulated by PPARs, c-Myc or the Wnt/β-catenin pathway [[19], [20], [21]]. Cytokines (e.g., TNFα) have been reported to inhibit ketone body production in animal models and isolated rat hepatocytes [[22], [23], [24]]. However, detailed mechanisms and the potential role of ketogenic enzymes, especially HMGCS2, in this process are not known.

In our current study, we investigated the regulation of HMGCS2 expression and βHB concentration by TNFα in intestinal epithelial cells and mouse intestinal organoids and the protein level of HMGCS2 in IBD patient tissues. We also determined the effect of HMGCS2 and βHB on TNFα-induced responses in the intestinal epithelial cells. Furthermore, we showed the effect of HMGCS2 on TNFα-induced reactive oxygen species (ROS) generation and the influence of ROS on ketogenesis regulation in intestinal cells. Our findings suggest a novel protective role for ketogenesis in TNFα-induced apoptosis and inflammatory responses in intestinal epithelial cells.

Section snippets

Materials and methods

Cell culture, transfection and treatment. Human colorectal cancer epithelial cell lines, HT29, Caco2 and DLD1, were obtained from American Type Culture Collection (ATCC, Manassas, VA). HT29 and Caco2 cells were cultured in McCoy's 5 A medium supplemented with 10% FBS and MEM with 15% FBS, 1% sodium pyruvate and 1% nonessential amino acids, respectively. DLD1 cells were grown in RPMI1640 with 10% FBS. The cells were maintained at 37 °C in a humidified 5% CO2 incubator. Transfection with

Results

TNFα inhibits ketogenesis through suppression of HMGCS2 expression in intestinal cells. Previous studies have shown that cytokines (e.g., TNFα) inhibit ketone body production in several animal models and isolated rat hepatocytes [[22], [23], [24]]. To determine how ketogenesis is regulated by TNFα, the expression of HMGCS2, a rate-limiting ketogenic enzyme, was determined in human colon epithelial cancer cell lines, HT29 and Caco2 cells. As shown in Fig. 1A, treatment of HT29 (left) and Caco2

Discussion

The intestinal epithelium closely interacts with the microbiota and immune system to maintain a tight equilibrium and balance between proliferation and programmed cell death. Disruption of this highly-regimented process can lead to a number of intestinal diseases [2,3]. Cytokines and chemokines play a critical role in the integrity of the intestinal epithelium and can initiate and drive intestinal damage [31,32]. Previously, the inhibitory effect of cytokines, especially TNFα, on ketone body

Declaration of competing interest

The authors have no conflicts of interest to disclose.

Acknowledgments

We thank the Markey Cancer Center Research Communications Office for editorial support and manuscript preparation. We also thank the Biostatistics and Bioinformatics Shared Resource Facility, the Biospecimen Procurement and Translational Pathology Shared Resource Facility, and the Flow Cytometry and Immune Monitoring Shared Resource Facility of the University of Kentucky Markey Cancer Center. This work was supported by the National Cancer Institute (R01 DK048498 and P30 CA177558).

References (68)

  • S. Xu et al.

    Rosiglitazone attenuates endothelial progenitor cell apoptosis induced by TNF-alpha via ERK/MAPK and NF-kappaB signal pathways

    J. Pharmacol. Sci.

    (2011)
  • J. Ye

    Regulation of PPARgamma function by TNF-alpha

    Biochem. Biophys. Res. Commun.

    (2008)
  • Y.F. Dang et al.

    New insights into molecular mechanisms of rosiglitazone in monotherapy or combination therapy against cancers

    Chem. Biol. Interact.

    (2018)
  • D. Yang et al.

    Pro-inflammatory cytokines increase reactive oxygen species through mitochondria and NADPH oxidase in cultured RPE cells

    Exp. Eye Res.

    (2007)
  • E.G. Neal et al.

    The ketogenic diet for the treatment of childhood epilepsy: a randomised controlled trial, the Lancet

    Neurology

    (2008)
  • H.H. Radeke et al.

    Interleukin 1-alpha and tumor necrosis factor-alpha induce oxygen radical production in mesangial cells

    Kidney Int.

    (1990)
  • C.N. Young et al.

    Reactive oxygen species in tumor necrosis factor-alpha-activated primary human keratinocytes: implications for psoriasis and inflammatory skin disease

    J. Invest. Dermatol.

    (2008)
  • M. Maalouf et al.

    Ketones inhibit mitochondrial production of reactive oxygen species production following glutamate excitotoxicity by increasing NADH oxidation

    Neuroscience

    (2007)
  • K.J. Maloy et al.

    Intestinal homeostasis and its breakdown in inflammatory bowel disease

    Nature

    (2011)
  • A. Negroni et al.

    Apoptosis, necrosis, and necroptosis in the gut and intestinal homeostasis

    Mediat. Inflamm.

    (2015)
  • D.S. Shouval et al.

    The role of environmental factors in the pathogenesis of inflammatory bowel diseases: a review

    JAMA Pediatr

    (2017)
  • S.S. Seyedian et al.

    A review of the diagnosis, prevention, and treatment methods of inflammatory bowel disease

    J Med Life

    (2019)
  • E. Domenech et al.

    An overview of the natural history of inflammatory bowel diseases

    Dig. Dis.

    (2014)
  • J. Pedersen et al.

    Inflammatory pathways of importance for management of inflammatory bowel disease

    World J. Gastroenterol.

    (2014)
  • B. Ruder et al.

    Tumour necrosis factor Alpha in intestinal homeostasis and gut related diseases

    Int. J. Mol. Sci.

    (2019)
  • B. Gareb et al.

    Review: local tumor necrosis factor-alpha inhibition in inflammatory bowel disease

    Pharmaceutics

    (2020)
  • J.M. Blander

    Death in the intestinal epithelium-basic biology and implications for inflammatory bowel disease

    FEBS J.

    (2016)
  • J.M. Blander

    On cell death in the intestinal epithelium and its impact on gut homeostasis

    Curr. Opin. Gastroenterol.

    (2018)
  • R. Franca et al.

    Therapeutic drug monitoring to improve outcome of anti-TNF drugs in pediatric inflammatory bowel disease

    Expet Opin. Drug Metabol. Toxicol.

    (2019)
  • M. Grabacka et al.

    Regulation of ketone body metabolism and the role of PPARalpha

    Int. J. Mol. Sci.

    (2016)
  • F.G. Hegardt

    Mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase: a control enzyme in ketogenesis

    Biochem. J.

    (1999)
  • N. Camarero et al.

    Ketogenic HMGCS2 Is a c-Myc target gene expressed in differentiated cells of human colonic epithelium and down-regulated in colon cancer

    Mol. Canc. Res.

    (2006)
  • J.T. Kim et al.

    Regulation of ketogenic enzyme HMGCS2 by wnt/beta-catenin/PPARgamma pathway in intestinal cells

    Cells

    (2019)
  • M. Beylot et al.

    Inhibition of hepatic ketogenesis by tumor necrosis factor-alpha in rats

    Am. J. Physiol.

    (1992)
  • Cited by (13)

    View all citing articles on Scopus
    View full text