Elsevier

Biochemical Pharmacology

Volume 70, Issue 3, 1 August 2005, Pages 394-406
Biochemical Pharmacology

HDAC inhibition prevents NF-κB activation by suppressing proteasome activity: Down-regulation of proteasome subunit expression stabilizes IκBα

https://doi.org/10.1016/j.bcp.2005.04.030Get rights and content

Abstract

The short chain fatty acid (SCFA) butyrate (BA) and other histone deacetylase (HDAC) inhibitors can rapidly induce cell cycle arrest and differentation of colon cancer cell lines. We found that butyrate and the specific HDAC inhibitor trichostatin A (TSA) can reprogram the NF-κB response in colon cancer cells. Specifically, TNF-α activation is suppressed in butyrate-differentiated cells, whereas IL-1β activation is largely unaffected. To gain insight into the relationship between butyrate-induced differentiation and NF-κB regulation, we determined the impact of butyrate on proteasome activity and subunit expression. Interestingly, butyrate and TSA reduced the cellular proteasome activity in colon cancer cell lines. The drop in proteasome activity results from the reduced expression of the catalytic β-type subunits of the proteasome at both the protein and mRNA level. The selective impact of HDAC inhibitors on TNF-α-induced NF-κB activation appears to relate to the fact that the TNF-α-induced activation of NF-κB is mediated by the proteasome, whereas NF-κB activation by IL-1β is largely proteasome-independent. These findings indicate that cellular differentation status and/or proliferative capacity can significantly impact proteasome activity and selectively alter NF-κB responses in colon cancer cells. This information may be useful for the further development and targeting of HDAC inhibitors as anti-neoplastic and anti-inflammatory agents.

Introduction

Butyrate (BA) is a short chain fatty acid (SCFA) produced by bacterial fermentation of dietary fiber within the gastrointestinal tract. It is actively absorbed by intestinal epithelial cells and is the major luminal source of energy for colonocytes [1], [2]. In addition to being a fuel source, butyrate has a number of other profound biological effects [3], [4], [5], [6]. For instance, butyrate can readily induce apoptosis of transformed cell lines [7], [8]. The ability for butyrate to induce apoptosis of cancer cells may contribute to the cancer preventive effects ascribed to dietary fiber [9], [10], [11]. Short chain fatty acids, particularly butyrate, can also suppress intestinal inflammation. Clinical trials have shown that topical butyrate applications alleviate symptoms in patients with mild and moderate ulcerative colitis [12]. Butyrate is also effective at treating other inflammatory conditions of the distal gastrointestinal tract [6], [13], [14]. In association, deficiencies in luminal butyrate production have been linked to colonic inflammation [9]. Such results suggest that butyrate may play an important role in regulating intestinal inflammation, as well as suppressing cellular transformation.

Biochemically, butyrate is a histone deacetylase (HDAC) inhibitor [15]. HDAC inhibitors in general have been noted for their ability to induce cell cycle arrest, differentiation and apoptosis of a wide spectrum of transformed cells. It has been proposed that HDAC inhibitors serve to normalize HDAC activity in transformed cells, which often express elevated levels of certain HDAC proteins [16], [17], [18], [19]. These findings have prompted clinical trials to assess the cancer therapeutic activity of HDAC inhibitors (such as SAHA and LAQ824) [20], [21], [22]. Gene expression profiles of cells treated with HDAC inhibitors have revealed that roughly 2% of the expressed genes are altered following HDAC inhibition [23]. Interestingly, within this population of genes, roughly half increase in expression, while the other half decreases. These selective changes in gene expression likely result from the enhanced acetylation of gene-regulatory transcription factors (e.g., GATA, p53, Sp1 and Sp3), in addition to the enhanced acetylation of histone proteins [24], [25], [26], [27].

Recently, HDAC inhibitors have been reported to modulate the activity of the transcription factor NF-κB in a number of different cell types including colon cancer cell lines and macrophages isolated from the lamia propria of the colon [3], [28], [29], [30], [31]. NF-κB is a central mediator of the immune and inflammatory response and has been implicated in promoting tumorogenesis by protecting cancer cells from apoptosis [32]. Upon activation, NF-κB rapidly enhances the expression of proinflammatory genes such as cytokines and cell adhesion molecules, as well as genes involved in promoting proliferation, angiogenesis and cell survival [33], [34], [35]. The ability of butyrate and other HDAC inhibitors to modulate NF-κB activity coincides with its proposed cancer suppressing and anti-inflammatory activities.

NF-κB is regulated through the binding of inhibitory molecules collectively referred to as the IκB proteins [36], [37]. IκB family members include IκBα, IκBɛ, IκBγ, IκBζ, Bcl-3, p105, p100, splicing variants IκBβ1 and IκBβ2 [37], [38], [39], [40], [41], [42]. Perhaps the most important and well-characterized inhibitor of the IκB family is IκBα. It is the most abundant inhibitor overall, and is responsible for the rapid activation of NF-κB [43], [44], [45], [46], [47]. During NF-κB activation, IκBα is phophorylated by the IκB kinase (IKK) complex and subsequently ubiquitinated by the multisubunit E3 ubiquitin-ligation enzyme, SCFβTrCP[48], [49]. Ubiquitinated IκBα is then rapidly degraded by the proteasome, releasing NF-κB to influence target gene expression [50], [51], [52]. It has been reported that butyrate's ability to suppress NF-κB activity depends in part on its ability to suppress cellular proteasome activity [30], [53].

Here, we provide evidence that HDAC inhibitors butyrate and trichostatin A (TSA) suppress proteasome activity by down-regulating the expression of select proteasome subunits. This ultimately prevents the ubiquitin-mediated, proteasome-dependent degradation of IκBα limiting NF-κB activation. Furthermore, we find that HDAC inhibitors selectively interfere with the proteasome-dependent activation of NF-κB, while having little effect on the proteasome-independent pathway. These findings further classify the cellular activities of a promising new class of anti-inflammatory/anti-neoplastic agents.

Section snippets

Cell culture and treatments

All cell lines were purchased from American Type Culture Collection (Manassas, VA). Caco-2 cells were propagated in minimal essential media containing 2 mM l-glutamine and Earle's salts (E-MEM) supplemented with 10% fetal bovine serum, 0.1 mM non-essential amino acids, 1 mM sodium pyruvate, streptomycin (50 mg/ml) and penicillin (50 U/ml). HT-29 and SW480 cells were propagated in McCoy's 5A medium supplemented with 10% fetal bovine serum, 0.1 mM non-essential amino acids and antibiotics. All medium

TNF-α-induced degradation of IκBα is proteasome-dependent in Caco-2 cells

Recent studies have indicated that not all activators of NF-κB induce the degradation of IκBα via the ubiquitin-proteasome system [56], [57], [58], [59]. In fact, it has been reported that the proteasome does not play a predominant role in the IL-1β-induced activation of NF-κB in colonic epithelial cells [60]. Therefore, we sought to determine the involvement of the proteasome in the induced degradation of endogenous IκBα by proinflammatory cytokines TNF-α and IL-1β in the Caco-2 colonic

Discussion

Numerous lines of evidence indicate that NF-κB promotes tumorigenesis [33], [35]. The sustained activation of NF-κB suppresses apoptosis by increasing the expression of cellular survival signals. In fact, the increased activity of NF-κB has been found in many types of solid tumors and transformed cell lines, including those isolated from the colonic epithelium [74], [75]. NF-κB also plays a pivitol role in the inflammatory response by inducing the expression of numerous inflammatory-related

Acknowledgement

This work was supported in part by an award from the National Cancer Institute to C.G. (R29CA79656)

References (99)

  • P.A. Baeuerle

    I kappa B–NF-kappaB structures: at the interface of inflammation control

    Cell

    (1998)
  • T.D. Gilmore et al.

    The I kappa B proteins: members of a multifunctional family

    Trends Genet

    (1993)
  • S. Haskill

    Characterization of an immediate-early gene induced in adherent monocytes that encodes I kappa B-like activity

    Cell

    (1991)
  • S. Yamazaki et al.

    A novel I kappa B protein, I kappa B-zeta, induced by proinflammatory stimuli, negatively regulates nuclear factor-kappa B in the nuclei

    J Biol Chem

    (2001)
  • R.F. Place

    Cytokine stabilization of newly synthesized I(kappa)B-alpha

    Biochem Biophys Res Commun

    (2001)
  • L. Catley

    NVP-LAQ824 is a potent novel histone deacetylase inhibitor with significant activity against multiple myeloma

    Blood

    (2003)
  • K.L. Rock

    Inhibitors of the proteasome block the degradation of most cell proteins and the generation of peptides presented on MHC class I molecules

    Cell

    (1994)
  • Z. Qin et al.

    A caspase-3-like protease is involved in NF-kappaB activation induced by stimulation of N-methyl-d-aspartate receptors in rat striatum

    Brain Res Mol Brain Res

    (2000)
  • F. Chen

    Calpain contributes to silica-induced I kappa B-alpha degradation and nuclear factor-kappa B activation

    Arch Biochem Biophys

    (1997)
  • Y. Han

    Tumor necrosis factor-alpha-inducible I kappa B alpha proteolysis mediated by cytosolic m-calpain. A mechanism parallel to the ubiquitin-proteasome pathway for nuclear factor-kappa B activation

    J Biol Chem

    (1999)
  • C. Wojcik et al.

    Analysis of Drosophila 26 S proteasome using RNA interference

    J Biol Chem

    (2002)
  • J.A. McBain

    Apoptotic death in adenocarcinoma cell lines induced by butyrate and other histone deacetylase inhibitors

    Biochem Pharmacol

    (1997)
  • I. Luque et al.

    Rel/NF-kappa B and I kappa B factors in oncogenesis

    Semin Cancer Biol

    (1997)
  • G. Rogler

    Nuclear factor kappaB is activated in macrophages and epithelial cells of inflamed intestinal mucosa

    Gastroenterology

    (1998)
  • P. Chen et al.

    Autocatalytic subunit processing couples active site formation in the 20S proteasome to completion of assembly

    Cell

    (1996)
  • M. Gaczynska

    Proteasome subunits X and Y alter peptidase activities in opposite ways to the interferon-gamma-induced subunits LMP2 and LMP7

    J Biol Chem

    (1996)
  • N. Chondrogianni

    Central role of the proteasome in senescence and survival of human fibroblasts: induction of a senescence-like phenotype upon its inhibition and resistance to stress upon its activation

    J Biol Chem

    (2003)
  • G. Mannhaupt

    Rpn4p acts as a transcription factor by binding to PACE, a nonamer box found upstream of 26S proteasomal and other genes in yeast

    FEBS Lett

    (1999)
  • T. Grune et al.

    Degradation of oxidized proteins in K562 human hematopoietic cells by proteasome

    J Biol Chem

    (1996)
  • D.H. Lee et al.

    Proteasome inhibitors: valuable new tools for cell biologists

    Trends Cell Biol

    (1998)
  • C.E. Denlinger

    Combined proteasome and histone deacetylase inhibition in non-small cell lung cancer

    J Thorac Cardiovasc Surg

    (2004)
  • C. Yu

    The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571

    Blood

    (2003)
  • A. Hershko

    Roles of ubiquitin-mediated proteolysis in cell cycle control

    Curr Opin Cell Biol

    (1997)
  • W.G. Bennett et al.

    Benefits of dietary fiber, Myth or medicine?

    Postgrad Med

    (1996)
  • B.S. Reddy

    Role of dietary fiber in colon cancer: an overview

    Am J Med

    (1999)
  • A. Toscani et al.

    Molecular analysis of sodium butyrate-induced growth arrest

    Oncogene Res

    (1988)
  • R.D. Fusunyan

    Butyrate switches the pattern of chemokine secretion by intestinal epithelial cells through histone acetylation

    Mol Med

    (1999)
  • A. Hague

    Sodium butyrate induces apoptosis in human colonic tumour cell lines in a p53-independent pathway: implications for the possible role of dietary fibre in the prevention of large-bowel cancer

    Int J Cancer

    (1993)
  • G.L. Thomas

    Enhanced apoptosis in transformed human lung fibroblasts after exposure to sodium butyrate

    In Vitr Cell Dev Biol Anim

    (1996)
  • O.C. Velazquez et al.

    Butyrate potential role in colon cancer prevention and treatment

    Adv Exp Med Biol

    (1997)
  • A. Csordas

    Butyrate, aspirin and colorectal cancer

    Eur J Cancer Prev

    (1996)
  • R.I. Breuer

    Rectal irrigation with short-chain fatty acids for distal ulcerative colitis. Preliminary report

    Dig Dis Sci

    (1991)
  • O. Kanauchi

    Butyrate from bacterial fermentation of germinated barley foodstuff preserves intestinal barrier function in experimental colitis in the rat model

    J Gastroenterol Hepatol

    (1999)
  • A. Wachtershauser et al.

    Rationale for the luminal provision of butyrate in intestinal diseases

    Eur J Nutr

    (2000)
  • H. Kawai

    Overexpression of histone deacetylase HDAC1 modulates breast cancer progression by negative regulation of estrogen receptor alpha

    Int J Cancer

    (2003)
  • J.H. Choi

    Expression profile of histone deacetylase 1 in gastric cancer tissues

    Jpn J Cancer Res

    (2001)
  • K. Halkidou

    Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer

    Prostate

    (2004)
  • P. Atadja

    Molecular and cellular basis for the anti-proliferative effects of the HDAC inhibitor LAQ824

    Novartis Found Symp

    (2004)
  • W.K. Kelly

    Phase I clinical trial of histone deacetylase inhibitor: suberoylanilide hydroxamic acid administered intravenously

    Clin Cancer Res

    (2003)
  • Cited by (161)

    • HDAC5-Mediated Acetylation of p100 Suppresses Its Processing

      2023, International Dental Journal
      Citation Excerpt :

      LMK-235 is a potent, selective inhibitor of HDAC4/5.14 Inhibition of HDACs was demonstrated to reduce NF-κB p65 activation in vitro.15 In this study, we explore the effect of LMK-235 on p100 deacetylation and its effect on PD and osteoclast differentiation.

    View all citing articles on Scopus
    View full text