Regular article
Rho protein-mediated changes in the structure of the actin cytoskeleton regulate human inducible NO synthase gene expression

https://doi.org/10.1016/S0014-4827(03)00129-0Get rights and content

Abstract

Rho proteins (Rho, Rac, Cdc 42) are known to control the organization of the actin cytoskeleton as well as gene expression. Inhibition of Rho proteins by Clostridium difficile toxin B disrupted the F-actin cytoskeleton and enhanced cytokine-induced inducible nitric oxide synthase (iNOS) expression in human epithelial cells. Also specific inhibition by Y-27632 of p160ROCK, which mediates Rho effects on actin fibers, caused a disruption of the actin cytoskeleton and a superinduction of cytokine-induced iNOS expression. Accordingly, direct disruption of the actin cytoskeleton by cytochalasin D, latrunculin B, or jasplakinolide enhanced cytokine-induced iNOS expression. The transcription factor serum response factor (SRF) has been described as mediating actin cytoskeleton-dependent regulation of gene expression. Direct targets of SRF are activating protein 1 (AP1)-dependent genes. All compounds used inhibited SRF- and AP1-dependent reporter gene expression in DLD-1 cells. However, the enhancing effect of the actin cytoskeleton-disrupting compounds on human iNOS promoter activity was much less pronounced than the effect on iNOS mRNA expression. Therefore, besides transcriptional mechanisms, posttranscriptional effects seem to be involved in the regulation of iNOS expression by the above compounds. In conclusion, our data suggest that Rho protein-mediated changes of the actin cytoskeleton negatively modulate the expression of human iNOS.

Introduction

The inducible NO synthase (iNOS),1 the high-output NOS, is normally absent from resting cells [1]. After activation of cells by different inducers (bacterial lipopolysaccharides, cytokines) iNOS is expressed. NO synthesized by iNOS can have beneficial effects, such as antimicrobial, antiatherogenic, and antiapoptotic actions. Also iNOS-generated NO has been shown to be involved in skin wound healing [2] and protection of liver cells against different types of stress [3]. In contrast, inappropriate iNOS induction can have detrimental consequences, such as cellular injury in arthritis, colitis, or septic shock [1], [4].

The regulation of iNOS expression is cell- and species-specific, with a wide variety of signal transduction pathways involved [1]. It has generally been believed that the expression of iNOS is regulated mainly at the level of transcription. The human iNOS promoter contains potential binding sites for a number of transcription factors, such as STAT-1 (signal transducer and activator of transcription 1), AP1 (jun/fos transcription factor), and NF-κB (nuclear factor κB). These transcription factors are known to participate in the induction of iNOS expression by cytokines [1], [5]. However, the steady-state level of a particular mRNA depends not only on its rate of synthesis, but also on its rate of degradation. Several authors have presented data showing that regulation of mRNA stability contributes to iNOS expression [6], [7].

Small GTP-binding proteins (G-proteins) are monomeric proteins with molecular masses of 20–40 kDa. Five subfamilies are known [8]. Two of these subfamilies, Ras and Rho proteins, have been described as regulators of gene expression [8]. Accumulating data point to the activation of the Jun N-terminal kinase (JNK) and the p38 mitogen-activated protein kinase (p38 MAPK) by Rho proteins (mostly Cdc42 and Rac), whereas Ras proteins have been shown to control activation of the p42/44 MAPK cascade [8]. Rho proteins have been reported to activate the transcription factors serum response factor (SRF) and NF-κB [8]. In addition, the Rho family of small G-proteins is known to regulate diverse cellular processes involving the cytoskeleton. These include actin polymerization, F-actin bundling, myosin-based contractility, focal adhesion formation, and cytokinesis [8]. A direct Rho A target, which mediates Rho-induced assembly of focal adhesions and stress fibers, is p160ROCK [8], which can be specifically inhibited by compound Y-27632 [9]. Recent data published by Sotiropoulos et al. [10] showed regulation of SRF activity by changes in actin dynamics. Therefore, reorganization of the actin cytoskeleton by Rho proteins may also be important for Rho-dependent gene regulation. In addition, several authors have described the regulation of gene expression by compounds like cytochalasin D and latrunculin B, which directly disrupt the F-actin fibers [11], [12].

The cytokines used to induce iNOS expression in different cell models are known to activate small G-proteins of the Ras/Rho family [13]. Indirect inhibition of the activity of these proteins with statins enhanced cytokine-induced iNOS expression in human epithelial cells [14]. This statin-related enhancement of iNOS expression was blocked by addition of geranylgeranylpyrophosphate (GGPP), but not farnesylpyrophosphate [14], pointing to an involvement of the Rho family of small G-proteins, as their activity depends on posttranslational geranylation.

In the current study, we sought to investigate the molecular mechanism of the negative regulation of iNOS expression by small G-proteins of the Rho family in human epithelial cells. Our data demonstrate that the negative regulation of iNOS induction in human cells seems to be related to a Rho A-mediated reorganization of the actin cytoskeleton. This effect may involve an activation of transcription factors SRF and AP1 and transcriptional and posttranscriptional mechanisms.

Section snippets

Reagents

Trypsin, glutamine, and pyruvate solutions, agarose, tRNA, cytochalasin D, TRITC-labeled phalloidin, formaldehyde, TPA (12-O-tetradecanoyl-phorbol-13-acetate), Triton X-100 and BSA were purchased from Sigma, Deisenhofen, Germany. Isotopes were obtained from NEN/Dupont, Köln, Germany. Restriction enzymes, Taq polymerase, Klenow DNA polymerase, T7-Sequencing Kit, dNTPs, and NTPs were purchased from Amersham-Biosciences, Freiburg, Germany. T3 and T7 RNA polymerase, RNase A, RNase T1, DNase I, and

Calculations

All data are presented as means ± SEM. Differences between groups were tested for statistical significance using factorial ANOVA (StatView, SAS Institute Inc.) followed by Fisher’s PLSD test.

TcdB enhances cytokine-induced iNOS expression in human cells

Previous research in our laboratory had demonstrated that cytokine-induce iNOS expression was enhanced by statins in human and murine cells. This effect of statins had been attributed to an indirect inhibition of the activity of Rho proteins caused by reduced cellular GGPP levels [14]. Here we show in human epithelial DLD-1 cells that direct inhibition of the Rho proteins with TcdB [20] also enhanced cytokine-induced iNOS mRNA and NO production (Fig. 1A, 1B and 1D, [14]). TcdB did not induce

Conclusion

Our data demonstrate that TcdB and compound Y-27632 potentiate cytokine-induced iNOS expression via inhibition of Rho A-dependent reorganization of the actin cytoskeleton. Compounds directly disrupting the actin cytoskeleton such as cytochalasin D, latrunculin B, and jasplakinolide mimicked this effect on iNOS expression. All of these compounds seem to exert a dual effect on the iNOS gene: increased promoter activity and stabilization of the mRNA.

Acknowledgements

The expert technical assistance of I. Ihrig-Biedert and K. Masch is gratefully acknowledged. The plasmid pNOS(16)Luc was a generous gift of Dr. D.A. Geller, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA. This work was supported by Grant 8312-38 62 61 from the Innovation Foundation of the State of Rhineland-Palatinate (to H.K. and U.F.), Grant K1 1020/4-1 from the Deutsche Forschungsgemeinschaft (to H.K.), and by the Collaborative Research Center SFB 553 (Project A7 to

References (40)

  • C.J Sympson et al.

    Cytochalasin D-induced actin gene expression in murine erythroleukemia cells

    Exp. Cell Res.

    (1993)
  • M.R Bubb et al.

    Jasplakinolide, a cytotoxic natural product, induces actin polymerization and competitively inhibits the binding of phalloidin to F-actin

    J. Biol. Chem.

    (1994)
  • M.R Bubb et al.

    Effects of jasplakinolide on the kinetics of actin polymerizationan explanation for certain in vivo observations

    J. Biol. Chem.

    (2000)
  • T Henics

    Microfilament-dependent modulation of cytoplasmic protein binding to TNFalpha mRNA AU-rich instability element in human lymphoid cells

    Cell Biol. Int.

    (1999)
  • G Zambetti et al.

    Disruption of the cytoskeleton with cytochalasin D induces c-fos gene expression

    Exp. Cell Res.

    (1991)
  • D Rajotte et al.

    Contribution of both STAT and SRF/TCF to c-fos promoter activation by granulocyte–macrophage colony-stimulating factor

    Blood

    (1996)
  • T Kizaki et al.

    Negative regulation of LPS-stimulated expression of inducible nitric oxide synthase by AP-1 in macrophage cell line J774A.1

    Biochem. Biophys. Res. Commun.

    (2001)
  • K.D Kröncke et al.

    Inducible nitric oxide synthase in human diseases

    Clin. Exp. Immunol.

    (1998)
  • H Kleinert et al.

    Cytokine induction of NO synthase II in human DLD-1 cellsroles of the JAK-STAT, AP-1 and NF-κB-signaling pathways

    Br. J. Pharmacol.

    (1998)
  • Y Vodovotz et al.

    Mechanisms of suppression of macrophage nitric oxide release by transforming growth factor-β1

    J. Exp. Med.

    (1993)
  • Cited by (45)

    • ROCK induced inflammation of the microcirculation during endotoxemia mediated by nitric oxide synthase

      2011, Microvascular Research
      Citation Excerpt :

      The response to Rho and ROCK inhibition is conflicting, with evidence for both increased and decreased iNOS expression. One study using hearts from diabetic rats, indicated that activation of the Rho/ROCK pathway is involved in iNOS up-regulation (Soliman et al. 2008) and this has been supported in a number of models (Ikeda et al., 2001; Machida et al., 2008; Tiftik et al., 2008; Witteck et al., 2003) whereas decreased iNOS expression has been observed in preadipocytes (Dobashi et al., 2008). This conflicting evidence may be due to the different in vitro and in vivo models and may explain the lack of response with the higher dose of fasudil (10 mg kg− 1) in our model.

    • Regulation of the expression of inducible nitric oxide synthase

      2010, Nitric Oxide - Biology and Chemistry
    • Transcriptional and post-transcriptional regulation of iNOS expression in human chondrocytes

      2010, Biochemical Pharmacology
      Citation Excerpt :

      This implies that iNOS induction in chondrocytes needs some “superconfluency-derived” stress signals to enable the cytokines to activate iNOS expression. As iNOS expression is regulated by Rho-mediated changes in the structure of the actin cytoskeleton [34], it is tempting to speculate that the above-mentioned stress signals may relate to superconfluency-induced changes in the structure of the actin cytoskeleton. In addition, there is evidence that the Rho-mediated modulation of the actin cytoskeleton is involved in the expressional changes essential for chondrogenesis [35].

    View all citing articles on Scopus

    This article contains data from the theses of A.W. and Y.Y.

    View full text