Calcineurin inhibitors recruit protein kinases JAK2 and JNK, TLR signaling and the UPR to activate NF-κB-mediated inflammatory responses in kidney tubular cells

https://doi.org/10.1016/j.taap.2013.08.011Get rights and content

Highlights

  • Molecular mechanisms modulating CNI renal inflammation were investigated.

  • Kinases, immune receptors and ER stress mediate the inflammatory response to CNIs.

  • Several intracellular pathways activate NF-κB in CNIs-treated tubular cells.

  • A NF-κB-dependent cytokine profile characterizes CNIs-induced inflammation.

  • CNI nephrotoxicity was associated to inflammatory events in mice and human.

Abstract

The calcineurin inhibitors (CNIs) cyclosporine (CsA) and tacrolimus are key drugs in current immunosuppressive regimes for solid organ transplantation. However, they are nephrotoxic and promote death and profibrotic responses in tubular cells. Moreover, renal inflammation is observed in CNI nephrotoxicity but the mechanisms are poorly understood. We have now studied molecular pathways leading to inflammation elicited by the CNIs in cultured and kidney tubular cells.

Both CsA and tacrolimus elicited a proinflammatory response in tubular cells as evidenced by a transcriptomics approach. Transcriptomics also suggested several potential pathways leading to expression of proinflammatory genes. Validation and functional studies disclosed that in tubular cells, CNIs activated protein kinases such as the JAK2/STAT3 and TAK1/JNK/AP-1 pathways, TLR4/Myd88/IRAK signaling and the Unfolded Protein Response (UPR) to promote NF-κB activation and proinflammatory gene expression. CNIs also activated an Nrf2/HO-1-dependent compensatory response and the Nrf2 activator sulforaphane inhibited JAK2 and JNK activation and inflammation. A murine model of CsA nephrotoxicity corroborated activation of the proinflammatory pathways identified in cell cultures. Human CNIs nephrotoxicity was also associated with NF-κB, STAT3 and IRE1α activation.

In conclusion, CNIs recruit several intracellular pathways leading to previously non-described proinflammatory actions in renal tubular cells. Identification of these pathways provides novel clues for therapeutic intervention to limit CNIs nephrotoxicity.

Introduction

Calcineurin inhibitors (CNIs) are key drugs in current immunosuppressive regimes for solid organ transplantation. However, both cyclosporine A (CsA) and tacrolimus (formerly known as FK506) are toxic drugs that may cause acute and chronic nephrotoxicity (Naesens et al., 2009). The three key biological processes underlying acute and chronic CNIs nephrotoxicity are kidney cell death, inflammation and residual fibrosis. In acute kidney injury, fibrosis is only evident in later stages when kidney regeneration fails to fully restore kidney structure and function. A body of evidence has shown that CsA and tacrolimus promote tubular cell death and pro-fibrotic changes in tubular cells, including epithelial-to-mesenchymal transition (EMT) (Berzal et al., 2012, Justo et al., 2003, Neria et al., 2009). However, there is very little information on the effects of CNIs on kidney inflammation, despite the fact that inflammation plays a key role in nephrotoxic kidney injury induced by some drugs and has been observed in CNIs nephrotoxicity. Specifically, the cisplatin-elicited inflammatory response in tubular cells plays a key amplification role in cisplatin nephrotoxicity (Pabla and Dong, 2008).

Inflammation is an essential event in the progression of renal disease. The transcription factor NF-κB is a key promoter of renal inflammation that integrates intracellular signals from many stimuli and drives the expression of cytokines and chemokines (Sanz et al., 2008). NF-κB contributes to lymphocyte activation following recruitment by the calcineurin/NFAT signaling pathway. CsA and tacrolimus inhibit calcineurin in lymphocytes, thus hindering the formation of cooperative NFAT, NF-κB and AP-1 complexes and hence, NF-κB-dependent IL-2 and IL-8 production and clonal T cell expansion (Granelli-Piperno et al., 1990, Nishiyama et al., 2005). This antiinflammatory effect of CNIs in lymphocytes contrasts to tubulointerstitial inflammation observed in animals chronically treated with CsA (Ling et al., 2003, Mizui et al., 2004) or in those with deletion of the α isoform of calcineurin A (Gooch et al., 2007). However, the molecular mechanisms of kidney inflammation in response to CNIs have not been studied and it is unknown whether CNIs have direct pro-inflammatory actions on renal cells. The response of tubular cells and leukocytes to CNIs may differ. Thus, CsA promotes cell death in tubular cells but protects from cell death in macrophages (Hortelano et al., 1999, Hortelano et al., 2000).

Numerous molecules and signaling pathways contribute to CNIs toxicity. Thus, CNIs elicit mitochondrial injury, endoplasmic reticulum (ER) stress and activate protein kinases (including MAPK and JAK/STAT) and caspase cascades to produce apoptosis (Justo et al., 2003, Neria et al., 2009, Xiao et al., 2013). However, whereas some of these pathways are also able to recruit NF-κB and elicit inflammation in other cellular systems or in tubular cells exposed to other stressors, their role in the generation of inflammation in CNIs nephrotoxicity has not been addressed.

The Unfolded Protein Response (UPR) is an ancient physiological adaptive mechanism to cope with potential deleterious protein misfolding in the ER. CsA- and tacrolimus-induced synthesis of the UPR protein GADD153/CHOP restrained TNFα-elicited inflammation in tubular cells (Du et al., 2009). This antiinflammatory action of CNIs was proposed to potentially protect renal tissue after transplantation. However, the fact remains that CNIs contribute to chronic allograft nephropathy, a process characterized by tubulointerstitial inflammation. Innate immunity responses involving toll like receptors (TLR) may also lead to NF-κB activation during infection as well as during sterile inflammation (Gonçalves et al., 2011, Loiarro et al., 2010). In this regard, increased TLR2 and TLR4 levels have also been observed in chronic human CsA nephrotoxicity (Lim et al., 2005, Lim et al., 2009). In resting macrophages, calcineurin may restrain TLR basal activity since CNIs favored TLR downstream signaling (Kang et al., 2007, Loiarro et al., 2010), but the interaction of CNIs with TLRs in kidney cells has not been explored.

Renal tubular cells compose most of the mass of the functioning kidney and they are thought to be a central cell type in renal inflammation and CNIs nephrotoxicity (Daha and van Kooten, 2000). We have now explored the hypothesis that CNIs trigger early proinflammatory signaling directly on tubular cells. We show that CNIs engage several protein kinase-dependent pathways that converge at and result in NF-κB activation and hence in inflammation. Both the UPR and TLR natural immunity also contributed to NF-κB activation.

Section snippets

Cells and reagents

MCT cells are a cultured line of murine proximal tubular epithelial cells originally obtained from Eric Neilson (Vanderbildt University, Nashville, TN) that have been extensively characterized and used as model to study renal inflammation (Haverty et al., 1988, Sanz et al., 2008, Ucero et al., 2013). MCT proximal tubular epithelial cells were cultured in RPMI 1640 (GIBCO, Grand Island, NY) supplemented with 10% decomplemented fetal bovine serum (DFBS), 2 mM glutamine, 100 U/mL penicillin and 10 

Transcriptomic analysis of inflammatory master regulatory genes and NF-κB target genes in murine tubular cells treated with CNIs

To evaluate the potential of CNIs to elicit inflammatory responses in tubular cells and to search for relevant molecules that may contribute to CNIs-dependent inflammation, the transcriptome of murine MCT proximal tubular cells exposed to 10 μg/ml CsA or 20 μg/ml tacrolimus for 6 h was analyzed. Several genes closely related to the NF-κB pathway were found differentially expressed in cells treated with both CNIs for 6 h (Table 1). This response encompassed the increased expression of the NF-κB

Discussion

The main finding of this work is that CNIs directly trigger early NF-κB-dependent inflammatory events in tubular cells by engaging several previously unrecognized intracellular pathways, thus providing several avenues for both therapeutically targeting nephrotoxicity and preventing CKD progression. Inflammation is an early feature of CNIs nephrotoxicity that predates the characteristic kidney fibrosis.

Despite inflammation being recognized as one of the hallmarks of chronic CNIs nephrotoxicity,

Conflict of interest statement

The authors declare no conflicts of interest.

Acknowledgments

Grant support: ISCIII and FEDER funds FIS PS09/00447, ISCIII-RETIC REDinREN RD12/0021, Comunidad de Madrid/CIFRA/S2010/BMD-2378, PI08/1043, PI11/02242. Salary support: Fundación Conchita Rábago de Jiménez Díaz to CGG, COS, SB; RedinRen to SC; UAM to JE; FIS to BFF (Programa Rio Hortega), AO (Programa Intensificación Actividad Investigadora, ISCIII/Agencia Laín-Entralgo/CM) and AMR (Programa Miguel Servet). IIS-Fundacion Jimenez Diaz biobank (RD09/0076/00101) for samples.

Author contributions

References (58)

  • J.K. Roche et al.

    CXCL1/KC and CXCL2/MIP-2 are critical effectors and potential targets for therapy of Escherichia coli O157:H7-associated renal inflammation

    Am. J. Pathol.

    (2007)
  • H. Sakurai

    Targeting of TAK1 in inflammatory disorders and cancer

    Trends Pharmacol. Sci.

    (2012)
  • E. Sarró et al.

    A pharmacologically-based array to identify targets of cyclosporine A-induced toxicity in cultured renal proximal tubule cells

    Toxicol. Appl. Pharmacol.

    (2012)
  • D.H. Shin et al.

    The NRF2-heme oxygenase-1 system modulates cyclosporin A-induced epithelial–mesenchymal transition and renal fibrosis

    Free Radic. Biol. Med.

    (2010)
  • K. Volkmann et al.

    Potent and selective inhibitors of the inositol-requiring enzyme 1 endoribonuclease

    J. Biol. Chem.

    (2011)
  • J. Xu et al.

    Procarcinogenic effects of cyclosporine A are mediated through the activation of TAK1/TAB1 signaling pathway

    Biochem. Biophys. Res. Commun.

    (2011)
  • Z.H. Zong et al.

    Implication of Nrf2 and ATF4 in differential induction of CHOP by proteasome inhibition in thyroid cancer cells

    Biochim. Biophys. Acta

    (2012)
  • S. Berzal et al.

    GSK3, snail, and adhesion molecule regulation by cyclosporine A in renal tubular cells

    Toxicol. Sci.

    (2012)
  • N. Bouvier et al.

    Cyclosporine triggers endoplasmic reticulum stress in endothelial cells: a role for endothelial phenotypic changes and death

    Am. J. Physiol. Renal Physiol.

    (2009)
  • P. Carmona-Saez et al.

    GENECODIS: a web-based tool for finding significant concurrent annotations in gene lists

    Genome Biol.

    (2007)
  • M.M. Chaturvedi et al.

    NF-κB addiction and its role in cancer: ‘one size does not fit all’

    Oncogene

    (2011)
  • C.H. Cheng et al.

    Cyclosporine-induced tubular vacuolization: the role of Bip/Grp78

    Nephron Exp. Nephrol.

    (2013)
  • F.Y. Chow et al.

    Monocyte chemoattractant protein-1-induced tissue inflammation is critical for the development of renal injury but not type 2 diabetes in obese db/db mice

    Diabetologia

    (2007)
  • B.C. Cross et al.

    The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1-binding small molecule

    Proc. Natl. Acad. Sci. U. S. A.

    (2012)
  • M.R. Daha et al.

    Is the proximal tubular cell a proinflammatory cell?

    Nephrol. Dial. Transplant.

    (2000)
  • M.H. de Borst et al.

    c-Jun NH2-terminal kinase is crucially involved in renal tubulo-interstitial inflammation

    J. Pharmacol. Exp. Ther.

    (2009)
  • S. Du et al.

    Suppression of NF-kappaB by cyclosporin a and tacrolimus (FK506) via induction of the C/EBP family: implication for unfolded protein response

    J. Immunol.

    (2009)
  • S. Fougeray et al.

    Metabolic stress promotes renal tubular inflammation by triggering the unfolded protein response

    Cell Death Differ.

    (2011)
  • S. Gonçalves et al.

    Tyrphostins as potential therapeutic agents for acute kidney injury

    Curr. Med. Chem.

    (2010)
  • Cited by (37)

    • Overcoming Compensatory Mechanisms toward Chronic Drug Administration to Ensure Long-Term, Sustainable Beneficial Effects

      2020, Molecular Therapy Methods and Clinical Development
      Citation Excerpt :

      The resulting renal damage is due to tubular cell death, pro-fibrotic effects, and increased renal inflammation. Several pro-inflammatory pathways are activated by these drugs, including JAK2/STAT3 and TAK1/JNK/AP-1 pathways, TLR4/Myd88/IRAK signaling, and the unfolded protein response, which can promote nuclear factor κB (NF-κB) activation and pro-inflammatory gene expression.35 Antipsychotic drugs can cause type 2 diabetes by inhibiting insulin signaling pathways in muscle cells, hepatocytes, and adipocytes and by increasing free fatty acid levels and inflammation, thus inducing direct damage and apoptosis of β cells.36

    • Suppression of TRPM2 reduces renal fibrosis and inflammation through blocking TGF-β1-regulated JNK activation

      2019, Biomedicine and Pharmacotherapy
      Citation Excerpt :

      In our study, we also found that TRPM2 knockout significantly improved nephrin expression in renal tissues of UUO-challenged mice, possibly helping for the suppression of inflammatory response regulated by NF-κB. JNK MAPK is intricately linked to the pathobiology of various renal diseases, including kidney fibrosis [25,49]. Accordingly, there is a considerable cross-talk between TGF-β1 and JNK MAPK signaling pathway in the synthesis and turnover of the ECM by fibroblast-like cells in the kidney.

    • Modulation of eNOS/iNOS by nebivolol protects against cyclosporine A-mediated nephrotoxicity through targeting inflammatory and apoptotic pathways

      2019, Environmental Toxicology and Pharmacology
      Citation Excerpt :

      In addition, the results showed that CsA induced renal NF-κB expression, which has been traditionally considered a prototypical pro-inflammatory signaling pathway that can induce the expression of pro-inflammatory genes including cytokines, chemokines, and adhesion molecules. The current results are in line with those of other studies (González-Guerrero et al., 2013; Park et al., 2016). Furthermore, in the current study, CsA had an apoptotic effect on kidney tissue, as indicated by up-regulation of the pro-apoptotic marker, fas ligand.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text