Elsevier

Cell Calcium

Volume 82, September 2019, 102057
Cell Calcium

Review
Deranged hepatocyte intracellular Ca2+ homeostasis and the progression of non-alcoholic fatty liver disease to hepatocellular carcinoma

https://doi.org/10.1016/j.ceca.2019.102057Get rights and content

Highlights

  • Lipid accumulation in hepatocytes substantially alters intracellular Ca2+ homeostasis.

  • Deranged intracellular Ca2+ enhances further lipid accumulation.

  • Altered intracellular Ca2+ creates an environment which promotes mutagenesis and cell growth.

  • Reactive oxygen species, ER stress, CaMKII, CamKK2 and Nrf2 mediate progression to liver cancer.

Abstract

Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related deaths in men, and the sixth in women. Non-alcoholic fatty liver disease (NAFLD) is now one of the major risk factors for HCC. NAFLD, which involves the accumulation of excess lipid in cytoplasmic lipid droplets in hepatocytes, can progress to non-alcoholic steatosis, fibrosis, and HCC. Changes in intracellular Ca2+ constitute important signaling pathways for the regulation of lipid and carbohydrate metabolism in normal hepatocytes. Recent studies of steatotic hepatocytes have identified lipid-induced changes in intracellular Ca2+, and have provided evidence that altered Ca2+ signaling exacerbates lipid accumulation and may promote HCC. The aims of this review are to summarise current knowledge of the lipid-induced changes in hepatocyte Ca2+ homeostasis, to comment on the mechanisms involved, and discuss the pathways leading from altered Ca2+ homeostasis to enhanced lipid accumulation and the potential promotion of HCC. In steatotic hepatocytes, lipid inhibits store-operated Ca2+ entry and SERCA2b, and activates Ca2+ efflux from the endoplasmic reticulum (ER) and its transfer to mitochondria. These changes are associated with changes in Ca2+ concentrations in the ER (decreased), cytoplasmic space (increased) and mitochondria (likely increased). They lead to: inhibition of lipolysis, lipid autophagy, lipid oxidation, and lipid secretion; activation of lipogenesis; increased lipid; ER stress, generation of reactive oxygen species (ROS), activation of Ca2+/calmodulin-dependent kinases and activation of transcription factor Nrf2. These all can potentially mediate the transition of NAFLD to HCC. It is concluded that lipid-induced changes in hepatocyte Ca2+ homeostasis are important in the initiation and progression of HCC. Further research is desirable to better understand the cause and effect relationships, the time courses and mechanisms involved, and the potential of Ca2+ transporters, channels, and binding proteins as targets for pharmacological intervention.

Introduction

Hepatocellular carcinoma (HCC) involves the accumulation of mutations in hepatocytes, the principle cell type present in the liver. HCC is an important health issue in both Western and Asian countries [[1], [2], [3]]. Nonalcoholic fatty liver disease (NAFLD) is a major risk factor for the development of HCC [2,3]. Changes in the concentration of Ca2+ in the cytoplasmic space, endoplasmic reticulum (ER) and mitochondria are essential for the normal regulation of carbohydrate, lipid and protein metabolism, as well as many other pathways such as the excretion of bile acids [[4], [5], [6]]. Studies conducted over the past 10 years have pointed to significant lipid-induced alterations in Ca2+ signaling in steatotic hepatocytes (reviewed in [[7], [8], [9], [10], [11]]). This, in turn, leads to further lipid accumulation and to the creation of an environment which is thought to promote mutagenesis and the progression of HCC [7,[11], [12], [13], [14], [15], [16]] (Fig. 1). The aim of this review is to summarise the changes in intracellular Ca2+ signaling caused by lipid accumulation in hepatocytes, to describe the proposed links between altered Ca2+ signaling, lipids, and HCC, and to discuss the evidence that lipid-induced changes in intracellular Ca2+ signaling promote the development of HCC.

Mutations in many Ca2+ transporters, channels, and Ca2+ binding proteins have been identified in DNA derived from liver tumors in patients with HCC, and in animal models of HCC [[16], [17], [18]]. Moreover, the expression of a number of Ca2+ signaling and Ca2+ binding proteins is altered in HCC tissue [16,[19], [20], [21]]. While some of these mutations or changes in protein expression may involve DNA from non-hepatocyte cell types present in liver, the observations suggest that, in the transformed hepatocytes which constitute liver tumors, Ca2+ signaling is altered [16]. While these changes in Ca2+ signaling are undoubtedly important in the progression and migration of established HCC, the focus of this review is on lipids and the liver environment which is proposed to set up the initiation and promotion of HCC.

Section snippets

Hepatocellular carcinoma

The term “primary liver cancer” encompasses HCC, intrahepatic cholangiocarcinoma, fibrolamellar hepatocellular cancer and pediatric hepatoblastoma, all of which originate in the liver [22]. HCC is the second leading cause of cancer-related deaths in men, and the sixth in women, world-wide [[1], [2], [3]]. HCC is often not noticed in the early stages, and is difficult to treat in later stages [[1], [2], [3]]. Major risk factors which pre-dispose to the development of HCC are hepatitis B (HBV),

Non-alcoholic fatty liver disease as a risk factor for hepatocellular carcinoma

The term non alcoholic fatty liver disease refers to a spectrum of liver disorders which range from the accumulation of excess lipid in hepatocytes (simple steatosis) to NASH in which lipid accumulation over time leads to hepatocyte injury, inflammation, and peri-cellular fibrosis [16,[31], [32], [33]]. The progression from simple steatosis to NASH is principally mediated by toxic free fatty acids (lipotoxicity) and an increase in reactive oxygen species (ROS) (oxidative stress) [16,[31], [32],

Changes in endoplasmic reticulum and mitochondrial calcium homeostasis in steatotic hepatocytes

Results obtained in a number of laboratories have shown that lipid accumulation in steatotic hepatocytes causes substantial changes in intracellular Ca2+ signaling (summarized, with references, in Table 1). The Ca2+ channels and transporters which have so far been identified as being altered in steatotic hepatocytes, and which have been studied in some detail, are the sarco/endoplasmic reticulum (Ca2++Mg2+)ATP-ase2b (SERCA2b), the type 1 inositol trisphosphate receptor (InsP3R1), and

Changes in store-operated Ca2+ entry in steatotic hepatocytes

In addition to the effects of intracellular lipid accumulation on the ER, SERCA2b, InsP3R1 and on the transfer of Ca2+ between the ER and mitochondria, lipid accumulation in steatotic hepatocytes also inhibits SOCE (Table 1). SOCE is one of the main pathways of Ca2+ entry to hepatocytes, and plays a central role in maintaining adequate concentrations of [Ca2+]ER under conditions of hormonal Ca2+ signaling involving InsP3R-mediated ER Ca2+ efflux and Ca2+ extrusion from the cell via plasma

Lipid-induced inhibition of store-operated Ca2+ homeostasis and deranged intracellular Ca2+ homeostasis exacerbate lipid accumulation in hepatocytes

Several different experimental approaches have provided evidence that altered intracellular Ca2+ homoeostasis causes and/or exacerbates lipid accumulation in hepatocytes, as well as in other cell types. The main initiating change in Ca2+ transporters, channels, and Ca2+ concentrations together with the proposed mechanisms are summarized in Table 2. These involve decreased lipolysis, inhibition of lipid autophagy, inhibition of beta-oxidation, increased lipogenesis and decreased secretion of

Deranged intracellular Ca2+ homeostasis in steatotic hepatocytes creates an environment which could promote the development and progression of hepatocellular carcinoma

As discussed above, chronically elevated lipid in steatotic hepatocytes of NAFLD subjects is a major risk factor for the development of HCC [2,23]. Animal studies, principally with mice and rats, have shown that a high fat diet enhances the development of HCC, although the nature and extent of HCC depends on the agent employed to induce mutagenesis [[92], [93], [94], [95], [96]]. A recent study involving a mouse model of steatohepatitis has provided evidence that incorporation of cholesterol

Hepatocyte Ca2+ channels and transporters as potential therapeutic targets in preventing the development of hepatocellular carcinoma in non alcoholic fatty liver disease

The information presented above indicates that altered hepatocyte intracellular Ca2+ homeostasis leads to intracellular and intra organ environments which could promote the development of HCC. This suggests that normalization of intracellular Ca2+ in the hepatocytes of NAFLD patients may reduce the likelihood of NAFLD proceeding to NASH and HCC. Therefore, pharmaceutical interventions which could normalize hepatocyte intracellular Ca2+ homeostasis in NAFLD are of potential value. Table 4 lists

Conclusions

Lipid-induced alterations in intracellular Ca2+ homeostasis in steatotic hepatocytes play important roles in HCC pathology, including exacerbation of lipid accumulation, generation of ER stress and ROS, activation of Ca2+ calmodulin-sensitive kinases and activation of transcription factors. Temporal changes in Ca2+ transporters, channels and intracellular Ca2+ concentrations during the progression of simple steatosis to NASH and HCC, and the mechanisms involved, are yet to be fully understood.

Declaration of Competing Interest

The authors have no conflict of interest to declare.

Acknowledgements

This research was supported by grants from the Australian Research Council (DP140100259), Flinders Center for Innovation in Cancer, and the Flinders Foundation.

References (126)

  • R. Karagozian et al.

    Obesity-associated mechanisms of hepatocarcinogenesis

    Metabolism

    (2014)
  • C. Lebeaupin et al.

    Endoplasmic reticulum stress signalling and the pathogenesis of non-alcoholic fatty liver disease

    J. Hepatol.

    (2018)
  • E.S. Wires et al.

    High fat diet disrupts endoplasmic reticulum calcium homeostasis in the rat liver

    J. Hepatol.

    (2017)
  • G.J. Barritt et al.

    Ca2+ -permeable channels in the hepatocyte plasma membrane and their roles in hepatocyte physiology

    Biochimica et biophysica acta

    (2008)
  • E.C. Aromataris et al.

    Store-operated Ca2+ channels and stromal interaction molecule 1 (STIM1) are targets for the actions of bile acids on liver cells

    Biochimica et Biophysica Acta – Mol. Cell Res.

    (2008)
  • A. Auld et al.

    Store-operated Ca2+ inflow in Reuber hepatoma cells is inhibited by voltage-operated Ca2+ channel antagonists and, in contrast to freshly isolated hepatocytes, does not require a pertussis toxin-sensitive trimeric GTP-binding protein

    Biochimica et Biophysica Acta

    (2000)
  • H.M. Brereton et al.

    Novel variants of voltage-operated calcium channel alpha 1-subunit transcripts in a rat liver-derived cell line: deletion in the IVS4 voltage sensing region

    Cell Calcium

    (1997)
  • F. Striggow et al.

    Verapamil and diltiazem inhibit receptor-operated calcium channels and intracellular calcium oscillations in rat hepatocytes

    FEBS Lett.

    (1993)
  • R. Bataller et al.

    In vitro and in vivo activation of rat hepatic stellate cells results in de novo expression of L-type voltage-operated calcium channels

    Hepatology

    (2001)
  • S. Roth-Eichhorn et al.

    Transformation-dependent calcium influx by voltage-operated calcium channels in stellate cells of rat liver

    J. Hepatol.

    (1999)
  • E.S. Ali et al.

    The glucagon-like peptide-1 analogue exendin-4 reverses impaired intracellular Ca2+ signalling in steatotic hepatocytes

    Biochimica et biophysica acta

    (2016)
  • M.W. Greene et al.

    PKC{delta} is activated in a dietary model of steatohepatitis and regulates endoplasmic reticulum stress and cell death

    J. Biol. Chem.

    (2010)
  • M.C. Petersen et al.

    Roles of diacylglycerols and ceramides in hepatic insulin resistance

    Trends Pharmacol. Sci.

    (2017)
  • V.T. Samuel et al.

    Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease

    J. Biol. Chem.

    (2004)
  • T. Kawasaki et al.

    Protein kinase C-induced phosphorylation of Orai1 regulates the intracellular Ca2+ level via the store-operated Ca2+ channel

    J. Biol. Chem.

    (2010)
  • X. Zhu-Mauldin et al.

    Modification of STIM1 by O-linked N-acetylglucosamine (O-GlcNAc) attenuates store-operated calcium entry in neonatal cardiomyocytes

    J. Biol. Chem.

    (2012)
  • D. Holowka et al.

    Polyunsaturated fatty acids inhibit stimulated coupling between the ER Ca2+ sensor STIM1 and the Ca2+ channel protein Orai1 in a process that correlates with inhibition of stimulated STIM1 oligomerization

    Biochimica et biophysica acta

    (2014)
  • J.P. Kao et al.

    Practical aspects of measuring intracellular calcium signals with fluorescent indicators

    Methods Cell Biol.

    (2010)
  • R.M. Paredes et al.

    Chemical calcium indicators

    Methods

    (2008)
  • J. Baumbach et al.

    A Drosophila in vivo screen identifies store-operated calcium entry as a key regulator of adiposity

    Cell Metab.

    (2014)
  • M. Maus et al.

    Store-operated Ca2+ entry controls induction of lipolysis and the transcriptional reprogramming to lipid metabolism

    Cell Metab.

    (2017)
  • D.M.F. Cooper

    Store-operated Ca2+-entry and adenylyl cyclase

    Cell Calcium

    (2015)
  • I. Ganan-Gomez et al.

    Oncogenic functions of the transcription factor Nrf2

    Free Radic. Biol. Med.

    (2013)
  • K. Endo et al.

    Efficacy of combination therapy with transcatheter arterial chemoembolization and radiofrequency ablation for intermediate-stage hepatocellular carcinoma

    Scand. J. Gastroenterol.

    (2018)
  • P. Rawla et al.

    Update in global trends and aetiology of hepatocellular carcinoma

    Contemp. Oncol. (Pozn.)

    (2018)
  • L.D. Gaspers et al.

    Intercellular calcium waves integrate hormonal control of glucose output in the intact liver

    J. Physiol.

    (2019)
  • E.S. Ali et al.

    Metabolic disorders and cancer: hepatocyte store-operated Ca2+ channels in nonalcoholic fatty liver disease

    Adv. Exp. Med. Biol.

    (2017)
  • C.H. Wilson et al.

    Steatosis inhibits liver cell store-operated Ca2+ entry and reduces ER Ca2+ through a protein kinase C-dependent mechanism

    Biochem. J

    (2015)
  • B.A. Ersoy et al.

    Thioesterase-mediated control of cellular calcium homeostasis enables hepatic ER stress

    J. Clin. Invest.

    (2018)
  • J. Rieusset et al.

    Disruption of calcium transfer from ER to mitochondria links alterations of mitochondria-associated ER membrane integrity to hepatic insulin resistance

    Diabetologia

    (2016)
  • A.P. Arruda et al.

    Defective STIM-mediated store operated Ca2+ entry in hepatocytes leads to metabolic dysfunction in obesity

    Elife

    (2017)
  • M. Maus et al.

    Store-operated Ca2+ entry controls induction of lipolysis and the transcriptional reprogramming to lipid metabolism

    Cell Metab.

    (2017)
  • H.W. Park et al.

    Pharmacological correction of obesity-induced autophagy arrest using calcium channel blockers

    Nat. Commun.

    (2014)
  • F. Lin et al.

    The camKK2/camKIV relay is an essential regulator of hepatic cancer

    Hepatology

    (2015)
  • M. Grohmann et al.

    Obesity drives STAT-1-dependent NASH and STAT-3-dependent HCC

    Cell

    (2018)
  • J.Q. Liang et al.

    Dietary cholesterol promotes steatohepatitis related hepatocellular carcinoma through dysregulated metabolism and calcium signaling

    Nat. Commun.

    (2018)
  • J. Huang et al.

    Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma

    Nat. Genet.

    (2012)
  • Y. Zhang et al.

    Integrated analysis of mutation data from various sources identifies key genes and signaling pathways in hepatocellular carcinoma

    PLoS One

    (2014)
  • H.G. Hass et al.

    Gene-expression analysis identifies specific patterns of dysregulated molecular pathways and genetic subgroups of human hepatocellular carcinoma

    Anticancer Res.

    (2016)
  • B.D. Tang et al.

    Inhibition of Orai1-mediated Ca2+ entry enhances chemosensitivity of HepG2 hepatocarcinoma cells to 5-fluorouracil

    J. Cell. Mol. Med.

    (2017)
  • Cited by (38)

    • Expression and functions of transient receptor potential channels in liver diseases

      2023, Acta Pharmaceutica Sinica B
      Citation Excerpt :

      Longstanding high-fat diet intake triggers NAFLD progression by early non-alcoholic fat liver (NAFL) with excessive fat accumulation in hepatocytes to non-alcoholic steatohepatitis (NASH) with steatosis, necro-inflammation, hepatocyte injury and different degrees of fibrosis26. Excessive fat accumulation in hepatocytes yields impaired intracellular Ca2+ homeostasis27 and aberrant Ca2+ redistribution among organelles further enhances lipid accumulation by positive feedback, fuels the progression to NASH and raises insulin resistance28,29. As important channels for sustaining intracellular Ca2+ levels, TRP channels are involved in the pathological process.

    • Targeting ion channels in hepatic cancer

      2022, Theranostics and Precision Medicine for the Management of Hepatocellular Carcinoma, Volume 2: Diagnosis, Therapeutic Targets, and Molecular Mechanisms
    • Mitochondrial metabolism in progression of liver cancer

      2022, Theranostics and Precision Medicine for the Management of Hepatocellular Carcinoma, Volume 1: Biology and Pathophysiology
    View all citing articles on Scopus
    View full text