Elsevier

Clinica Chimica Acta

Volume 506, July 2020, Pages 72-83
Clinica Chimica Acta

Review
Bnip3 in mitophagy: Novel insights and potential therapeutic target for diseases of secondary mitochondrial dysfunction

https://doi.org/10.1016/j.cca.2020.02.024Get rights and content

Highlights

Abstract

The present review is a summary of the recent literature concerning Bnip3 expression, function, and regulation, along with its implications in mitochondrial dysfunction, disorders of mitophagy homeostasis, and development of diseases of secondary mitochondrial dysfunction. As a member of the Bcl-2 family of cell death-regulating factors, Bnip3 mediates mPTP opening, mitochondrial potential, oxidative stress, calcium overload, mitochondrial respiratory collapse, and ATP shortage of mitochondria from multiple cells. Recent studies have discovered that Bnip3 regulates mitochondrial dysfunction, mitochondrial fragmentation, mitophagy, cell apoptosis, and the development of lipid disorder diseases via numerous cellular signaling pathways. In addition, Bnip3 promotes the development of cardiac hypertrophy by mediating inflammatory response or the related signaling pathways of cardiomyocytes and is also responsible for raising abnormal mitophagy and apoptosis progression through multiple molecular signaling pathways, inducing the pathogenesis and progress of hepatocellular carcinoma (HCC). Different molecules regulate Bnip3 expression at both the transcriptional and post-transcriptional level, leading to mitochondrial dysfunction and unbalance of mitophagy in hepatocytes, which promotes the development of non-alcoholic fatty liver disease (NAFLD). Thus, Bnip3 plays an important role in mitochondrial dysfunction and mitophagy homeostasis and has emerged as a promising therapeutic target for diseases of secondary mitochondrial dysfunction.

Introduction

Bnip3, encoded by the BNIP3 gene at human chromosome 10q26.3, was first known as “NIP3” and considered to belong to the Bcl-2 family of cell death-regulating factors, which is an atypical BH3 domain only containing members of the Bcl2 family of proteins [1], [2]. Bnip3 is extensively expressed in various cells and is involved in multiple cellular functions via participation in numerous cellular signaling pathways, including mitochondrial dysfunction, mitochondrial autophagy (mitophagy), and cell apoptosis [3], [4], [5], [6]. Furthermore, as a transcriptional target in response to specific dysfunctional mitochondria, Bnip3 primarily regulates mitochondrial fragmentation and mitophagy via interaction with microtuble-associated protein light chain 3 (LC-3) and its related molecular receptors [7], [8].

Growing evidence has supported Bnip3′s association with the process of dysfunctional mitochondria and mitophagy [9] and attests to a strong relationship between Bnip3 and mitochondrial morphology. More specifically, overexpression of the BNIP3 gene has been found to result in the loss of mitochondrial membrane potential (ΔΨm) and mitochondrial permeability transition pore (mPTP) opening in cardiomyocytes, which, in turn, causes aberrant mitochondrial function and cardiac cell death [10]. Conversely, the bolstering of the homeostasis of the mitophagy process can be prompted through the inhibition of BNIP3 gene expression in cardiac myocytes, which decreases mPTP opening, rescues ΔΨm level, and suppresses mitochondrial perturbation [10]. A similar relationship can be seen in the way Bnip3 precipitates cell apoptosis and death in cardiomyocytes and hepatocytes, and how, Bnip3 deficiency, in contrast, confers a decidedly reduced risk of hepatic and cardiovascular disease [10], [11], [12], [13]. As can be seen, the overall findings from Bnip3 studies lend credibility to the hypothesis that Bnip3 figures significantly in the disorders of mitophagy homeostasis and diseases of secondary mitochondrial dysfunction.

The disorders of mitophagy homeostasis imply that abnormal fission and/or fusion of mitochondria, along with mitochondrial dysfunction, is involved in cell apoptosis and death [9], [14]. PTEN-induced kinase-1 (PINK1) and Parkin were previously considered to be the general signaling pathways for the mitophagy process [15]. As a serine-threonine kinase, PINK1 is imported into mitochondria, cleaved by the inner membrane protease presenilin-associated rhomboid-like (PARL), released into the cytosol, and degraded by the proteasome [15]. If less PINK1 is imported into mitochondria as a result of mitochondrial depolarization, it stabilizes on the outer mitochondrial membrane (OMM) and then recruits Parkin to mitochondria. This eventually leads to the clearance of depolarized mitochondria and initiates the mitophagy process [16]. Bnip3 has been implicated in mitophagy in different pathophysiological conditions, wherein Bnip3 manipulates many receptors for the incorporation of mitochondria into autophagosomes [3], [12], [17]. Therefore, the investigation of the regulatory mechanisms involved in the maintenance of Bnip3-mediated mitophagy homeostasis is of great significance for the prevention and therapy of diseases of secondary mitochondrial dysfunction.

Recently, Bnip3 has gained much attention due to its potential role in mitophagy and diseases of secondary mitochondrial dysfunction [18], [19]. Despite the emerging knowledge concerning Bnip3′s involvement in dysfunctional mitochondria, the regulatory mechanisms of Bnip3 in mitophagy are still poorly understood. We therefore aimed to review the recent investigations into the molecular structure and cell biology of Bnip3, discuss the roles of Bnip3 in mitochondrial dysfunction and disorders of mitophagy homeostasis, and further provide a framework to explain how Bnip3 plays a pathogenic role in diseases of secondary mitochondrial dysfunction, including in cardiac hypertrophy, hepatocellular carcinoma (HCC), and non-alcoholic fatty liver disease (NAFLD). Further research into the regulatory mechanisms of Bnip3 actions in mitochondrial dysfunction and mitophagy could lead to the development of novel therapeutic platforms for combatting these diseases of secondary mitochondrial dysfunction.

Section snippets

Description of the structure, function, and expression of Bnip3

Originally, B-cell leukemia/lymphoma 2 (BCL2) family was identified by chromosomal translocations that activated BCL2 gene expression, although this family was later identified through their composition of a series of shared BCL2 homology (BH) motifs (BH1, BH2, BH3, and BH4) [20]. The general structure of members in this family consists of a hydrophobic α-helix surrounded by amphipathic α-helices. Importantly, BH3, functioning as a protein-protein interaction mediator, is the only motif that is

The destructive role of Bnip3 on mitochondrial autophagy

It is well established that the abnormal development of mitochondrial autophagy (mitophagy) is the key contributor to numerous diseases, and multiple studies of abnormal mitophagy have resulted in the further identification of Bnip3 expression and/or activity in mitochondrial dysfunction [5], [36], [37]. The bulk of relevant research has demonstrated that Bnip3 is indeed a significant contributor and is strongly associated with mitophagy [18]. After autophagic stimulation, Bnip3 causes

Bnip3 is involved in mitophagy-induced cardiac hypertrophy

Growing evidence demonstrates that cardiac hypertrophy, especially irreversible ventricular remodeling, is the primary culprit for the development of heart failure [11], [83]. The heart is a vital organ that returns or pumps blood through the blood vessels of the circulatory system. When an increase in the thickness of the cardiomyocytes without a corresponding increase in ventricular size occurs with the heart under ischemic conditions, the cardiomyocytes will proliferate irrepressibly, and

Bnip3 promotes the development of hepatocellular carcinoma (HCC) by provoking mitophagy disorder

A strong association between Bnip3 and hepatocellular carcinoma has been widely acknowledged (HCC) [13], [97]. A massive epigenetic regulation investigation confirmed that the BNIP3 gene is closely linked with HCC induced by mitophagy [13], [97]. Multiple in vitro and in vivo experiments have further demonstrated that Bnip3 is intimately related to the regulation of transcription activity by methylation of promoter regions, which causes HCC-related gene silencing or abnormalities in the

Bnip3 is involved in the process of non-alcoholic fatty liver disease induced by mitophagy

Recently, incidence rates for the elderly population of all major racial and ethnic populations have increased substantially, while growth rates of obesity and metabolic syndrome suggest that the prevalence of NAFLD will continue to rise [101]. NAFLD is a metabolic syndrome characterized by the disorder of hepatic lipid homeostasis and intrahepatic accumulation of a large amount of lipid [111]. Chronic liver disease, along with its progression to NAFLD, has become a major socio-economic burden.

Conclusions and perspectives

This review summarizes the crucial roles and underlying mechanisms of Bnip3 in mitophagy and diseases of secondary mitochondrial dysfunction, including cardiac hypertrophy, HCC, and NAFLD. Bnip3 manipulates the morphology, function, and homeostasis of fission and fusion on mitochondria through various signaling pathways [11], [12], [80], [96], [122]. It also regulates mitochondrial dysfunction, mitochondrial fragmentation, mitophagy, cell apoptosis, and the development of lipid disorder

Declaration of Competing Interest

The authors declare that they have no conflicts of interests. The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Acknowledgments

This work is funded by the National Natural Science Foundation of China (NO. 81503074).

References (125)

  • M.N. Quinsay et al.

    Bnip3 mediates permeabilization of mitochondria and release of cytochrome c via a novel mechanism

    J. Mol. Cell. Cardiol.

    (2010)
  • H. Lee et al.

    Regulation of BNIP3 in normal and cancer cells

    Mol. Cells

    (2006)
  • Y.H. Yook et al.

    Nitric oxide induces BNIP3 expression that causes cell death in macrophages

    Biochem. Biophys. Res. Commun.

    (2004)
  • H. Zhang et al.

    Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia

    J. Biol. Chem.

    (2008)
  • K. Okamoto et al.

    Mitochondria-anchored receptor Atg32 mediates degradation of mitochondria via selective autophagy

    Dev. Cell

    (2009)
  • D. Arnoult et al.

    Bax/Bak-dependent release of DDP/TIMM8a promotes Drp1-mediated mitochondrial fission and mitoptosis during programmed cell death

    Curr. Biol.

    (2005)
  • G. Twig et al.

    Mitochondrial fusion, fission and autophagy as a quality control axis: The bioenergetic view

    BBA-Bioenergetics

    (2008)
  • Y. Zhu et al.

    Lactate accelerates calcification in VSMCs through suppression of BNIP3-mediated mitophagy

    Cell. Signal.

    (2019)
  • R. Li et al.

    Therapeutic effect of Sirtuin 3 on ameliorating nonalcoholic fatty liver disease: The role of the ERK-CREB pathway and Bnip3-mediated mitophagy

    Redox Biol.

    (2018)
  • S. Ghavami et al.

    Role of BNIP3 in TNF-induced cell death–TNF upregulates BNIP3 expression

    BBA

    (2009)
  • A.M.J. Sanchez et al.

    AMP-activated protein kinase stabilizes FOXO3 in primary myotubes

    Biochem. Biophys. Res. Commun.

    (2018)
  • Q. Zhang et al.

    30 Years of NF-kappaB: A blossoming of relevance to human pathobiology

    Cell

    (2017)
  • G.J. Zhao et al.

    NF-kappaB suppresses the expression of ATP-binding cassette transporter A1/G1 by regulating SREBP-2 and miR-33a in mice

    Int. J. Cardiol.

    (2014)
  • Y. Chen et al.

    A nucleus-targeted alternately spliced nix/Bnip3L protein isoform modifies nuclear factor κB (NFκB)-mediated cardiac transcription

    J. Biol. Chem.

    (2013)
  • S. Almasi et al.

    TRPM2 channel-mediated regulation of autophagy maintains mitochondrial function and promotes gastric cancer cell survival via the JNK-signaling pathway

    J. Biol. Chem.

    (2018)
  • C. Shi et al.

    Yap promotes hepatocellular carcinoma metastasis and mobilization via governing cofilin/F-actin/lamellipodium axis by regulation of JNK/Bnip3/SERCA/CaMKII pathways

    Redox Biol.

    (2018)
  • L.H.M. Bozi et al.

    Endoplasmic reticulum stress impairs cardiomyocyte contractility through JNK-dependent upregulation of BNIP3

    Int. J. Cardiol.

    (2018)
  • Y.P. Rong et al.

    Targeting Bcl-2-IP3 receptor interaction to reverse Bcl-2's inhibition of apoptotic calcium signals

    Mol. Cell.

    (2008)
  • J.C. Nault et al.

    The role of molecular enrichment on future therapies in hepatocellular carcinoma

    J. Hepatol.

    (2018)
  • D.A. Kubli et al.

    Bnip3 functions as a mitochondrial sensor of oxidative stress during myocardial ischemia and reperfusion

    Am. J. Physiol. Heart Circulatory Physiol.

    (2008)
  • P.A. Fordjour et al.

    Targeting BNIP3 in inflammation-mediated heart failure: a novel concept in heart failure therapy

    Heart Fail. Rev.

    (2016)
  • C. Yuan et al.

    BNIP3/Bcl-2-mediated apoptosis induced by cyclic tensile stretch in human cartilage endplate-derived stem cells

    Exp. Ther. Med.

    (2018)
  • Kubasiak LA, Hernandez OM, Bishopric NH, et al. Hypoxia and acidosis activate cardiac myocyte death through the Bcl-2...
  • C.W. Park et al.

    BNIP3 is degraded by ULK1-dependent autophagy via MTORC1 and AMPK

    Autophagy

    (2013)
  • J. Zhang et al.

    Role of BNIP3 and NIX in cell death, autophagy, and mitophagy

    Cell Death Differ.

    (2009)
  • A.H. Chaanine et al.

    Potential role of BNIP3 in cardiac remodeling, myocardial stiffness, and endoplasmic reticulum: mitochondrial calcium homeostasis in diastolic and systolic heart failure

    Circ. Heart Failure

    (2013)
  • D.F. Calvisi et al.

    Mechanistic and prognostic significance of aberrant methylation in the molecular pathogenesis of human hepatocellular carcinoma

    J. Clin. Investig.

    (2007)
  • M. Hirano et al.

    Emerging therapies for mitochondrial diseases

    Essays Biochem.

    (2018)
  • J. Park et al.

    Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin

    Nature

    (2006)
  • D.P. Narendra et al.

    PINK1 is selectively stabilized on impaired mitochondria to activate Parkin

    PLoS Biol

    (2010)
  • L. Sun et al.

    Upregulation of BNIP3 mediated by ERK/HIF-1alpha pathway induces autophagy and contributes to anoikis resistance of hepatocellular carcinoma cells

    Future Oncol.

    (2014)
  • S. Roperto et al.

    Mitophagy mediated by BNIP3 and BNIP3L/NIX in urothelial cells of the urinary bladder of cattle harbouring bovine papillomavirus infection

    Vet. Microbiol.

    (2019)
  • G. Chen et al.

    The E1B 19K/Bcl-2-binding protein Nip3 is a dimeric mitochondrial protein that activates apoptosis

    J. Exp. Med.

    (1997)
  • K.L. O'Neill et al.

    Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane

    Genes Dev.

    (2016)
  • M. Panel et al.

    Mitochondria and aging: A role for the mitochondrial transition pore?

    Aging Cell

    (2018)
  • C. Vande Velde et al.

    BNIP3 and genetic control of necrosis-like cell death through the mitochondrial permeability transition pore

    Mol. Cell. Biol.

    (2000)
  • G. Karpathiou et al.

    Autophagy and Bcl-2/BNIP3 death regulatory pathway in non-small cell lung carcinomas

    APMIS

    (2013)
  • F. Liu et al.

    Verticillin A overcomes apoptosis resistance in human colon carcinoma through DNA methylation-dependent upregulation of BNIP3

    Cancer Res.

    (2011)
  • M. Vara-Perez et al.

    BNIP3 contributes to the glutamine-driven aggressive behavior of melanoma cells

    Biol. Chem.

    (2019)
  • H.W. Chiu et al.

    Combination of the novel histone deacetylase inhibitor YCW1 and radiation induces autophagic cell death through the downregulation of BNIP3 in triple-negative breast cancer cells in vitro and in an orthotopic mouse model

    Mol. Cancer

    (2016)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    View full text