Mitochondrial dysfunction induced by knockdown of mortalin is rescued by Parkin

https://doi.org/10.1016/j.bbrc.2011.05.116Get rights and content

Abstract

Mutations in the parkin gene are the most common cause of autosomal recessive Parkinson’s disease (PD). As an E3-ubiquitin ligase, Parkin is associated with mitochondrial dynamics and mitophagy. Mortalin, a molecular chaperone, is located primarily in mitochondria, where it functions to maintain mitochondrial homeostasis and antagonize oxidative stress injury. A reduced expression level of mortalin has been observed in the affected brain regions of PD patients. Mortalin also interacts with a variety of PD-related proteins and plays an indispensible role in helping native protein refolding and importing proteins into the mitochondrial matrix. Thus, the main aims of the present study were to investigate mitochondrial dysfunction induced by knockdown of mortalin and to test whether Parkin overexpression could rescue this effect. We found that lentivirus-mediated knockdown of mortalin in HeLa cells resulted in a collapse of mitochondrial membrane potential, an abnormal accumulation of reactive oxygen species and apparent alterations in mitochondrial morphology under H2O2-induced stress conditions. Remarkably, Parkin overexpression rescued these mitochondrial abnormalities. In HeLa cells expressing Parkin, co-immunoprecipitation of endogenous mortalin and wild-type Parkin was detected when they were treated with carbonyl cyanide 3-chlorophenylhydrazone (CCCP). In conclusion, we indicate that the relatively decreased mortalin expression level and its impaired interaction with Parkin could affect its roles in mitochondrial function.

Highlights

► Mortalin interacts with Parkin. ► Knockdown of mortalin induces a collapse of mitochondrial membrane potential, abnormal accumulation of ROS and apparent changes in mitochondrial morphology under stress conditions. ► Parkin overexpression rescues the mitochondrial dysfunction induced by knockdown of mortalin.

Introduction

Parkinson’s disease (PD) is characterized by dopaminergic neurodegeneration in the substantia nigra pars compacta (SNpc) and by the appearance of cytoplasmic inclusions, namely Lewy bodies. PD is a highly prevalent neurodegenerative disease, yet its pathogenesis remains elusive. The accumulation of abnormal proteins and mitochondrial oxidative stress may lead to impaired or apoptotic dopaminergic neurons in PD patients. Mutations in the parkin gene are the most common cause of autosomal recessive PD, and may also play a role in sporadic cases of PD. Recent work on Parkin underscores the importance of mitochondrial dysfunction as a central mechanism of PD pathogenesis [1], [2], [3]. As a mitochondrially localized E3 ubiquitin–protein ligase [3], Parkin is recruited selectively to impaired mitochondria, mediates Lys27-linked polyubiquitination of voltage-dependent anion channel 1 (VDAC1) and initiates mitophagy through the autophagy–lysosome pathway [1]. Parkin is also involved in the regulation of mitochondrial morphology [4], antagonizing oxidative damage to mtDNA and activating mitochondrial self-repair mechanisms [2].

Mortalin (also known as HSPA9, GRP75 and PBP74) is a molecular chaperone 70 family member that is located primarily in mitochondria, where it functions to maintain mitochondrial homeostasis and quality control. As a central component of the mitochondrial protein import machinery, mortalin binds to nuclear-encoded proteins and drives them into the mitochondrial matrix by an ATP-dependent mechanism with the help of other co-chaperones [5]. As an anti-apoptotic protein, mortalin protects cells by inhibiting the accumulation of reactive oxygen species (ROS) during glucose deprivation [6]. Increasing evidence linking mortalin to PD include reduced expression levels in affected brain regions, mortalin mutations in PD patients and interactions of mortalin with the PD-associated proteins [7], [8].

Therefore, this study primarily focuses on studying the effects of mortalin knockdown on mitochondrial homeostasis and the recruitment of Parkin to mitochondria, as well as testing whether Parkin overexpression can protect against mitochondrial dysfunction induced by knockdown of mortalin under H2O2-induced oxidative stress.

Section snippets

Plasmid construction

Parkin variants (T240R/R275W/R1 domain del) were fusion PCR generated and subcloned into pcDNA3.1 (−) (Invitrogen, Carlsbad, CA) or pEGFP-N2 (Clontech, Palo Alto, CA). The pLKO.1-TRC, pCMV-dR8.2 and pCMV-VSVG vectors were purchased from Sigma–Aldrich (Sigma–Aldrich, St. Louis, MO). All constructs were verified by sequencing. Primers used in this experiment were listed below (Table 1).

Cell culture and transfection

HeLa and HEK293T cells were maintained at 37 °C with 5% CO2 in Dulbecco’s modified Eagle’s medium (DMEM;

The CCCP induced interaction of endogenous mortalin with Parkin in vivo

To determine whether CCCP can induce the interaction of endogenous mortalin with Parkin in vivo, we first examined the subcellular localization of Parkin and mortalin. Because of undetectable Parkin levels in HeLa cells, the cells were transiently transfected with pEGFP (for immunofluorescence analysis) or pcDNA 3.1(−) (for subcellular fractionation analysis) vector encoding wild-type (WT) Parkin and treated with 10 μM CCCP for 1 h. The results of immunofluorescence confirmed that mortalin is

Discussion

Our data show that mortalin knockdown induces an unexpected collapse of MMP, abnormal accumulation of ROS and a significant decrease of mtDNA copy numbers under H2O2-induced oxidative stress conditions. Moreover, mortalin interacts with the PD-related protein Parkin, and Parkin overexpression notably rescues the mitochondrial dysfunction that is induced by knockdown of mortalin.

Acknowledgments

This work was supported by the Specialized Research Fund for the Doctoral Program of Higher Education of China (Grant No. 20100071110036) and the Shanghai Municipal Natural Science Foundation, China (Grant No. 10ZR1403200). The authors thank Prof. Zhuohua Zhang for the kind gift of the Parkin plasmid.

References (20)

There are more references available in the full text version of this article.

Cited by (50)

  • Proteomic characterization of early lung response to breast cancer metastasis in mice

    2019, Experimental and Molecular Pathology
    Citation Excerpt :

    Oxidative stress significantly imposed to reorganization of tissue structure as described earlier. Additionally, ROS-related activation of the different mechanisms enabled the formation of tumor-promoting changes e.g. inhibition of cell death, lead to reduced sensitivity of cancer cells to hydrogen peroxide and ROS, enabled the protection of oncoproteins, reflected by alterations in Ywhaz (Pennington et al., 2018), Selenbp1 (Zhao et al., 2016), Hsp90 (Beck et al., 2009; Garcia-Cardena et al., 1998), Hspa9 (Yang et al., 2011), G6pdx (Efferth et al., 2006). The other changes related to formation of the tumor microenvironment and alterations in pathways regulating tumorigenesis, which lead to distant metastases formation, were observed in proteins involved in defense mechanisms - heat shock proteins, as well as Selenbp1.

  • Fasudil, a Rho kinase inhibitor, promotes the autophagic degradation of A53T α-synuclein by activating the JNK 1/Bcl-2/beclin 1 pathway

    2016, Brain Research
    Citation Excerpt :

    Neurite outgrowth and elongation were scored by measuring the length of the longest neurite from the cell body to the tip. The cell counting and neurite length measurements were performed in a blinded manner by two independent examiners using ImageJ 1.44j (Wayne Rasband, NIH, Bethesda, MD)(Liu et al., 2013; Yang et al., 2011). The cells were seeded, treated with fasudil, and fixed as described above, following by the overnight incubation with rabbit anti-human LC3 primary antibody (1:50, monoclonal, #12741, CST, USA) and the incubation with Alex flour 488 (goat anti-rabbit, 1:3000, Life Technologies, USA) for 90 min.

  • Involvement of mortalin/GRP75/mthsp70 in the mitochondrial impairments induced by A53T mutant α-synuclein

    2015, Brain Research
    Citation Excerpt :

    The following studies about mechanism indicate that mortalin overexpression alleviates the Aβ-induced neurotoxicity through a mitochondria-dependent mechanism, including inhibiting the activation of mitochondrial permeability transition pore (mPTP), improving mitochondrial function, and the reducing oxidative stress (Qu et al., 2012, 2011). Furthermore, in conditions mimicking PD induced by H2O2 or 6-hydroxydopamine, mortalin overexpression seems to attenuate the mitochondrial injury and cell apoptosis (Chiasserini, 2010; Yang et al., 2011a, 2011b), similar to the results detected in glucose deprivation conditions or AD models. However, the majority of these studies were performed in the acute mitochondrial abnormalities induced by neurotoxins, which may not recapitulate the protein interactions in PD pathogenesis.

  • Neuroprotective effects of extract of Acanthopanax senticosus harms on SH-SY5Y cells overexpressing wild-type or A53T mutant α-synuclein

    2014, Phytomedicine
    Citation Excerpt :

    In addition, α-synuclein, the toxicity induced by which can be suppressed by parkin (Oluwatosin-Chigbu et al., 2003; Petrucelli et al., 2002), induces apoptotic cell death in the neuronal cells via a caspase-3 signaling cascade (Adamczyk et al., 2010). Parkin purges damaged organelles from the vital mitochondrial network (Tanaka, 2010) and rescues mitochondrial dysfunction induced by knockdown of mortalin, mutation in whose gene is the most common cause of autosomal recessive PD (Yang et al., 2011). Pre-study has reported caspase-3 is expressed in postmitotic dopaminergic neurons and caspase-3 activation could result in dopaminergic neuronal apoptosis in PD model, which may represent a common integration point and constitute an attractive target for antiapoptotic therapy in PD (Hartmann et al., 2000).

View all citing articles on Scopus
1

These authors contributed equally to this work.

View full text