Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

ARTICLE IN PRESS

Experimental Gerontology xxx (2007) xxxxxx


www.elsevier.com/locate/expgero

Mini Review

Three faces of mortalin: A housekeeper, guardian and killer


Sunil C. Kaul a, Custer C. Deocaris
a

a,b

, Renu Wadhwa

a,b,*

National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki 305 8562, Japan
b
Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Hongo, Tokyo 113-8656, Japan
Received 1 August 2006; received in revised form 5 October 2006; accepted 24 October 2006

Abstract
Mortalin was rst cloned as a mortality factor that existed in the cytoplasmic fractions of normal, but not in immortal, mouse broblasts. A decade of eorts have expanded its persona from a house keeper protein involved in mitochondrial import, energy generation
and chaperoning of misfolded proteins, to a guardian of stress that has multiple binding partners and to a killer protein that contributes
to carcinogenesis on one hand and to old age disorders on the other. Being proved to be an attractive target for cancer therapy, it also
warrants attention from the perspectives of management of old age diseases and healthy aging.
 2006 Elsevier Inc. All rights reserved.
Keywords: Mortalin/mthsp70/Grp75/PBP74; Overexpression; Lifespan extension; Immortalization; Cancer; Silencing; Growth arrest; Chaperone; Old
age diseases

1. The discovery of twin mortalins


Mortalin has come a long way since it was rst cloned
and identied to be associated with cellular mortality by
virtue of its presence in the cytosolic fractions of serially
passaged mouse embryonic broblasts (MEF), and of
mortal hybrids obtained by the fusion of mortal
(MEF) and immortal (MN48-1, derivative of NIH 3T3)
cells. Immortal cells seemed to lack this protein in their
cytosolic fractions. cDNA cloning and homology search
placed it in the heat shock protein 70 (hsp70) family
(Wadhwa et al., 1993a). Mouse immortal cells were later
found to harbor an allelic form of mortalin, mot-2 that
dier from mot-1 by two amino acids and located in
the perinuclear region of a cell (Wadhwa et al.,
1993b,c; Kaul et al., 2000a). Two forms of murine
mortalins were seen to have contrasting functions:
whereas an overexpression of the pancytosolic mot-1
protein to NIH 3T3 cells induced cellular senescence,
the perinuclear mortalin expressed by mot-2 cDNA
induced their malignant transformation (Wadhwa et al.,
*

Corresponding author. Tel.: +81 29 861 9464; fax: +81 29 861 2900.
E-mail address: renu-wadhwa@aist.go.jp (R. Wadhwa).

1993c; Kaul et al., 1998). The phenomenon of dierential


subcellular distribution of mortalin in mortal and immortal broblasts was conserved in human broblasts (Wadhwa et al., 1993b; Ran et al., 2000). However, genetic
cloning has not revealed the existence of more than
one human mortalin cDNAs. Human mortalin cDNA
was found to have transforming activity similar to mouse
mot-2 protein and thus was called hmot-2 (Kaul et al.,
1998). By chromosomal mapping mouse and human
mortalin have been assigned to chromosomes 18 and
5q31.1 (gene name HSPA9B), respectively (Kaul
et al., 1995; Ohashi et al., 1995). Expression and genomic
analysis of human mortalin revealed the presence of
2.8 kb human mortalin transcribed from an 18 kb region
on chromosome 5q31.1 that consisted of 17 exons with
boundaries almost identical to its murine counterpart
(Michikawa et al., 1993; Xie et al., 2000).
Mortalin is a 679 amino acids long (molecular weight
73,913 daltons) heat un-inducible member of Hsp70 family
of proteins. It has a high degree of identity with other
members of the Hsp70 family, including Escherichia coli
DnaK, Saccharomyces cerevisiae SSClp, the constitutive
cytosolic Hsp70 from rat, Hsc70 and the rat endoplasmic
reticulum isoform, BiP. The precursor protein has a

0531-5565/$ - see front matter  2006 Elsevier Inc. All rights reserved.
doi:10.1016/j.exger.2006.10.020

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
2

S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

46-amino acid long mitochondrial-targeting signal peptide.


It undergoes Ca-dependent autophosphorylation, has multiple subcellular sites and binding partners, and functions
related to the control of cell proliferation and stress
response (Kaul et al., 1993; Wadhwa et al., 1993b,
2002a). The crystal structure of mortalin has not been
elucidated so far. Based on its evolutionary conservation
within the Hsp70 family, it is expected to have two principal domains (the N-terminal ATPase and C-terminal
region) joined by a protease-sensitive site with its chaperone activities intimately linked with ATP-hydrolysis

(Fig. 1) (Deocaris et al., 2006a). A kettle pot model structure has been proposed in which the ATPase domain
(44-kDa) is expected to consist of four sub-domains that
fold into a pair of lobes forming a deep catalytic-cleft
(Fig. 1) (Sriram et al., 1997; Deocaris et al., 2006a). Studies
on E. coli Hsp70, DnaK, have demonstrated that its
ATPase activity can be cyclically stimulated by co-chaperones DnaJ and GrpE. DnaJ permits the hydrolysis of
Hsp70-bound ATP allowing the ADP-bound Hsp70 to
interact more strongly with unfolded proteins. The nucleotide exchange factor GrpE enables the recycling of Hsp70

Fig. 1. Homology modeling based structure of mortalin. Kettle pot model of the structure that includes its N-terminal ATP binding domain (pot handle),
middle substrate-binding domain (SBD) (pot) and C-terminal (lid) is shown on the left. Structure of the SBD and the lid as derived from the homology to
E. coli DnaK, were produced by SWISS-MODEL (http://swissmodel.expasy.org), INSIGHT II (Accelrys) and MOLSCRIPT (Kraulis, 1991). Universal
latch and mortalin-specic latch (that arises because of the electrostatic interactions among Arg574, Arg578 and Asp628, and should be largely perturbed
by the Gly624 Arg mutation) are shown. Chaperone function of mortalin adapted to its kettle pot structure is diagrammed to shown entry of unfolded
peptides into the SBD, chaperoning in the closed kettle pot and its release by unlocking and opening of the lid ensured by structural features of the protein.

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

back into an ATP-bound state permitting the ecient


release of its substrate (Harrison, 2003; Deocaris et al.,
2006a). As these cycles of ATP-hydrolysis occur, allosteric
changes in the N-terminus are transmitted to the 18-kDa
substrate-binding domain (SBD), made-up of two sets of
four-stranded anti-parallel beta-sheets that forms a twisted
sandwich (Fig. 1). While the ATPase domains are strictly
conserved, the SBD domain shows a greater sequence variation resulting in the diversication of the client peptides
and substrate specicity. Flanking the SBD is a 10-kDa
long C-terminal helix made-up of ve helical domains
(AE). This interesting structure, called the substrate
lid, does not contact the peptide substrate directly and
has the ability to ip-op to allow entry of unfolded peptides and release of folded peptides after chaperoning
(Fig. 1). The lid also functions as a molecular latch that
locks-in substrates during an ADP-bound state (Zhu et al.,
1996; Moro et al., 2004, 2005; Fernandez-Saiz et al., 2006)
(Fig. 1). Recent biophysical studies have provided new
clues to the mystery underlying the opposing functions of
the two mouse mortalin alleles (Deocaris et al., 2006c).
The helix A shows high sequence conservation among
the Hsp70 proteins whereas the distal portion of helix
(BE) is divergent. The two-amino acid dierences between
the two murine mortalin isoforms reside within this region
of the C-terminal helix. The V618M (mot-l:mot-2) amino
acid substitution is buried in the helix C and the R624G
(mot-l:mot-2) occurs near a critical helical bend and the
substitution of G (mot-2) into R (mot-1) causes some
changes in the inter-domain salt bridges. This can be appreciated from the understanding of the exible lid structure
above the substrate cleft of mortalin where electrostatic
latches between the lid and the cleft appear to be important for its functioning as a chaperone. One latch consisting of Asp477, Arg513, Glu586 and His590 is
common with other HSP70s (Mayer et al., 2000), while
an additional latch that we identied on the opposite
end, consisting of Arg574, Arg578 and Glu628, is mortalin-specic. A replacement of Gly624 (in mot-2), located
at the C-terminus of a-helix C, by Arg (in mot-1) is likely
to extend the a-helix C. In contrast, Gly, a strong helix
breaker, shortens the L3 (CD) loop. The latter should perturb the structure of the mortalin-specic latch, presumably pulling apart the electrostatic attractions that could
result in the loss of the chaperone action of the molecule.

tion of yeast mitochondria (Kawai et al., 2001). During


import of mitochondrial-targeted proteins, the proteins
pass through the membranes via the outer membrane
(TOM) and followed by the inner membrane (TIM) channels. In the process, bulky proteins are required to unfold
to permeate through a translocation pore and are then
refolded back to their native conformers to regain function
(Voos and Rottgers, 2002). There are two controversial
hypotheses on how the mortalin machinery performs its
protein translocation function across the two mitochondrial membranes, the trapping (Strub et al., 2000; Geissler
et al., 2001) and the motor models (Glick, 1995; Horst
et al., 1997; Voos et al., 1996; Deocaris et al., 2006a).
Yamamoto et al. (2005) have identied 15-kDa Timl5/
Ziml7 protein that associates and cooperates with mortalin/mthsp70 to facilitate import of presequence-containing
proteins into the matrix. Self-aggregation of mortalin/
mthsp70 is prevented by Hepl (Sichting et al., 2005). Following the translocation of proteins into the mitochondria,
they refold back and assemble to its native oligomeric state
and sort into various compartments of the organelle to
perform their functions. Newly imported mitochondrial
pre-proteins interact with mortalin/mthsp70 and Hsp60
as soon as they reach the matrix compartment (Mahlke
et al., 1990; Langer and Neupert, 1991; Hartl et al.,
1992). The activity of these mitochondrial chaperones is
life-essential as was demonstrated in yeast and mammalian
cells (Cheng et al., 1989; Wadhwa et al., 2005; Deocaris
et al., 2006a). Interestingly, overexpression of mortalin,
but not hsp60, resulted in lifespan extension of normal
human broblasts (Wadhwa et al., 2005). A recent study
identied mortalin in the lipid rafts from dierent mouse
organs, supporting its role as a component of the oxidationreduction respiratory chain (Kim et al., 2006). Rivolta and Holley (2002) have reported that the mitochondria
in general, and mortalin in particular, were asymmetrically
distributed during cell division and are involved in cell fate
determination. Another house keeping function of mortalin is predicted from its involvement in proteasomal degradation of proteins, mediated by its interactions with CHIP
(carboxyl terminus of Hsc70-Interacting Protein with ubiquitin E3-ligase activity) (Li et al., 2005; Deocaris et al.,
unpublished observation).

2. Mortalin in its housekeeping mode

The biological impact of mortalin function is not


restricted to its mitochondrial locale. Subcellular fractionation and immunouorescence microscopy have revealed
that mortalin is not only present in mitochondria but also
in other extra-mitochondrial sites (Ran et al., 2000; Poindexter et al., 2002). In parallel, its multiple binding partners
have revealed its diverse functional skills (Fig. 2). Although
mortalin did not consent with other members of heat shock
protein 70 family due to its un-inducibility to heat shock
(Domanico et al., 1993; Wadhwa et al., 1993a; Bhattacharyya et al., 1995; Mitchell et al., 2002), various mild stress

Mortalin is the major mitochondrial protein (Bhattacharyya et al., 1995) and it plays a central role in the elaborate translocation system for ecient import and export
of proteins (Koehler, 2004; Rehling et al., 2004; Wiedemann et al., 2004). Its role in cell viability and mitochondrial biogenesis was underscored by experimental data
including (i) the yeast cells knocked out for mortalin
homologue (Sscl) were lethal (Craig et al., 1987) and (ii)
the loss of function mutations of mthsp70 caused aggrega-

3. Mortalin in its guardian mode

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
4

S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

3.2. Internalization of receptors

Fig. 2. A diagram showing mortalin (grey star) and its interacting proteins
(white ovals).

Interleukin-1 (IL-1) is a major proinammatory cytokine


mediating local and systemic responses of the immune system. Two types of IL-1 receptors are known, but only the
IL-1 receptor type I initiates biological responses. It was
shown that mortalin is associated with the IL-1 receptor
type I and is involved in internalization of the receptor
(Sacht et al., 1999). Receptor for hyaluronan mediated
motility (RHAMM), a centrosomal and microtubal, hyaluronan-binding protein was shown to interact with mortalin
in interphase microtubules, but not in mitotic spindles. It
was proposed that mortalinRHAMM complexes may
stabilize microtubules in the interphase (Kuwabara et al.,
2006). Mortalin was also found to be upregulated in
isolated rodent islets exposed to cytokines. In two rat strains
that showed dierent sensitivity to the toxic eects of cytokines, a signicant dierences in IL-1beta mediated mortalin expression was observed suggesting its role in cytokine
induced beta-cell destruction (Johannesen et al., 2004).
3.3. Protein modications

responses including glucose-deprivation, low doses of ionizing radiation and calorie restriction were shown to induce
mortalin (Massa et al., 1995; Merrick et al., 1997; Sadekova et al., 1997; Gao et al., 2003; Um et al., 2003b; Tsuchiya et al., 2004; Liu et al., 2005). Besides, it ensures
nal destiny of other proteins, modies their functions
and act as a guardian against stress and apoptosis (Taurin
et al., 2002; Um et al., 2003a; Craven et al., 2005; Jin et al.,
2006a) (Fig. 2).
3.1. Intracellular tracking
Fibroblast growth factor-1 (FGF-1) regulates cell
growth and dierentiation. It lacks signal peptide and is
intracellularly localized as a result of endogenous expression or endocytosis. Mortalin was isolated as a FGF-1binding protein from rat L6 cells. Additionally, the two
proteins were found to share the same intracellular niche
and interact physically suggesting that mortalin is involved
in the tracking and/or signaling by FGF-1 (Mizukoshi
et al., 1999, 2001). FGF-1 added to the culture medium
of quiescent BALB/c3T3 cells was shown to interact with
mortalin in the cells in a regulated manner. Although both
the internalized FGF-1 and mortalin were present at high
levels throughout the FGF-1-initiated cell cycle, their interaction became apparent only in late Gl phase. Mortalin
was preferentially tyrosine phosphorylated at Gl. When
cells were treated with vanadate, a strong interaction
between mortalin and FGF-1 was established. Conversely,
treatment with tyrosine phosphatase abrogated mortalin
FGF-1 interactions suggesting that the interactions occur
preferentially in late Gl phase of the cell cycle, and are regulated by tyrosine phosphorylation of mortalin (Mizukoshi
et al., 1999, 2001).

Mevalonate pyrophosphate decarboxylase (MPD), an


enzyme that furnish prenyl groups required for prenylation
(protein modication that is essential for the activity of
many proteins including p21Ras) was identied as a binding partner for mortalin. A functional link between mot-2
(mortalin), MPD and p21Ras in control of cell proliferation was proposed (Wadhwa et al., 2003b). Voltage-dependent anion-selective channel 1 (VDAC1), a eukaryotic
porin that functions as a channel in membranous structures, was found to interact with the dynein light chain
Tctex-1 and mortalin in vivo. The functional relevance of
the identied protein interactions was analyzed in planar
lipid bilayer (PLB) experiments in which both recombinant
binding partners altered the electrophysiological properties
of hVDACl. While rTctex-1 increases the voltage-dependence of hVDAC1 slightly, mortalin drastically minimized
the voltage-dependence, indicating a modulation of channel properties (Schwarzer et al., 2002). Grp94, Glucose regulated protein 94, a member of the hsp90 family, was
shown to interact with mortalin (Takano et al., 2001).
3.4. Stress and immune responses
Several studies have indicated that an upregulation of
mortalin suppresses the engagement of apoptosis from various stressors, e.g., arsenite in rat lung epithelial cells (Lau
et al., 2004), mercury in renal cells (Stacchiotti et al., 2006;
dierentiation agent 1,25-dihydroxyvitamin D3 in rat gliomas (Baudet et al., 1998), and glucose starvation and ischemia reperfusion in Chinese Hamster Lung (CHL) cells
(Gao et al., 2003). High level of mortalin has been detected
as an adaptive response to high activity such as treadmill
running (Mattson et al., 2000). Taurin et al. (2002) have
shown that the transient transfection of vascular smooth

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

muscle cells (VSMCs) with mortalin cDNA led to a delayed


apoptotic response after serum deprivation. The expression
of tumor suppressor gene, p53, in mortalin transfected cells
was delayed to the same extent, and the expressed protein
showed abnormal perinuclear distribution suggesting that
p53 is retained and inactivated by mortalin. The study
dened a novel mortalin-mediated [Na+]i/[K+]i-responsive
signaling pathway that may play an important role in the
regulation of programmed cell death in VSMCs. Remodeling of vessel walls is one of the major determinants of longterm blood pressure elevation and an independent risk factor for cardiovascular morbidity and mortality. Drugs that
are known to protect vascular cells from apoptotic stimuli,
such as cardiotonic steroids (CTS), have been shown to target the Na/K-ATPase pump. Another study has detected
the role of mortalin in CTS-inhibited apoptosis of vascular
smooth muscles cells (Orlov and Hamet, 2006). Associated
with onset of glucose-deprivation treatment is the rapid
increase in ROS accumulation that is reduced with mortalin overexpression suggesting that mortalin could inhibit
the ROS accumulation, and has a cytoprotective eect
(Liu et al., 2005). In another study, mortalin/mthsp70
was identied as one of the proteins stimulated after transient exposure of human endothelial cells to sub-lethal levels of hydroperoxides suggesting its involvement in
oxidative stress response (Mitsumoto et al., 2002). Orsini
et al. (2004) showed that a fraction of cytosolic p66Shc
(regulates lifespan in mammals and is a critical component
of the apoptotic response to oxidative stress) localizes within mitochondria where it forms a complex with mitochondrial Hsp70/mortalin. Mortalin was shown to inhibit
p66Shc function activated during oxidative stress (ultraviolet radiation) that induced the dissociation of p66Shc
mortalin complexes. Saretzki et al. (2004) have identied
mortalin as one of the factors responsible for superior
stress defense in murine embryonic stem cells. It was identied to mediate cellular response to low dose ionizing radiations. Treatment of cells with antisense mortalin
oligonucleotide was found to sensitize cells to ionizing radiation (Sadekova et al., 1997). Um et al. (2003a) have proposed mortalin/mthsp70 as a DNA-PK regulated protein
(plays a protective role against drug-induced apoptosis)
that can be a determinant of drug sensitivity.
Another important major function of surface-expressed
mortalin is its captivating role in immune response, i.e.,
antigen presentation and in innate immunity. Complement-mediated cell death is caused by C5b-9, the membrane attack complex (MAC) that inicts damage to the
target cells. For protection, cells eliminate the MAC from
their surface either by ectocytosis (direct emission of membrane vesicles) or by endocytosis (internalization). Recently, the involvement of mortalin in MAC elimination has
been suggested. Extracellular application of antibodies
directed to mortalin increased cell sensitivity to MAC-mediated lysis. Other pore formers, such as streptolysin O and
melittin, did not induce release of mortalin. Mortalin was
shown to bind to complement C8 and C9 and is shed in

vesicles containing C9 and complement MACs. These


results suggest that mortalin promotes the shedding of
membrane vesicles loaded with complement MAC and protects cells from complement-mediated lysis (Pilzer and
Fishelson, 2005; Pilzer et al., 2005).
As described above, besides playing a prime role in bioenergetics-associated transport, sorting and refolding, as
mortalin travels outside the mitochondria, it collaborates
with a repertoire of binding partners and acquires more
expansive cellular roles. These mortalin-mediated manifold
of intracellular trajectories are exploited in a two-way
sword: while strengthening the yin of cellular energetic
and stress pathways, it leads to yang for mortalins killer
instincts behind many old age-associated diseases and cancer in particular.

4. Mortalin in its killer mode


4.1. In cancers
Tumors are known to lead more stressful lives compared to the normal cells. This is particularly the case considering that its immortal state carries added demands for
continuous rapid proliferation; competition for basic cellular needs (space, nutrient and oxygen); hostility of new cellular environments, particularly for adventurous
metastases; and most signicantly, the accumulation of
mutated proteins as a result of genomic instability. Just
as senescence is considered, at least in the Darwinian
school of thought, consequential to the declining force of
natural selection as a function of biological time; cancer
can be considered as a corrupted phenotype that escaped
this selective pressure. A notable survival strategy of cancer
cells is its considerable investments in bolstering its innate
molecular chaperones allowing the endowment of survival
advantages, such as, (a) in counteracting the stress of
senescence and apoptosis, (b) in its function as an micro-evolutionary buer that neutralizes conformational
consequences of mutant proteins, (c) aids in the acquisition
of gain-of-functions of chaperone-stabilized rogue proteins, (d) promotion of invasiveness and cell motility, (e)
co-ordination hyper-activation of proliferation signals
and (f) resistance to radiation, heat, hormones and chemotherapeutic agents (Soti and Csermely, 2000, 2002; Soti
et al., 2003).
Hsps serve as safeguards to maintain homeostasis and
integrity of protein interactions. The observation that
tumor cells often have elevated levels of Hsps may be associated to a pre-malignant cells response to the selection
process occurring during tumorigenesis. By virtue of their
activities as molecular chaperones, several of these Hsps
not just contribute to cellular immortalization, but also
enhance a neoplasias survival advantages. Primarily the
Hsp70 family, the constitutive Hsc70 (HS7C) and the heat
shock-inducible Hsp70 (HSPA1A); the Hsp90 family,
Hsp90b (HSPCB) and Hsp90a; and the mitochondrial

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
6

S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

Hsp27 (HSPB1), have been most widely ascribed with


oncogenic roles (Mosser and Morimoto, 2004).
The level of mortalin expression was found to be
elevated in many human tumors, and in all of the tumorderived and in vitro immortalized cells examined (Wadhwa
et al., 2006). In human embryonic broblasts immortalized
with an expression plasmid for hTERT, the telomerase catalytic subunit, with or without human papillomavirus E6
and E7 genes, the subclones with spontaneously increased
mortalin expression levels became anchorage-independent
and acquired the ability to form tumors in nude mice.
Furthermore, overexpression of mortalin was sucient
to increase the malignancy of breast carcinoma cells
suggesting that an upregulation of mortalin contributes signicantly to tumorigenesis (Wadhwa et al., 2006). Quantitative estimation of level of mortalin expression has
revealed that the tumor cells with higher level of mortalin
expression have more aggressive tumor phenotypes such
as metastasis (Wadhwa et al., 2006) (Fig. 3). The role of
mortalin has been more extensively probed in three tumor
types: brain, colon and leukemia.
4.1.1. Brain tumors
Immunohistochemical studies of mortalin in normal and
tumor human brain sections revealed that in normal brain
sections, the expression was seen mainly as being conned
to neurons. Normal astrocytes showed undetectable
expression of mortalin. Three grades of astrocyte tumors
(low-grade astrocytoma, anaplastic astrocytoma and glioblastoma), however, showed an increasing number of
mortalin-positive cells. Other types of brain tumors, such
as meningiomas, neurinomas, pituitary adenomas and
metastases, also showed elevated levels of mortalin expression compared to those in the normal brain. An increase in
number of mortalin-positive cells with malignant progression of brain tumors and its correlation with Ki-67 (a cell
proliferation marker)-positive cells further suggested an
involvement of nonpancytosolic mortalin(s) in malignant
transformation of cells in vivo (Takano et al., 1997).

Fig. 3. A quantitative estimation of mortalin expression in human


transformed cells showing the relationship of mortalin expression level
and metastatic property.

4.1.2. Colon cancers


Comparative proteomic analysis identied overexpression of mortalin in colorectal adenocarcinomas. By immunostaining on a colorectal cancer tissue microarray linked
to a patient database, mortalin overexpression was found
to correlate with poor patient survival. The ndings demonstrated that mortalin overexpression may predict outcome in colorectal cancer and suggested that mortalin is
involved in colorectal neoplasia (Dundas et al., 2004).
4.1.3. Leukemia
Human Chromosome 5q31.1 is frequently deleted in
myeloid leukemias and myelodysplasia suggesting that
mortalin may be a candidate gene (Xie et al., 2000). Myelodysplastic syndrome (MDS) comprises a heterogeneous
group of clonal blood disorders characterized by ineective
hematopoiesis arising from dysplasia and accelerated apoptotic death of multipotential hematopoietic progenitors
and their progeny. These syndromes display signicant
clinical variability that ranges from anemia, erythroid dysplasia, fatal multi-lineage peripheral cytopenia or acute
myeloid leukemia (AML). The majority of patients with
MDS die within 34 years. Deletions at 5q31 are associated
with more aggressive forms of MDS, often progressing rapidly to leukemia. A critical deleted region (CDR) at chromosome 5q31 has been dened containing nine candidate
genes, including mortalin/HSPA9B, but causative genes
have yet to be identied and are rather critical for diagnosis, improved therapies, and prevention.
Hematopoiesis is well characterized in zebrash and has
been demonstrated to have a high degree of conservation
with mammalian counterparts. zebrash mutants with
abnormalities at various stages in blood development have
been isolated by genetic screens and present mutants that
model human hematopoietic diseases. A developmental
blood mutant, crimsonless (crs), is anemic at the onset of
circulation and display defective blood cell dierentiation
followed by apoptosis and a reduction in erythrocytes,
granulocytes and hematopoietic progenitors resembling
the symptoms of MDS. Using positional cloning, RNA rescue and morpholino knockdown, Craven et al. (2005) have
identied the mutated gene in crs mutants in zebrash.
Hspa9b (mortalin/mthsp70/GRP75) that shows 84.8%
identity and 89.4% similarity with human HSPA9B. A single amino acid mutant (G492E) within the substrate-binding domain of HSPA9B was shown to be the cause of the
crs phenotype. A near-identical mutation in the conserved
glycine at position 443 in DnaK (53.5% identity and 63.3%
similarity to zebrash mortalin) completely abolished propeptide binding, rendering the chaperone functionless
(Burkholder et al., 1996). To verify that the mutation in
Hspa9b is sucient to cause the crs phenotype, Craven
et al. (2005) had used both rescue and antisense morpholino knockdown strategies. Injection of capped RNA encoding wild-type HSPA9B rescued approximately 95% (53 of
56) injected mutant embryos. The blood of rescued animals
was no longer hypochromic. Conversely, inactivation of

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

Zebrash mortalin using antisense morpholino-modied


oligonucleotides that targeted the 5 0 untranslated region
(UTR) and rst methionine recapitulated the anemic phenotype observed in crs mutants in 80% of the injected animals. This study has very strongly proved that a single
amino acid mutation, G492E that abolishes chaperone
function of mortalin is the cause of crs phenotype in Zebrash, a model of human MDS. In an independent comparative proteomic study, Pizzatti et al. (2006) have identied
mortalin as one of the 31 dierentially expressed proteins in
protein proles from chronic myeloid leukemia and healthy
bone marrow donors.
Mortalin and p53 interactions were rst identied in the
cytoplasm of tumor cells. It was shown that mortalin
sequesters p53 in the cytoplasm and inhibits normal transcriptional activation function of p53 (Wadhwa et al.,
1998, 2000a,b, 2002b; Kaul et al., 2001; Taurin et al.,
2002; Mihara et al., 2003). Mortalin binding p53 peptides
were shown to reactivate p53 function, associated with
growth arrest of cancer cells (Kaul et al., 2005). Most
recently, wild-type p53 and mortalin interactions were also
demonstrated in the cytoplasm of the soft shell leukemic,
but not normal, clam hemocytes that develop a fatal neoplasm sharing molecular similarity with an unrelated group
of human cancers. Mortalinp53 interactions were abrogated both in the mammalian and clam cells by a cationic
inhibitor of mortalin (MKT-077) resulting in reactivation
of wild-type p53 function (Wadhwa et al., 2000b, 2002a;
Walker et al., 2006). A recent study on the comparative
mass spectrometric analysis of unduplicated and duplicated
centrosomes identied mortalin as a protein that associates
preferentially with duplicated centrosomes. For the rst
time, it was localized to centrosomes where it formed physical complexes with p53 during late G1, S and G2, and dissociated from centrosomes during mitosis. Overexpression
of mortalin over-ruled the p53-dependent suppression of
centrosome duplication, assigning it as an upstream regulator of p53 function in control of centrosome duplication
(Ma et al., 2006). Mortalinp53 complexes in the cytoplasm or in centrosome compromise wild-type p53 activities leading to uncontrolled proliferation, a hallmark of
cancers. Overexpression of mortalin in normal human cells
was able to extend their in vitro lifespan of broblasts
(Kaul et al., 2000b; Wadhwa et al., 2005). Since mortalinp53 interactions have not been detected in normal cells,
it is likely that lifespan extension is the result of mortalin
functions independent to that of p53-inactivation.
4.2. In other old age diseases
4.2.1. Diabetes
Hyperglycemia induces the production of reactive oxygen species (ROS) from the mitochondria. During diabetic
hyperglycemia, levels and induction response of Hsp70, but
not Hsp105 and Hsp90, are markedly decreased in the liver
(Yamagishi et al., 2004; Muranyi et al., 2005). An antidiabetic response based on the enhanced eciency of

import of anti-oxidative enzymes such as superoxide dismutase and glutathione peroxidase with the boosting of
the mitochondrial import machinery has been proposed
(Matsuoka et al., 2005).
4.2.2. Neurodegenerative diseases
The demise of specic neuronal populations due to the
accumulation of abnormal polypeptides forms the etiologic
basis of several neurodegenerative diseases, such as Alzheimers disease, Parkinsons disease, Huntington disease,
spinocerebellar ataxia, etc. Such misfolded protein conformers have a strong tendency to form neurotoxic insoluble protein aggregates. A very important consequence is
the impairment of the ubiquitin-proteasome degradation
system and suppression of the heat shock and oxidative
stress response. Accumulation of the aggregated proteins
also activates signal transduction pathways that lead to cell
death (Soti and Csermely, 2002).
A study on the hippocampus from ApoE knockout
mice, a model for Alzheimers disease, revealed that mortalin is among the six oxidized proteins suggesting it to be
involved in risk and progression of Alzheimers disease
(Choi et al., 2004). In another study, on unbiased quantitative proteomic approach, nigral mitochondrial proteins
of Parkinsons disease (PD) patients were compared with
those from age-matched control. Mortalin was found to
be substantially decreased in PD brains. Manipulations
of mortalin level in dopaminergic neurons resulted in signicant changes in sensitivity to PD phenotypes via pathways involving mitochondrial and proteasomal function
as well as oxidative stress suggesting that it is a target
for oxidative stress, a marker for PD pathology (Jin
et al., 2006a). It is suggestive that mortalin is a regulator
of oxidative stress and apoptosis, and contributes to aging
and old age pathologies. Li et al. (2005) have shown that
DJ-1, a causative gene for familial form of the PD, associates with chaperones including Hsp70, CHIP, and mortalin/mtHsp70 and gets translocated to mitochondria upon
oxidative stress. Another study has identied mortalin as
one of the ve major proteins (mortalin, nucleolin, grp94,
calnexin and clathrin) binding to a-synuclein and DJ-1,
two critical proteins in PD pathogenesis (Jin et al.,
2006b). In parallel to the extended lifespan of
worms overexpressing mortalin/hsp70F, it was seen to
decrease during normal nematode aging. Of note,
HSP70F siRNA caused reduction in worm lifespan and
early appearance of progeria like phenotype and age pigments (Yokoyama et al., 2002) (Kimura et al., personal
communication).
These studies have suggested that mortalin may serve as
a longevity factor due to its diverse functions described
above. In the battle of its longevity functions, its activities
may be compromised by its structural and chemical modications. Some modications may even convert it from an
ecient-chaperone to a sick-chaperone or to an anti-chaperone molecule that would build up the garbage catastrophe often considered as a marker of old age disorders.

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
8

S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

Some examples of such modications of mortalin have


already been found (Bruschi et al., 1993; Bruschi and Lindsay, 1994; Cooper et al., 2001; Choi et al., 2004).
5. Perspectives: mortalin-based interventions
Bearing in mind the three egos of mortalin i.e., as an
essential housekeeping gene, as a guardian chaperone that
is activated during stress and cause killing of the old, it may
seem rather audacious to therapeutically control this chaperones temperaments. Some hints, however, at ne-tuning
these mortalin paradoxes for achieving healthy aging and
to forward novel approaches in treating of aging-associated
diseases, such as cancer, can be glimpsed from several key
basic studies over the past decade.
5.1. Strengthening the guardian
The level of mortalin protein was found to decrease in
senescent human broblasts (MRC5 and HFF) and in
aged worms (unpublished data). Its overexpression leads
to the lifespan extension in both systems suggesting that
the decline in mortalin level and thus its function(s) determines xed proliferation potential of normal cells and
aging, respectively. The lifespan of human foreskin broblasts (HFF5), cultured under standard in vitro conditions, was extended slightly by expression of exogenous
mortalin, but not by the catalytic subunit of telomerase,
hTERT. Together, mot-2 (mortalin) and hTERT permitted bypass of senescence, a substantial extension of lifespan, and possibly immortalization demonstrating that
mortalin and telomerase can cooperate in the immortalization process (Kaul et al., 2003). Since upregulation of
mortalin expression and activation of telomerase are common features of cancers, it is likely that these cooperate
in vivo during the development of human cancers. On a
similar context, transient induction of hsp70F led to a
slight (<13%) extension in the Caenorhabditis elegans lifespan; and transgenic worms that constitutively over-expressed hsp70F predominantly in muscle showed
lifespan extension (approximately 43% for mean and
45% for maximum lifespan) as compared to the wild-type
worms (Yokoyama et al., 2002). Together, the ndings
that (i) the level of mortalin decreases during normal
aging of cells and in PD brain (Jin et al., 2006a), and
(ii) its overexpression leads to lifespan extension of normal cells are suggestive that mortalin may be a lifespan
determining factor, possibly due to its role as a mitochondrial importer, chaperone, intracellular tracking and
ROS management. Strengthening of these functions by
mortalin supplementation may lead to youthful rejuvenation of normal cells. However, it is important to control
the dose of mortalin so that the rejuvenation does not
lead to transformation. Oxidation-resistant engineered
mortalin that may functionally sustain itself even in damaging cellular environment and self-regulating mortalin
are attractive possibilities.

5.1.1. Estrogen therapy


The use of hormones constitutes a potentially new
option in the prevention and treatment of age-related diseases, howevever, it is not an established therapy. There
is an accumulation of evidence of estrogens as a neuroprotective agent, with the mitochondria as an important target
(Singh et al., 2006). Of note, together with SOX5, RBM15,
dynein, mortalin was shown to be regulated by estrogen
receptor-alpha gene (ESR1) via the homeobox transcription factor BAEX2 (Stevens and Meech, 2006). So far, this
is the rst study that has been able to elucidate the eectors
in the transcriptional control of mortalin. Thyroid hormone was also found to induce elevations in mortalin/
mthsp70 along with an outer membrane receptor Tom20
(Craig et al., 1998).
5.1.2. Exercise
In chronically stimulated skeletal muscles, increased
level of mortalin expression was correlated with increased
mitochondrial activity. It has been posited that such induction is needed to keep pace with mitochondrial biogenesis
and to ensure the rate of protein import in chronically stimulated skeletal muscle (Ornatsky et al., 1995). The phenomenon of mitochondrial adaptation to exercise appears to
occur rapidly. During a 3-month program of treadmill running, soleus muscle of Wistar rats show upregulated levels
of mortalin as well as the b-subunit of the mitochondrial
Hsp60, F1-ATPase, Hsp72 and GRP78. However, when
the animals have rested for 3 days, the rates of synthesis
of mortalin and HSP72 drop to levels that are even lower
than that of sedentary animals (Gonzalez et al., 2000;
Mattson et al., 2000).
5.1.3. Glycerol therapy
The concept of glycerol therapy as potential anti-aging
modulation has recently been reported, based on this
chemical chaperones ability to alleviate protein aggregation (Deocaris et al., 2006b). More importantly, glycerol
was found to induce mortalin expression, its chaperoning
and proteasomal functions (Deocaris et al., unpublished
observation).
5.2. Weakening the killer
Killer function of mortalin is viewed as its proproliferative support to cancer cells as described above. Suppression
of mortalin in cancer cells is, therefore, suggested as a therapeutic tool. Indeed, compromising mortalin expression by
functional small RNAs including ribozymes, antisense and
siRNA in human cancer cells led to their growth arrest or
apoptosis (Wadhwa et al., 2003a, 2004; unpublished
observation).
5.2.1. Small molecules
MKT-077, a cationic rhodacyanine dye analogue, has
been under preclinical cancer therapeutical trials (because
of its selective toxicity to cancer cells) binds to mortalin

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

(mot-2) and abrogates its interactions with the tumor suppressor protein, p53. In cancer cells, but not in normal
cells, MKT-077 induced release of wild-type p53 from cytoplasmically sequestered p53mot-2 complexes and rescued
its transcriptional activation function. Thus, MKT-077
could be one of the candidates for therapy of cancers with
wild-type p53 (Wadhwa et al., 2000b; Walker et al., 2006).
Induction of senescence like growth arrest by bromodeoxyuridine (Michishita et al., 1999) and of 5-aza-2 0 deoxycytidine (Widodo et al., unpublished observation) also caused
shift of subcellular distribution of mortalin from perinuclear to pancytoplasmic type.
5.2.2. Synthetic peptides
Mortalin binds to a carboxyl terminus region of the
tumor suppressor protein p53. By in vivo co-immunoprecipitation of mortalin with p53 and its deletion mutants,
the mot-2-binding site of p53 was mapped to carboxyl terminus 312352 amino acid residues of mortalin. Furthermore, mot-2p53 interactions were disrupted by
overexpression of the short p53 carboxyl-terminal peptides
that bind to mortalin. This was accompanied by nuclear
translocation of p53 and growth arrest of human osteosarcoma and breast carcinoma cells (Kaul et al., 2005).
5.2.3. Immunotherapy
A challenging hypothesis mimotope-hormesis, which
shows evidence for epitope mimicry (mimotopy) between
mortalin and various hsp70s of infectious agents has been
proposed (Deocaris et al., 2005). From this mimotope phenomenon, it has been proposed that assaults of infection
during early adulthood could fortify the immune system
to evoke more potent defenses against late-onset diseases,
such as cancer, via autoimmunity. Interestingly, both
experimental and clinical data support the benecial role
of autoimmunity in long-term cancer survivors. Thus,
among the clinical applications, mortalin-based vaccine
or antibody treatment will certainly be a powerful tool in
our ght against cancer.
Acknowledgement
Authors thank Zeenia Kaul for the artwork.
References
Baudet, C., Perret, E., Delpech, B., Kaghad, M., Brachet, P., Wion, D.,
Caput, D., 1998. Dierentially expressed genes in C6.9 glioma cells
during vitamin D-induced cell death program. Cell Death Dier. 5,
116125.
Bhattacharyya, T., Karnezis, A.N., Murphy, S.P., Hoang, T., Freeman,
B.C., Phillips, B., Morimoto, R.I., 1995. Cloning and subcellular
localization of human mitochondrial hsp70. J. Biol. Chem. 270, 1705
1710.
Bruschi, S.A., Lindsay, J.G., 1994. Mitochondrial stress protein actions
during chemically induced renal proximal tubule cell death. Biochem.
Cell. Biol. 72, 663667.
Bruschi, S.A., West, K.A., Crabb, J.W., Gupta, R.S., Stevens, J.L., 1993.
Mitochondrial HSP60 (PI protein) and a HSP70-like protein (mort-

alin) are major targets for modication during S-(1,1,2,2-tetrauoroethyl)-L-cysteine-induced nephrotoxicity. J. Biol. Chem. 268, 23157
23161.
Burkholder, W.F., Zhao, X., Zhu, X., Hendrickson, W.A., Gragerov, A.,
Gottesman, M.E., 1996. Mutations in the C-terminal fragment of
DnaK aecting peptide binding. Proc. Natl. Acad. Sci. USA 93,
1063210637.
Cheng, M.Y., Hartl, F.U., Martin, J., Pollock, R.A., Kalousek, F.,
Neupert, W., Hallberg, E.M., Hallberg, R.L., Horwich, A.L.,
1989. Mitochondrial heat-shock protein hsp60 is essential for
assembly of proteins imported into yeast mitochondria. Nature
337, 620625.
Choi, J., Forster, M.J., McDonald, S.R., Weintraub, S.T., Carroll, C.A.,
Gracy, R.W., 2004. Proteomic identication of specic oxidized
proteins in ApoE-knockout mice: relevance to Alzheimers disease.
Free Radic. Biol. Med. 36, 11551162.
Cooper, A.J., Wang, J., Gartner, C.A., Bruschi, S.A., 2001. Co-purication of mitochondrial HSP70 and mature protein disulde isomerase
with a functional rat kidney high-M(r) cysteine S-conjugate beta-lyase.
Biochem. Pharmacol. 62, 13451353.
Craig, E.A., Kramer, J., Kosic-Smithers, J., 1987. SSC1, a member of the
70-kDa heat shock protein multigene family of Saccharomyces
cerevisiae, is essential for growth. Proc. Natl. Acad. Sci. USA 84,
41564160.
Craig, E.E., Chesley, A., Hood, D.A., 1998. Thyroid hormone modies
mitochondrial phenotype by increasing protein import without altering
degradation. Am. J. Physiol. 275, C1508C1515.
Craven, S.E., French, D., Ye, W., de Sauvage, F., Rosenthal, A., 2005.
Loss of Hspa9b in zebrash recapitulates the ineective hematopoiesis
of the myelodysplastic syndrome. Blood 105, 35283534.
Deocaris, C.C., Taira, K., Kaul, S.C., Wadhwa, R., 2005. Mimotope-hormesis and mortalin/grp75/mthsp70: a new hypothesis on
how infectious disease-associated epitope mimicry may explain
low cancer burden in developing nations. FEBS Lett. 579, 586
590.
Deocaris, C.C., Kaul, S.C., Wadhwa, R., 2006a. On the brotherhood of
the mitochondrial chaperones mortalin and heat shock protein 60. Cell
Stress Chaperones 11, 116128.
Deocaris, C.C., Shrestha, B.G., Kraft, D.C., Yamasaki, K., Kaul, S.C.,
Rattan, S.I., Wadhwa, R., 2006b. Geroprotection by glycerol: insights
to its mechanisms and clinical potentials. Ann. NY Acad. Sci. 1067,
488492.
Deocaris, C.C., Yamasaki, K., Kaul, S.C., Wadhwa, R., 2006c. Structural
and functional dierences between mouse mot-1 and mot-2 proteins
that dier in two amino acids. Ann. NY Acad. Sci. 1067, 220223.
Domanico, S.Z., DeNagel, D.C., Dahlseid, J.N., Green, J.M., Pierce,
S.K., 1993. Cloning of the gene encoding peptide-binding protein 74
shows that it is a new member of the heat shock protein 70 family.
Mol. Cell. Biol. 13, 35983610.
Dundas, S.R., Lawrie, L.C., Rooney, P.H., Murray, G.I., 2004. Mortalin
is over-expressed by colorectal adenocarcinomas and correlates with
poor survival. J. Pathol. 205, 7481.
Fernandez-Saiz, V., Moro, F., Arizmendi, J.M., Acebron, S.P., Muga, A.,
2006. Ionic contacts at DnaK substrate binding domain involved in the
allosteric regulation of lid dynamics. J. Biol. Chem. 281, 74797488.
Gao, C.X., Zhang, S.Q., Yin, Z., Liu, W., 2003. Molecular chaperone
GRP75 reprove cells from injury caused by glucose deprivation. Shi
Yan Sheng Wu Xue Bao 36, 381387.
Geissler, A., Rassow, J., Pfanner, N., Voos, W., 2001. Mitochondrial
import driving forces: enhanced trapping by matrix Hsp70 stimulates
translocation and reduces the membrane potential dependence of
loosely folded preproteins. Mol. Cell. Biol. 21, 70977104.
Glick, B.S., 1995. Pathways and energetics of mitochondrial protein
import in Saccharomyces cerevisiae. Methods Enzymol. 260, 224231.
Gonzalez, B., Hernando, R., Manso, R., 2000. Stress proteins of 70 kDa
in chronically exercised skeletal muscle. Pugers Arch. 440, 4249.
Harrison, C., 2003. GrpE, a nucleotide exchange factor for DnaK. Cell
Stress Chaperones 8, 218224.

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
10

S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

Hartl, F.U., Martin, J., Neupert, W., 1992. Protein folding in the cell: the
role of molecular chaperones Hsp70 and Hsp60. Annu. Rev. Biophys.
Biomol. Struct. 21, 293322.
Horst, M., Oppliger, W., Rospert, S., Schonfeld, H.J., Schatz, G., Azem,
A., 1997. Sequential action of two hsp70 complexes during protein
import into mitochondria. EMBO J. 16, 18421849.
Jin, J., Hulette, C., Wang, Y., Zhang, T., Pan, C., Wadhwa, R., Zhang, J.,
2006a. Proteomic identication of a stress protein, mortalin/mthsp70/
GRP75: relevance to Parkinsons disease. Mol. Cell Proteomics 5,
11931204.
Jin, J., Li, G.J., Davis, J., Zhu, D., Wang, Y., Pan, C, Zhang, J., 2006b.
Identication of novel proteins interacting with both a-synuclein and
DJ-1. Mol. Cell Proteomics [Epub ahead of print].
Johannesen, J., Pie, A., Karlsen, A.E., Larsen, Z.M., Jensen, A., Vissing,
H., Kristiansen, O.P., Pociot, F., Nerup, J., 2004. Is mortalin a
candidate gene for T1DM? Autoimmunity 37, 423430.
Kaul, S.C., Wadhwa, R., Komatsu, Y., Sugimoto, Y., Mitsui, Y., 1993.
On the cytosolic and perinuclear mortalin: an insight by heat shock.
Biochem. Biophys. Res. Commun. 193, 348355.
Kaul, S.C., Wadhwa, R., Matsuda, Y., Hensler, P.J., Pereira-Smith, O.M.,
Komatsu, Y., Mitsui, Y., 1995. Mouse and human chromosomal
assignments of mortalin, a novel member of the murine hsp70 family
of proteins. FEBS Lett. 361, 269272.
Kaul, S.C., Duncan, E.L., Englezou, A., Takano, S., Reddel, R.R.,
Mitsui, Y., Wadhwa, R., 1998. Malignant transformation of NIH3T3
cells by overexpression of mot-2 protein. Oncogene 17, 907911.
Kaul, S.C., Duncan, E., Sugihara, T., Reddel, R.R., Mitsui, Y., Wadhwa,
R., 2000a. Structurally and functionally distinct mouse hsp70 family
members Mot-1 and Mot-2 proteins are encoded by two alleles. DNA
Res. 7, 229231.
Kaul, S.C., Reddel, R.R., Sugihara, T., Mitsui, Y., Wadhwa, R., 2000b.
Inactivation of p53 and life span extension of human diploid
broblasts by mot-2. FEBS Lett. 474, 159164.
Kaul, S.C., Reddel, R.R., Mitsui, Y., Wadhwa, R., 2001. An N-terminal
region of mot-2 binds to p53 in vitro. Neoplasia 3, 110114.
Kaul, S.C., Yaguchi, T., Taira, K., Reddel, R.R., Wadhwa, R., 2003.
Overexpressed mortalin (mot-2)/mthsp70/GRP75 and hTERT cooperate to extend the in vitro lifespan of human broblasts. Exp. Cell Res.
286, 96101.
Kaul, S.C., Aida, S., Yaguchi, T., Kaur, K., Wadhwa, R., 2005.
Activation of wild type p53 function by its mortalin-binding,
cytoplasmically localizing carboxyl terminus peptides. J. Biol. Chem.
280, 3937339379.
Kawai, A., Nishikawa Si, S., Hirata, A., Endo, T., 2001. Loss of the
mitochondrial Hsp70 functions causes aggregation of mitochondria in
yeast cells. J. Cell Sci. 114, 35653574.
Kim, K.B., Lee, J.W., Lee, C.S., Kim, B.W., Choo, H.J., Jung, S.Y., Chi,
S.G., Yoon, Y.S., Yoon, G., Ko, Y.G., 2006. Oxidationreduction
respiratory chains and ATP synthase complex are localized in
detergent-resistant lipid rafts. Proteomics 6, 24442453.
Koehler, C.M., 2004. New developments in mitochondrial assembly.
Annu. Rev. Cell Dev. Biol. 20, 309335.
Kraulis, P., 1991. MOLSCRIPT: a program to produce both detailed and
schematic plots of protein structures. J. Appl. Cryst. 24, 946950.
Kuwabara, H., Yoneda, M., Hayasaki, H., Nakamura, T., Mori, H., 2006.
Glucose regulated proteins 78 and 75 bind to the receptor for
hyaluronan mediated motility in interphase microtubules. Biochem.
Biophys. Res. Commun. 339, 971976.
Langer, T., Neupert, W., 1991. Heat shock proteins hsp60 and hsp70: their
roles in folding, assembly and membrane translocation of proteins.
Curr. Top Microbiol. Immunol. 167, 330.
Lau, A.T., He, Q.Y., Chiu, J.F., 2004. A proteome analysis of the arsenite
response in cultured lung cells: evidence for in vitro oxidative stressinduced apoptosis. Biochem. J. 382, 641650.
Li, H.M., Niki, T., Taira, T., Iguchi-Ariga, S.M., Ariga, H., 2005.
Association of DJ-1 with chaperones and enhanced association and
colocalization with mitochondrial Hsp70 by oxidative stress. Free
Radic. Res. 39, 10911099.

Liu, Y., Liu, W., Song, X.D., Zuo, J., 2005. Eect of GRP75/mthsp70/
PBP74/mortalin overexpression on intracellular ATP level, mitochondrial membrane potential and ROS accumulation following glucose
deprivation in PC 12 cells. Mol. Cell Biochem. 268, 4551.
Ma, Z., Izumi, H., Kanai, M., Kabuyama, Y., Ahn, N.G., Fukasawa, K.,
2006. Mortalin controls centrosome duplication via modulating
centrosomal localization of p53. Oncogene 25, 53775390.
Mahlke, K., Pfanner, N., Martin, J., Horwich, A.L., Hartl, F.U., Neupert,
W., 1990. Sorting pathways of mitochondrial inner membrane
proteins. Eur. J. Biochem. 192, 551555.
Massa, S.M., Longo, F.M., Zuo, J., Wang, S., Chen, J., Sharp, F.R., 1995.
Cloning of rat grp75, an hsp70-family member, and its expression in
normal andischemic brain. J. Neurosci. Res. 40, 807819.
Matsuoka, T., Wada, J., Hashimoto, I., Zhang, Y., Eguchi, J., Ogawa, N.,
Shikata, K., Kanwar, Y.S., Makino, H., 2005. Gene delivery of tim44
reduces mitochondrial superoxide production and ameliorates neointimal proliferation of injured carotid artery in diabetic rats. Diabetes
54, 28822890.
Mattson, J.P., Ross, C.R., Kilgore, J.L., Musch, T.I., 2000. Induction of
mitochondrial stress proteins following treadmill running. Med. Sci.
Sport. Exer. 32, 365369.
Mayer, M.P., Rudiger, S., Bukau, B., 2000. Molecular basis for
interactions of the DnaK chaperone with substrates. Biol. Chem.
381, 877885.
Merrick, B.A., Walker, V.R., He, C., Patterson, R.M., Selkirk, J.K., 1997.
Induction of novel Grp75 isoforms by 2-deoxyglucose in human and
murine broblasts. Cancer Lett. 119, 185190.
Michikawa, Y., Baba, T., Arai, Y., Sakakura, T., Kusakabe, M., 1993.
Structure and organization of the gene encoding a mouse mitochondrial stress-70 protein. FEBS Lett. 336, 2733.
Michishita, E., Nakabayashi, K., Suzuki, T., Kaul, S.C., Ogino, H., Fujii,
M., Mitsui, Y., Ayusawa, D., 1999. 5-Bromodeoxyuridine induces
senescence-like phenomena in mammalian cells regardless of cell type
or species. J. Biochem. 126, 10521059.
Mihara, M., Erster, S., Zaika, A., Petrenko, O., Chittenden, T., Pancoska,
P., Moll, U.M., 2003. p53 has a direct apoptogenic role at the
mitochondria. Mol. Cell 11, 577590.
Mitchell, C.R., Harris, M.B., Cordaro, A.R., Starnes, J.W., 2002. Eect of
body temperature during exercise on skeletal muscle cytochrome c
oxidase content. J. Appl. Physiol. 93, 526530.
Mitsumoto, A., Takeuchi, A., Okawa, K., Nakagawa, Y., 2002. A subset
of newly synthesized polypeptides in mitochondria from human
endothelial cells exposed to hydroperoxide stress. Free Radic. Biol.
Med. 32, 2237.
Mizukoshi, E., Suzuki, M., Loupatov, A., Uruno, T., Hayashi, H.,
Misono, T., Kaul, S.C., Wadhwa, R., Imamura, T., 1999. Fibroblast
growth factor-1 interacts with the glucose-regulated protein GRP75/
mortalin. Biochem. J. 2, 461466.
Mizukoshi, E., Suzuki, M., Misono, T., Loupatov, A., Munekata, E.,
Kaul, S.C., Wadhwa, R., Imamura, T., 2001. Cell-cycle dependent
tyrosine phosphorylation on mortalin regulates its interaction with
broblast growth factor-1. Biochem. Biophys. Res. Commun. 280,
12031209.
Moro, F., Fernandez-Saiz, V., Muga, A., 2004. The lid subdomain of
DnaK is required for the stabilization of the substrate-binding site. J.
Biol. Chem. 279, 1960019606.
Moro, F., Fernandez-Saiz, V., Slutsky, O., Azem, A., Muga, A., 2005.
Conformational properties of bacterial DnaK and yeast mitochondrial
Hsp70. Role of the divergent C-terminal alpha-helical subdomain.
FEBS J. 272, 31843196.
Mosser, D.D., Morimoto, R.I., 2004. Molecular chaperones and the stress
of oncogenesis. Oncogene 23, 29072918.
Muranyi, M., He, Q.P., Fong, K.S., Li, P.A., 2005. Induction of heat
shock proteins by hyperglycemic cerebral ischemia. Brain Res. Mol.
Brain Res. 139, 8087.
Ohashi, M., Oyanagi, M., Hatakeyama, K., Inoue, M., Kominami, R.,
1995. The gene encoding PBP74/CSA/motalin-1, a novel mouse hsp70,
maps to mouse chromosome 18. Genomics 30, 406407.

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx
Orlov, S.N., Hamet, P., 2006. The death of cardiotonic steroid-treated
cells: evidence of Na+i, K+i-independent H+i-sensitive signalling.
Acta Physiol. (Oxf.) 187, 231240.
Ornatsky, O.I., Connor, M.K., Hood, D.A., 1995. Expression of stress
proteins and mitochondrial chaperonins in chronically stimulated
skeletal muscle. Biochem. J. 311, 119123.
Orsini, F., Migliaccio, E., Moroni, M., Contursi, C., Raker, V.A., Piccini,
D., Martin-Padura, L., Pelliccia, G., Trinei, M., Bono, M., Puri, C.,
Tacchetti, C., Ferrini, M., Mannucci, R., Nicoletti, I., Lanfrancone,
L., Giorgio, M., Pelicci, P.G., 2004. The life span determinant p66Shc
localizes to mitochondria where it associates with mitochondrial heat
shock protein 70 and regulates trans-membrane potential. J. Biol.
Chem. 279, 2568925695.
Pilzer, D., Fishelson, Z., 2005. Mortalin/GRP75 promotes release of
membrane vesicles from immune attacked cells and protection from
complement-mediated lysis. Int. Immunol. 17, 12391248.
Pilzer, D., Gasser, O., Moskovich, O., Schierli, J.A., Fishelson, Z., 2005.
Emission of membrane vesicles: roles in complement resistance,
immunity and cancer. Springer Semin. Immunopathol. 27, 375387.
Pizzatti, L., Sa, L.A., de Souza, J.M., Bisch, P.M., Abdelhay, E., 2006.
Altered protein prole in chronic myeloid leukemia chronic phase
identied by a comparative proteomic study. Biochim. Biophys. Acta
1764, 929942.
Poindexter, B.J., Pereira-Smith, O., Wadhwa, R., Buja, L.M., Bick, R.J.,
2002. 3D reconstruction and localization of mortalin by deconvolution
microscopy. Micros. Anal. 89, 2123.
Ran, Q., Wadhwa, R., Kawai, R., Kaul, S.C., Sifers, R.N., Bick, R.J.,
Smith, J.R., Pereira-Smith, O.M., 2000. Extramitochondrial localization of mortalin/mthsp70/PBP74/GRP75. Biochem. Biophys. Res.
Commun. 275, 174179.
Rehling, P., Brandner, K., Pfanner, N., 2004. Mitochondrial import and
the twin-pore translocase. Nat. Rev. Mol. Cell Biol. 5, 519530.
Rivolta, M.N., Holley, M.C., 2002. Asymmetric segregation of mitochondria and mortalin correlates with the multi-lineage potential of inner
ear sensory cell progenitors in vitro. Brain Res. Dev. Brain Res. 133,
4956.
Sacht, G., Brigelius-Flohe, R., Kiess, M., Sztajer, H., Flohe, L., 1999.
ATP-sensitive association of mortalin with the IL-1 receptor type I.
Biofactors 9, 4960.
Sadekova, S., Lehnert, S., Chow, T.Y., 1997. Induction of PBP74/
mortalin/Grp75, a member of the hsp70 family, by low doses of
ionizing radiation: a possible role in induced radioresistance. Int. J.
Radiat. Biol. 72, 653660.
Saretzki, G., Armstrong, L., Leake, A., Lako, M., von Zglinicki, T., 2004.
Stress defense in murine embryonic stem cells is superior to that of
various dierentiated murine cells. Stem Cells 22, 962971.
Schwarzer, C., Barnikol-Watanabe, S., Thinnes, F.P., Hilschmann, N.,
2002. Voltage-dependent anion-selective channel (VDAC) interacts
with the dynein light chain Tctex1 and the heat-shock protein PBP74.
Int. J. Biochem. Cell Biol. 34, 10591070.
Sichting, M., Mokranjac, D., Azem, A., Neupert, W., Hell, K.,
2005. Maintenance of structure and function of mitochondrial
Hsp70 chaperones requires the chaperone Hepl. EMBO J. 24,
10461056.
Singh, M., Dykens, J.A., Simpkins, J.W., 2006. Novel mechanisms for
estrogen-induced neuroprotection. Exp. Biol. Med. (Maywood) 231,
514521.
Soti, C., Csermely, P., 2000. Molecular chaperones and the aging process.
Biogerontology 1, 225233.
Soti, C., Csermely, P., 2002. Chaperones and aging: role in neurodegeneration and in other civilizational diseases. Neurochem. Int. 41, 383
389.
Soti, C., Sreedhar, A.S., Csermely, P., 2003. Apoptosis, necrosis and
cellular senescence: chaperone occupancy as a potential switch. Aging
Cell 2, 3945.
Sriram, M., Osipiuk, J., Freeman, B., Morimoto, R., Joachimiak, A.,
1997. Human Hsp70 molecular chaperone binds two calcium ions
within the ATPase domain. Structure 5, 403414.

11

Stacchiotti, A., Ricci, F., Rezzani, R., Li Volti, G., Borsani, E., Lavazza,
A., Bianchi, R., Rodella, L.F., 2006. Tubular stress proteins and nitric
oxide synthase expression in rat kidney exposed to mercuric chloride
and melatonin. J. Histochem. Cytochem. 54, 11491157.
Stevens, T.A., Meech, R., 2006. BARX2 and estrogen receptor-alpha
(ESR1) coordinately regulate the production of alternatively spliced
ESR1 isoforms and control breast cancer cell growth and invasion.
Oncogene. 25, 54265435.
Strub, A., Lim, J.H., Pfanner, N., Voos, W., 2000. The mitochondrial
protein import motor. Biol. Chem. 381, 943949.
Takano, S., Wadhwa, R., Yoshii, Y., Nose, T., Kaul, S.C., Mitsui, Y.,
1997. Elevated levels of mortalin expression in human brain tumors.
Exp. Cell Res. 237, 3845.
Takano, S., Wadhwa, R., Mitsui, Y., Kaul, S.C., 2001. Identication and
characterization of molecular interactions between glucose-regulated
proteins (GRPs) mortalin/GRP75/peptide-binding protein 74 (PBP74)
and GRP94. Biochem. J. 357, 393398.
Taurin, S., Seyrantepe, V., Orlov, S.N., Tremblay, T.L., Thibault, P.,
Bennett, M.R., Hamet, P., Pshezhetsky, A.V., 2002. Proteome analysis
and functional expression identify mortalin as an antiapoptotic gene
induced by elevation of [Na+]i/[K+]i ratio in cultured vascular smooth
muscle cells. Circ. Res. 91, 915922.
Tsuchiya, T., Dhahbi, J.M., Cui, X., Mote, P.L., Bartke, A., Spindler,
S.R., 2004. Additive regulation of hepatic gene expression by dwarsm
and caloric restriction. Physiol. Genomics 17, 307315.
Um, J.H., Kang, C.D., Hwang, B.W., Ha, M.Y., Hur, J.G., Kim, D.W.,
Chung, B.S., Kim, S.H., 2003a. Involvement of DNA-dependent
protein kinase in regulation of the mitochondrial heat shock proteins.
Leuk. Res. 27, 509516.
Um, J.H., Kim, S.J., Kim, D.W., Ha, M.Y., Jang, J.H., Chung, B.S.,
Kang, C.D., Kim, S.H., 2003b. Tissue-specic changes of DNA repair
protein Ku and mtHSP70 in aging rats and their retardation by caloric
restriction. Mech. Ageing Dev. 124, 967975.
Voos, W., Rottgers, K., 2002. Molecular chaperones as essential mediators of mitochondrial biogenesis. Biochim. Biophys. Acta 2, 5162.
Voos, W., von Ahsen, O., Muller, H., Guiard, B., Rassow, J., Pfanner, N.,
1996. Dierential requirement for the mitochondrial Hsp70Tim44
complex in unfolding and translocation of preproteins. EMBO J. 15,
26682677.
Wadhwa, R., Kaul, S.C., Ikawa, Y., Sugimoto, Y., 1993a. Identication of
a novel member of mouse hsp70 family. Its association with cellular
mortal phenotype. J. Biol. Chem. 268, 66156621.
Wadhwa, R., Kaul, S.C., Mitsui, Y., Sugimoto, Y., 1993b. Dierential
subcellular distribution of mortalin in mortal and immortal mouse and
human broblasts. Exp. Cell Res. 207, 442448.
Wadhwa, R., Kaul, S.C., Sugimoto, Y., Mitsui, Y., 1993c. Induction of
cellular senescence by transfection of cytosolic mortalin cDNA in NIH
3T3 cells. J. Biol. Chem. 268, 2223922242.
Wadhwa, R., Takano, S., Robert, M., Yoshida, A., Nomura, H., Reddel,
R.R., Mitsui, Y., Kaul, S.C., 1998. Inactivation of tumor suppressor
p53 by mot-2, a hsp70 family member. J. Biol. Chem. 273, 29586
29591.
Wadhwa, R., Kaul, S.C., Takano, S., Reddel, R.R., Mitsui, Y., Wadhwa,
R., 2000a. Transcriptional inactivation of p53 by deletions and single
amino acid changes in mouse mot-1 protein. Biochem. Biophys. Res.
Commun. 279, 602606.
Wadhwa, R., Sugihara, T., Yoshida, A., Nomura, H., Reddel, R.R.,
Simpson, R., Maruta, H., Kaul, S.C., 2000b. Selective toxicity of
MKT-077 to cancer cells is mediated by its binding to the hsp70 family
protein mot-2 and reactivation of p53 function. Cancer Res. 60, 6818
6821.
Wadhwa, R., Taira, K., Kaul, S.C., 2002a. An Hsp70 family chaperone,
mortalin/mthsp70/PBP74/Grp75: what, when, and where? Cell Stress
Chaperones 7, 309316.
Wadhwa, R., Yaguchi, T., Hasan, M.K., Mitsui, Y., Reddel, R.R., Kaul,
S.C., 2002b. Hsp70 family member, mot-2/mthsp70/GRP75, binds to
the cytoplasmic sequestration domain of the p53 protein. Exp. Cell
Res. 274, 246253.

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

ARTICLE IN PRESS
12

S.C. Kaul et al. / Experimental Gerontology xxx (2007) xxxxxx

Wadhwa, R., Ando, H., Kawasaki, H., Taira, K., Kaul, S.C., 2003a.
Targeting mortalin using conventional and RNA-helicase-coupled
hammerhead ribozymes. EMBO Rep. 4, 595601.
Wadhwa, R., Yaguchi, T., Hasan, M.K., Taira, K., Kaul, S.C., 2003b.
Mortalin-MPD (mevalonate pyrophosphate decarboxylase) interactions and their role in control of cellular proliferation. Biochem.
Biophys. Res. Commun. 302, 735742.
Wadhwa, R., Takano, S., Taira, K., Kaul, S.C., 2004. Reduction in
mortalin level by its antisense expression causes senescence-like growth
arrest in human immortalized cells. J. Gene Med. 6, 439444.
Wadhwa, R., Takano, S., Kaur, K., Aida, S., Yaguchi, T., Kaul, Z.,
Hirano, T., Taira, K., Kaul, S.C., 2005. Identication and characterization of molecular interactions between mortalin/mtHsp70 and
HSP60. Biochem. J. 391, 185190.
Wadhwa, R., Takano, S., Kaur, K., Deocaris, C.C., Pereira-Smith, O.M.,
Reddel, R.R., Kaul, S.C., 2006. Upregulation of mortalin/mthsp70/
Grp75 contributes to human carcinogenesis. Int. J. Cancer 118, 2973
2980.
Walker, C., Bottger, S., Low, B., 2006. Mortalin-based cytoplasmic
sequestration of p53 in a nonmammalian cancer model. Am. J. Pathol.
168, 15261530.

Wiedemann, N., Frazier, A.E., Pfanner, N., 2004. The protein import
machinery of mitochondria. J. Biol. Chem. 279, 1447314476.
Xie, H., Hu, Z., Chyna, B., Horrigan, S.K., Westbrook, C.A., 2000.
Human mortalin (HSPA9): a candidate for the myeloid leukemia
tumor suppressor gene on 5q31. Leukemia 14, 21282134.
Yamagishi, N., Ishihara, K., Hatayama, T., 2004. Hsp105alpha suppresses
Hsc70 chaperone activity by inhibiting Hsc70 ATPase activity. J. Biol.
Chem. 279, 4172741733.
Yamamoto, H., Momose, T., Yatsukawa, Y., Ohshima, C., Ishikawa, D.,
Sato, T., Tamura, Y., Ohwa, Y., Endo, T., 2005. Identication of a
novel member of yeast mitochondrial Hsp70-associated motor and
chaperone proteins that facilitates protein translocation across the
inner membrane. FEBS Lett. 579, 507511.
Yokoyama, K., Fukumoto, K., Murakami, T., Harada, S., Hosono, R.,
Wadhwa, R., Mitsui, Y., Ohkuma, S., 2002. Extended longevity of
Caenorhabditis elegans by knocking in extra copies of hsp70F, a
homolog of mot-2 (mortalin)/mthsp70/Grp75. FEBS Lett. 516, 5357.
Zhu, X., Zhao, X., Burkholder, W.F., Gragerov, A., Ogata, C.M.,
Gottesman, M.E., Hendrickson, W.A., 1996. Structural analysis of
substrate binding by the molecular chaperone DnaK. Science 272,
16061614.

Please cite this article in press as: Kaul, S.C. et al., Three faces of mortalin: A housekeeper, guardian and killer, Exp. Gerontol.
(2007), doi:10.1016/j.exger.2006.10.020

You might also like