You are on page 1of 12

Novel CDNF/MANF Family of Neurotrophic Factors

Päivi Lindholm, Mart Saarma


Institute of Biotechnology, Viikinkaari 9, Viikki Biocenter, University of Helsinki, 00014 Helsinki, Finland

Received 16 July 2009; revised 14 September 2009; accepted 21 September 2009

ABSTRACT: Current therapeutic interventions structurally conserved cysteine residues, which deter-
for neurodegenerative diseases alleviate only disease mine the protein fold. The crystal structure analysis
symptoms, while treatments that could stop or reverse revealed that CDNF and MANF consist of two domains;
actual degenerative processes are not available. Parkin- an amino-terminal saposin-like domain that may inter-
son’s disease (PD) is a movement disorder with charac- act with lipids or membranes, and a presumably
teristic degeneration of dopaminergic neurons in the unfolded carboxy-terminal domain that may protect
midbrain. Few neurotrophic factors (NTFs) that pro- cells against endoplasmic reticulum stress. CDNF and
mote survival, maintenance, and differentiation of MANF protect midbrain dopaminergic neurons and
affected brain neurons are considered as potential ther- restore motor function in 6-hydroxydopamine rat model
apeutic agents for the treatment of neurodegenerative of PD in vivo. In line, Drosophila MANF is needed for
diseases. Thus, it is important to search and study new the maintenance of dopaminergic neurites and dopa-
NTFs that could also be used in therapy. In this review, mine levels in the fly, suggesting that the function of
we discuss novel evolutionary conserved family of NTFs CDNF/MANF proteins is evolutionary conserved.
consisting of two members in the vertebrates, cerebral Future studies will reveal the receptors and mode of
dopamine neurotrophic factor (CDNF) and mesence- action of these novel factors, which are potential thera-
phalic astrocyte-derived neurotrophic factor (MANF). peutic proteins for the treatment of PD. ' 2010 Wiley Peri-
Invertebrates, including Drosophila and Caenorhabditis odicals, Inc. Develop Neurobiol 70: 360–371, 2010
have a single protein homologous to vertebrate CDNF/ Keywords: neurotrophic factors; receptors; dopamine
MANF. Characteristic feature of these proteins is eight neurons; Parkinson’s disease; lipids; saposins

INTRODUCTION NTF theory that is largely based on the studies on


nerve growth factor (NGF), the first growth factor
Neurotrophic factors (NTFs) are an important group and NTF identified (Levi-Montalcini, 1987), excess
of secreted proteins regulating the life and death of neurons compete of NTFs that are secreted by the tar-
neurons during development. Different NTFs affect get tissue in limited amounts. Only neurons that
specific sets of neuronal subpopulations, which are receive sufficient trophic support survive and main-
genetically competent to respond, i.e., express cog- tain synaptic contacts with the target tissue, whereas
nate receptors on their surface, and have appropriate other neurons die by apoptosis. NTFs also regulate
transcriptional programs. During developmental peri- migration, differentiation, and maturation of postmi-
ods of programmed cell death (PCD), NTFs prevent totic neuronal precursors and promote the regenera-
apoptosis and regulate the number of neurons inner- tion of neurons from injury (Huang and Reichardt,
vating the target tissue. According to the classical 2001; Airaksinen and Saarma, 2002). The role of
NTFs is well-established in the development of the
Correspondence to: M. Saarma (mart.saarma@helsinki.fi). peripheral nervous system (PNS). However, the func-
' 2010 Wiley Periodicals, Inc. tion of NTFs in the development and maintenance of
Published online 22 February 2010 in Wiley InterScience (www.
interscience.wiley.com). the central nervous system (CNS) is less clear. NTFs,
DOI 10.1002/dneu.20760 e.g., brain-derived neurotrophic factor (BDNF) are
360
Novel CDNF/MANF Family of NTFs 361

known to regulate synaptic plasticity in adult CNS GDNF and BDNF have been detected in the substan-
(Lu et al., 2005). NTFs have important roles also in tia nigra (SN) of PD patients (Mogi et al., 1999;
non-neuronal tissues, and regulate, e.g., angiogenesis Parain et al., 1999; Howells et al., 2000; Chauhan et
(Zacchigna et al., 2008), kidney development, and al., 2001). Thus, delivery of NTFs may help to restore
spermatogenesis (Airaksinen and Saarma, 2002; the NTF deficiency in the disease.
Andressoo and Saarma, 2008). None of the existing therapeutic strategies for PD
In this review, we focus on the novel CDNF/ can stop or even slow down the degeneration of dopa-
MANF family of NTFs (Petrova et al., 2003; Lind- minergic neurons or induce neuronal recovery. The
holm et al., 2007). Traditionally, there are three fami- available therapies are based on dopamine-replace-
lies of NTFs: Neurotrophins NGF, BDNF, neurotro- ment therapy using either L-DOPA (l-3,4-dihydroxy-
phin-3 (NT-3) and NT-4 (Huang and Reichardt, phenylalanine) or dopamine agonists. These treat-
2001; Lu et al., 2005); glial cell line-derived neuro- ments only alleviate the disease symptoms, usually
trophic factor (GDNF) family ligands (GFLs) GDNF, by correcting striatal dopamine neurotransmission.
neurturin (NRTN), artemin (ARTN), and persephin Two highly potent NTFs for dopaminergic neurons,
(PSPN) (Airaksinen and Saarma, 2002; Bespalov and GDNF and NRTN have been taken to clinical trials
Saarma, 2007); and neuropoietic cytokines (also on PD patients, but the results from these trials have
referred as interleukin-6 (IL-6) family) (Heinrich been quite modest so far. Although direct infusion of
et al., 2003; Bauer et al., 2007). Members of these GDNF protein into the putamen of PD patients led to
families either signal through transmembrane recep- significant clinical improvement (Gill et al., 2003;
tor tyrosine kinases or via kinases that interact with Patel et al., 2005; Slevin et al., 2005), a double
their receptors (Bespalov and Saarma, 2007). Neuro- blinded phase II study showed no clinical efficacy of
trophic activities are also shown by other trophic and GDNF (Lang et al., 2006). Since several patients gen-
growth factors, e.g., members of transforming growth erated antisera against GDNF (Lang et al., 2006) and
factor b, vascular endothelial growth factor (VEGF), at high doses GDNF induced cerebellar lesions in pri-
insulin-like growth factor, and fibroblast growth mates (Hovland et al., 2007), clinical trials with
factor (FGF) families (Grothe and Timmer, 2007; GDNF protein have currently been interrupted.
Zacchigna et al., 2008). Recently, a novel evolution- Recent NRTN gene therapy trials have given
ary conserved protein Meteorin with neurotrophic slightly more positive outcome. A press release on
activities was characterized (Nishino et al., 2004; May 2009 by Ceregene on phase II study reported
Jørgensen et al., 2009). small positive effects on PD patients after 18 months
The question whether NTFs function in the devel- of treatment with CERE-120, the adeno-associated
opment of invertebrate nervous system has been virus serotype 2 vector carrying the gene for NRTN.
uncertain. However, a recent study described Dro- In any case, the development of NTF therapy is still
sophila Neurotrophin (DNT1; spätzle 2) a small part at a very early stage. Therefore, in addition to the
of which is structurally homologous to all known development of more efficient NTF delivery systems
neurotrophins (Zhu et al., 2008). Although GFL it is of great importance to search for and study novel
homologs have not been found in insect genomes NTFs that could offer new therapeutic approaches for
(Airaksinen et al., 2006), an orphan homolog for the treatment of neurodegenerative diseases.
GDNF transmembrane receptor tyrosine kinase RET The focus of this review is on the novel CDNF/
(rearranged during transfection) is present in Dro- MANF family of NTFs that are structurally unrelated
sophila (Sugaya et al., 1994). As we will discuss later to the classical NTF families, suggesting a
in more detail, Drosophila MANF (DmMANF) func- completely new mechanism of action and a novel
tions as a NTF for dopaminergic neurons in the fly therapeutic potential for the treatment of neurodege-
(Palgi et al., 2009). Thus, it seems now evident that nerative diseases.
NTFs have important roles also in the invertebrate
nervous system.
Since NTFs have many beneficial effects on neu-
rons, they are potential factors not only for the treat- CDNF AND MANF LIGANDS
ment of neurodegenerative diseases, including Par-
kinson’s disease (PD), Alzheimer’s disease (AD), Cerebral dopamine neurotrophic factor (CDNF; offi-
amyotrophic lateral sclerosis (ALS), but also for the cial name by HUGO gene nomenclature committee;
treatment of neuronal trauma, such as spinal previous name conserved dopamine NTF) and
cord injury. Shortage of NTFs is also seen in mesencephalic astrocyte-derived neurotrophic factor
neurodegenerative diseases, e.g., decreased levels of (MANF) form a novel evolutionary conserved protein
Developmental Neurobiology
362 Lindholm and Saarma

family showing neurotrophic activities (Petrova et al., acid identity with D. melanogaster and 50% identity
2003; Lindholm et al., 2007, 2008; Airavaara et al., with C. elegans MANF proteins, respectively. Differ-
2009; Palgi et al., 2009; Voutilainen et al., 2009). ently from neurotrophins and GDNF family ligands,
The founding member of the family, MANF, was CDNF and MANF proteins seem not to contain a pro-
identified from the culture medium of rat Type-1 sequence, suggesting that enzymatic cleavage is not
astrocyte ventral mesencephalic cell line 1 (VMCL1) necessary for their activity (Fig. 1A).
based on its survival promoting activities on cultured Consistently with the roles as extracellular NTFs,
embryonic dopaminergic neurons (Petrova et al., CDNF and MANF are secreted from transiently trans-
2003). The active rat protein was isolated and found fected cells (Lindholm et al., 2007, 2008; Palgi et al.,
homologous to a predicted human arginine-rich pro- 2009). Also the secretion of endogenous MANF has
tein (ARP) of 234 amino acids (aa; GenBank Acc. been demonstrated in vitro (Apostolou et al., 2008).
No. NP_006001) (Shridhar et al., 1996b; Petrova et Human CDNF contains one potential N-linked glyco-
al., 2003), which had never been expressed or purified sylation site, and both unglycosylated (Lindholm et
in vitro. Human ARP (ARMET; arginine-rich, al., 2007) and glycosylated (Apostolou et al., 2008)
mutated in early stage tumors) gene contains an form of human CDNF is secreted from transiently
amino-terminal arginine-rich region and mutations overexpressing cells. The nonglycosylated CDNF is
within were associated with different cancer types biologically active (Lindholm et al., 2007), indicating
(Shridhar et al., 1996a,b, 1997), although contrasting that glycosylation is not crucial for its neurotrophic
studies showed that variations in the arginine-rich activity. Differently from human CDNF, mouse
region of ARP represent normal polymorphisms CDNF, or human and mouse MANF do not have
(Evron et al., 1997; Tanaka et al., 2000; Piepoli et al., potential N- or O-linked glycosylation sites. In con-
2006). In line with that, the arginine-rich region of trast to the original report (Petrova et al., 2003),
ARP is apparently not translated in vivo (Petrova et secreted glycosylated MANF has not been detected in
al., 2003). Thus, the protein encoded by ARP was later studies (Apostolou et al., 2008; Lindholm et al.,
renamed MANF to better indicate its neurotrophic 2008). Both CDNF and MANF are highly soluble and
functions (Petrova et al., 2003). Human MANF to- monomeric at neutral pH 7 (Lindholm et al., 2007;
gether with the 21 amino acids long signal sequence Mizobuchi et al., 2007). Whether the secretion of en-
consists of 179 aa that equal amino acids 56–234 of dogenous CDNF or MANF is regulated by physiolog-
human ARP (Petrova et al., 2003) [Fig. 1(A)]. The ical stimuli or injury in vivo is currently unclear.
signal sequence is cleaved off cotranslationally Although secreted, MANF proteins are also par-
resulting into the mature MANF protein of 158 amino tially retained in the endoplasmic reticulum (ER)
acids. (Mizobuchi et al., 2007; Apostolou et al., 2008).
Mature CDNF (ARMETL1; GenBank Acc. No. MANF and CDNF have a C-terminal sequence
NM_177647) protein consists of 161 amino acid resi- (RTDL and KTEL, respectively) closely resembling
dues (preCDNF is 187 amino acids long), and is a the classical ER retention signal (KDEL), which may
vertebrate specific paralog of MANF. CDNF was first function as a partial ER retention signal (Raykhel et
identified by bioinformatics and then biochemically al., 2007; Apostolou et al., 2008). Interestingly,
characterized (Lindholm et al., 2007). CDNF and CDNF and MANF may have important functions also
MANF encoding genes are highly conserved in evo- in the ER, as discussed below.
lution. Based on sequence analyses, vertebrates have
CDNF and MANF genes (Lindholm et al., 2007),
whereas invertebrates, including C. elegans and TWO DOMAINS—TWO ACTIVITIES?
D. melanogaster, have a single homologous gene
more closely related to MANF than to CDNF (Palgi The structure of mammalian MANF and CDNF pro-
et al., 2009). Characteristic feature of CDNF and teins is unique among NTFs and growth factors, and
MANF proteins is eight cysteine residues, spacing of defines a novel type of proteins [Fig. 1(B)]. The crys-
which is conserved from vertebrates to invertebrates tal structure of the mature human MANF protein and
(Shridhar et al., 1996b; Petrova et al., 2003; Lind- the amino (N)-terminal domain of human CDNF
holm et al., 2007). DmMANF of 151 aa residues (pre- have been resolved (Parkash et al., 2009). The struc-
DmMANF is 173 aa) has been recently characterized ture of CDNF and MANF consists of two domains, a
(Palgi et al., 2009). Human CDNF shows 59% amino saposin-like N-terminal domain and a presumably
acid identity with human MANF, 49% identity with unstructured carboxy (C)-terminal domain with an
D. melanogaster, and 46% identity with C. elegans intradomain cysteine bridge in a CXXC motif. Inter-
MANF proteins. Human MANF shows 53% amino estingly, saposin-like proteins interact with lipids
Developmental Neurobiology
Novel CDNF/MANF Family of NTFs 363

are saposin-like proteins (SAPLIPs) granulysin


(Anderson et al., 2003) and NK-lysin (Liepinsh et al.,
1997) [Fig. 1(C)], which function as defense proteins
against bacterial cells and are able to disrupt target
cell membranes (Jongstra et al., 1987; Andersson et
al., 1995; Pena et al., 1997). Granulysin and NK-lysin
are proteolytically processed from propolypeptides in
cytolytic granules. In contrast, CDNF and MANF are
likely functional upon synthesis and secretion since
they apparently do not contain prosequences for
enzymatic activation. However, it is possible to
speculate the N-terminal domain represents the pro-
domain of CDNF/MANF family proteins and then
proteolytic cleavage in the linker region produces the
\mature" protein, i.e., the C-terminal domain.
Characteristic features of the membrane interac-
tions of SAPLIPs are oligomerization and conforma-
tional changes. Also the composition of membrane
lipids and pH value are known to affect membrane
binding. Interestingly, the saposin-like domain of

Figure 1 (A) Schematic presentation of the primary struc-


tures of human CDNF and MANF, and selected invertebrate
MANF proteins. Pre represents the signal sequence and the
numbers the length of the polypeptides in amino acids.
Yellow vertical bars show the conserved cysteine residues.
(B) Crystal structure of mature human MANF. Alpha-helices
(a) are numbered starting from the N-terminus. Below the
structural image is a schematic picture of the primary struc-
ture of mature MANF. The formation of cysteine bridges is
shown as connecting lines. (C) Structure of the N-terminal
domain of human MANF (blue) is superimposed on human
granulysin saposin (pink).

and/or membranes (Bruhn, 2005) suggesting that the


N-terminal domain of CDNF and MANF may also
interact with lipids. Furthermore, the C-terminal cys- Figure 2 Hypothetical modes of CDNF and MANF
action. (A) Change in pH or lipid binding may induce dime-
teine bridge of MANF is similar to the active site
rization of CDNF or MANF. (B) C-terminal domain of
motif of thiol/disulfide oxidoreductases that catalyze CDNF or MANF may facilitate the formation of disulfide
the formation of intramolecular disulphide bonds, bridges on secretory proteins in the ER. (C) CDNF or
suggesting that CDNF and MANF maybe involved in MANF may reduce disulfide bond on a transmembrane
protein folding in the ER. receptor protein on the cell surface. (D) CDNF or MANF
The closest structural homologs for CDNF and may interact with the cell surface membrane and subse-
MANF N-terminal domain (Parkash et al., 2009) quently activate transmembrane receptor protein.

Developmental Neurobiology
364 Lindholm and Saarma

CDNF crystallized as a dimer at pH 4.6, whereas the erties similar with those of saposins is also not
full-length, mature MANF crystallized as a monomer known. Hypothetical modes of CDNF and MANF
at neutral pH (Parkash et al., 2009). Saposin C and dimerization and membrane interaction are presented
Saposin A form oligomers in the presence of a deter- in Figure 2.
gent at low pH 4.8 (Ahn et al., 2006). Acidic pH also
increases the ability of Saposin C and Saposin D to
bind lipid vesicles and destabilize phospholipid-con- PROTECTION AGAINST ER STRESS
taining membranes (Vaccaro et al., 1995). Differently
from acidic saposins with pI values 4.6–4.8, CDNF Accumulating evidence suggests that MANF is an
and MANF SAPLIPs are slightly basic with pI values ER stress response protein (Lee et al., 2003; Mizobu-
7.7 and 8.5, respectively. It is likely that pH affects chi et al., 2007; Apostolou et al., 2008; Tadimalla et
the putative oligomerization [Fig. 2(A)] and mem- al., 2008) and is able to protect cells against ER
brane interactions of CDNF and MANF. stress-induced cell death in vitro (Apostolou et al.,
Hydrophobic residues located at conserved posi- 2008). In line, the crystal structure suggests that
tions of SAPLIPs form a hydrophobic core and may MANF and CDNF may help protein folding in the
have a role in the lipid interactions (Bruhn, 2005). ER. In the MANF C-terminal domain, the two cys-
Based on the structural analysis, the saposin-like teines in 127CKGC130 motif (132CRAC135 in CDNF)
domain of CDNF and MANF is in the closed confor- form a C-terminal disulfide bridge. Interestingly, a
mation and stabilized by three intramolecular disul- CXXC active site motif is found in thiol/disulfide
fide bonds, which connect a1 and C-terminal 310 oxidoreductases that catalyze the formation of intra-
helix, a1 and a5 helix, a2 and a3 helix, respectively molecular disulfide bonds. These proteins include thi-
(Parkash et al., 2009) [Fig. 1(B)]. Saposin B, a pro- oredoxins, like protein disulphide isomerases (PDIs),
tein with lipid transfer activity (Vogel et al., 1991), which function in the ER (Ellgaard and Ruddock,
crystallized in an open conformation as a homodimer 2005). Interestingly, nitrosylated PDI is found in
with bound phospholipid (Ahn et al., 2003), whereas brain samples derived from PD patients, indicating a
Saposin C crystallized in the presence of detergent as link between PDI dysfunction and protein misfolding
a monomer in an open conformation with hydropho- in neurodegenerative disorders (Uehara et al., 2006).
bic pocket exposed to solvent (Hawkins et al., 2005). MANF and CDNF may facilitate the formation of
Also at pH 4, Saposin C crystallized in an open con- cysteine bridges and protein folding in the ER
formation, but as a domain-swapped dimer that may [Fig. 2(B)], thus reducing the ER stress caused by
facilitate vesicle fusion (Rossmann et al., 2008). unfolded or incorrectly folded proteins.
Charge distribution on the surface of SAPLIPs Accumulation of unfolded proteins in the ER
may have an important role in the membrane target- causes ER stress and the induction of unfolded pro-
ing. NK-lysin (Liepinsh et al., 1997) and granulysin tein response (UPR). This is a signal-transduction
(Anderson et al., 2003) have a ring of positively pathway that counteracts ER stress (Szegezdi et al.,
charged residues around the protein, which may 2006b) by increasing protein folding capacity and
direct their interaction toward the negatively charged degradation of misfolded proteins in the cell. Also
lipid head groups in the membrane. The saposin-like protein translocation across the ER membrane is
domain of CDNF and MANF has conserved posi- reduced. Proteins are refolded by molecular chaper-
tively charged residues on the surface in two patches ones and misfolded proteins are removed by ER asso-
that may contribute to the membrane interactions ciated protein degradation. ER transmembrane recep-
(Parkash et al., 2009). tors mediate the UPR pathways, including receptors
We have recently confirmed the lipid interaction activating transcription factor 6 (ATF6), inositol-
of CDNF and MANF (Hongxia Zhao, Pekka Lappa- requiring enzyme 1 (IRE1), and pancreatic ER kinase
lainen and Mart Saarma, unpublished observations). (PKR)-like ER kinase (PERK) (Szegezdi et al.,
The role of lipids and membrane interactions in the 2006b). The expression of UPR target genes is also
biology of CDNF and MANF are completely regulated by the transcription factor X-box binding
unknown. Does lipid binding induce dimerization of protein 1 (XBP1) (Szegezdi et al., 2006b).
CDNF and MANF [Fig. 2(A)] and subsequent activa- Upregulation of Manf by ER stress conditions has
tion? It does not seem likely that CDNF and MANF been shown by microarray analyses (Lee et al., 2003;
disrupt the target membranes like granulysin and Mizobuchi et al., 2007; Belmont et al., 2008), sug-
NK-lysin, since cell death has not been observed gesting that Manf has a role in the quality control of
when cells were treated with CDNF or MANF. proteins during ER stress (Lee et al., 2003). Manf was
Whether CDNF and MANF have lipid transfer prop- also upregulated in cell lines by different UPR
Developmental Neurobiology
Novel CDNF/MANF Family of NTFs 365

inducers (Apostolou et al., 2008). Ischemia is known MANF protein (Mizobuchi et al., 2007), but further
to induce ER stress response (Szegezdi et al., 2006a). analyses are definitely needed.
In line, hypoxia induced upregulation of Manf in cell
lines in vitro (Romero-Ramirez et al., 2004), and
myocardial infarction upregulated Manf in vivo CDNF AND MANF ACTIVITIES IN VIVO
(Harpster et al., 2006; Tadimalla et al., 2008). Knock-
down of Armet expression by small interfering RNA In addition to the highly potent factors GDNF and
(siRNA) increased the susceptibility of cultured cells NRTN, which have been extensively studied using
to tunicamycin-induced death, whereas overexpres- toxin-induced rodent and primate models of PD, few
sion of Armet was protective supporting the role for trophic factors and growth factors, e.g., EPO (Pus-
ARMET/MANF as a protective protein against ER kovic et al., 2006; Kadota et al., 2009), VEGF family
stress-induced cell death (Apostolou et al., 2008). members (Yasuhara et al., 2005), and FGF family
Furthermore, recombinant MANF protein protected members (Fontan et al., 2002) induce the recovery of
cardiac myocytes from ischemia-induced death in dopaminergic function in vivo. MANF was discov-
vitro (Tadimalla et al., 2008). In line, MANF protein ered based on its survival promoting activity on mid-
protects against ischemic brain injury induced by brain dopaminergic neurons in vitro (Petrova et al.,
middle cerebral artery occlusion (MCAO) in rats in 2003). Since the activity of MANF or CDNF in vivo
vivo (Airavaara et al., 2009). The ER stress response on dopaminergic neurons had not been studied, we
element-II (ERSE-II) is located in the Armet pro- used a rat 6-OHDA model of PD to test their activ-
moter and is shown to regulate Armet expression ities on dopaminergic neurons (Lindholm et al., 2007,
(Mizobuchi et al., 2007). In contrast to Manf, Cdnf Voutilainen et al., 2009). The unilateral 6-hydroxydo-
(Armetl1) expression was not upregulated by ER- pamine (6-OHDA) rat model of PD is commonly
stressors in vitro, suggesting that CDNF acts constitu- used for preclinical studies of novel potential thera-
tively in the ER (Apostolou et al., 2008). ER stress peutic agents. Striatal 6-OHDA injection leads to pro-
had only a minor effect on the secretion of MANF tracted retrograde degeneration of the nigrostriatal
and CDNF proteins in vitro (Apostolou et al., 2008), pathway (Sauer and Oertel, 1994) [Fig. 3(A)]. When
suggesting that they have a cell-autonomous role in CDNF (Lindholm et al., 2007) or MANF (Vouti-
the ER stress. lainen et al., 2009) were injected into the striatum
Interestingly, the reduction and rearrangement of before striatal 6-OHDA, both factors were able to
disulfide bonds is a mechanism of controlling protein significantly reduce amphetamine-induced rotational
function on the cell surface (Hogg, 2003). Cell adhe- behavior in rats and protect tyrosine hydroxylase
sion, uptake of bacterial toxins and viral fusion with a (TH) positive dopaminergic cell bodies in the SN and
host cell membrane may depend on thiol-disulfide TH-positive fibers in the striatum [Fig. 3(B)]. Impor-
exchange reactions (Hogg, 2003). Oxidoreductases tantly, CDNF and MANF also showed prominent
are secreted and may associate on cell surfaces neurorestorative effects. When either CDNF or
(Jordan and Gibbins, 2006). Extracellular redox cata- MANF was given into the striatum 4 weeks after a
lysts have been shown to regulate the functional striatal 6-OHDA lesion, we observed recovery of
forms of cell surface proteins as well as receptor– motor function in the rats, suggesting that CDNF and
ligand interactions. For example, thioredoxin 1 MANF prevented further degeneration of dopaminer-
(TRX1) mediates disulfide reduction in CD30 (tumor gic neurons and maintained or increased the function-
necrosis factor receptor superfamily member 8; ality of remaining neurons [Fig. 3(C,D)]. Thus, we
TNFRS8), which induces a subsequent conforma- found that CDNF and MANF very efficiently stopped
tional change and affects functional properties of the the further death of degenerating dopamine neurons.
CD30 ectodomain thus regulating inflammatory Presumably they also induced dopaminergic neurite
response (Schwertassek et al., 2007). Extracellular growth and axonal sprouting. GDNF induces sprout-
PDI is known to mediate entry of human immunode- ing of striatal dopaminergic fibers and upregulation
ficiency virus 1 (HIV-1) into lymphoid cells by cata- of TH expression (Hoffer et al., 1994; Hudson et al.,
lyzing redox changes of the HIV-1 envelope glyco- 1995). Further studies are needed to reveal the effects
protein gp120 (Hogg, 2003). Whether extracellular or of CDNF and MANF on dopaminergic neurites and
cell-surface bound CDNF and MANF proteins regu- TH levels in vivo.
late reduction or oxidation of disulfide bonds on cell Effects of recombinant MANF protein have also
surface proteins, e.g., transmembrane receptor been studied in an experimental model of stroke in
[Fig. 2(C)] is not known. The first studies did not rats (Airavaara et al., 2009). Administration of
detect oxidoreductase activity or bound metal ions in MANF into the cerebral cortex before MCAO was
Developmental Neurobiology
366 Lindholm and Saarma

Figure 3 CDNF and MANF activities in vivo. (A) Unilateral 6-OHDA model of PD. 6-OHDA is
injected (arrow) into the striatum (STR) resulting in a gradual degeneration of nigro-striatal dopaminer-
gic pathway with cell bodies located in the substantia nigra (SN). (B) GDNF and CDNF were able to
protect striatal TH+ fibers (in brown) against unilateral 6-OHDA injection (arrow). Note that the right
hemisphere served as the control. (C, D) CDNF (C) and MANF (D) can restore the motor function in
6-OHDA-lesioned rats. Animals were first injected with 6-OHDA and 4 weeks later with CDNF or
MANF, and GDNF as the control. Striatal injection of CDNF, MANF, and GDNF reduced ampheta-
mine-induced ipsilateral rotational behavior as compared with the control group with maximum effect
measured at 12 weeks postlesion. (E) MANF protein diffuses in the brain more efficiently than GDNF.
(B) and (C) were reproduced with permission of Nature Publishing Group from Lindholm et al. (2007).
(D) was reproduced with permission of the Society for Neuroscience from Voutilainen et al. (2009).
(E) was kindly provided by Merja H. Voutilainen, Susanne Bäck, and Raimo K. Tuominen.

able to significantly reduce the infarction volume and MANF protein added to the culture medium of car-
reduce markers of apoptotic cell death in the ischemic diac myocytes prevented cell death induced by simu-
cortex (Airavaara et al., 2009). In support to that lated ischemia in vitro (Tadimalla et al., 2008). Manf
Developmental Neurobiology
Novel CDNF/MANF Family of NTFs 367

was transiently upregulated in adult rat brain after dorsal root ganglion (DRG) neurons, or early post-
status epilepticus (SE) and global forebrain ischemia natal sympathetic cervical ganglion (SCG) neurons
in vivo (Lindholm et al., 2008), suggesting that (Lindholm et al., 2007) differently from GDNF that
MANF may regulate neuronal survival and synaptic supports the survival of different neuronal popula-
plasticity in the cortex and hippocampus. tions including motoneurons and enteric, sensory,
Studies with Drosophila MANF-deficient flies parasympathetic, and sympathetic neurons (Airak-
indicate that DmMANF, which is expressed in the sinen and Saarma, 2002). Similar to the results
glia, functions as a NTF in the fly (Palgi et al., 2009). obtained with CDNF were also reported for the
The genomic-null flies die in early larval stage, and MANF protein, although MANF showed a small sur-
mutant embryos show axonal degeneration, cuticular vival promoting effect on DRG neurons (Petrova et
defects, and nonapoptotic cell death. Importantly, al., 2003). Additional studies using, e.g., in vitro
DmMANF is needed for the maintenance of dopami- organotypic cultures or in vivo models of neurode-
nergic neurites and dopamine levels, whereas moto- generation, including models of motoneuron degener-
neurons and serotonergic (5-HT) neurons remained ation (e.g., ALS models), models of Huntington’s dis-
unaffected (Palgi et al., 2009). Human MANF is able ease with striatal neurodegeneration, or models of
to rescue the fly DmMANF knockout phenotype AD are essential to reveal potential neuronal targets
(Palgi et al., 2009), suggesting evolutionary con- of CDNF and MANF.
served function for MANF protein in the vertebrates
and invertebrates.
RECEPTOR IS UNKNOWN

NOVEL NEUROTROPHIC FACTORS FOR Differently from GDNF and NRTN, the target mole-
DOPAMINERGIC NEURONS cules, i.e., putative receptor(s) and signaling path-
ways activated by CDNF and MANF are still
Although extensively studied, the role of NTFs in the unknown. Whether CDNF and MANF use the same
target-innervation of dopaminergic neurons has signaling receptors is also unclear. 6-OHDA is known
remained unclear (Andressoo and Saarma, 2008). to induce oxidative stress, inhibition of mitochondrial
Potential target-derived factors for midbrain dopami- respiratory chain complex I, apoptotic cell death, and
nergic neurons include GDNF, NRTN, BDNF, NT-4, inflammation (Schober, 2004). CDNF and MANF
and FGF2 (Krieglstein, 2004). presumably activate signaling pathways in cells that
Cdnf and Manf are expressed in the developing counteract some of these phenomena. Interestingly,
nigro-striatal system of the mouse at P1 and P10 in vitro studies have shown that ER stress and UPR
(Lindholm et al., 2007, 2008), that is around the two are also related to 6-OHDA toxicity (Silva et al.,
postnatal PCD peaks of midbrain dopaminergic neu- 2005). Could extracellularly delivered CDNF and
rons at P2 and P14 (Burke, 2004), suggesting that MANF inhibit ER stress induced by 6-OHDA in the
CDNF and MANF may have a role in the develop- brain tissue in vivo? 6-OHDA was shown to induce
ment of midbrain dopaminergic system. In the adult upregulation of Manf expression in dopaminergic
mouse, MANF protein was also localized in TH- MN9D cells in culture (Holtz et al., 2005). Whether
positive dopaminergic neurons in the SN (Lindholm 6-OHDA treatment affects to endogenous levels of
et al., 2008). Whether MANF or CDNF function as CDNF or MANF expression in rat brain in vivo is
target-derived NTFs for midbrain dopaminergic neu- under investigation.
rons is currently unknown. Future studies on CDNF- Whether the neurotrophic activities of CDNF and
and MANF-deficient mice should reveal the develop- MANF are mediated by the saposin-like domain, the
mental role of these new NTFs. The widespread C-terminal domain or whether both domains are
expression of CDNF and MANF (Lindholm et al., essential is also unknown. Neurotrophic activities of
2007, 2008) suggests that endogenous CDNF and SAPLIPs, e.g., prosaposin, saposin C, and a peptide
MANF secreted by other sources than the striatal tar- derived from them have been reported in vitro
get may affect maturation of rodent dopaminergic (O’Brien et al., 1994, 1995) and in vivo (Kotani et al.,
system in vivo. 1996; Liu et al., 2001), suggesting that the neurotro-
Whether CDNF and MANF affect PNS neurons or phic activity may reside in the N-terminal domain of
other neuronal types in addition to dopaminergic neu- CDNF and MANF (Parkash et al., 2009). It is also
rons and cortical neurons in the CNS in vivo is possible that the neurotrophic activities and cytopro-
unknown. In vitro, CDNF protein was unable to tection against ER stress are mediated by the same
promote the survival of embryonic motoneurons or structural motif. Whether the extracellular CDNF and
Developmental Neurobiology
368 Lindholm and Saarma

MANF is able to activate a transmembrane protein MANF is slightly more basic than CDNF, we assume
receptor, e.g., through interaction with lipids, and that CDNF should diffuse even better than MANF in
induce intracellular survival promoting signaling the brain tissue. The efficient diffusion of MANF may
[Fig. 2(D)] is unclear. Another possibility is that be of critical importance for its possible clinical use,
extracellular CDNF and MANF proteins are endocy- since growth factors delivered to human brain have to
tosed and subsequently able to induce neurotrophic/ diffuse significant distances to reach all target cells.
cytoprotective effects inside the cell. We definitely Indeed, the low benefit of GDNF and NRTN observed
need additional studies to reveal the mechanisms of in the recent clinical trials of PD may at least partially
CDNF and MANF neurotrophic and cytoprotective result from their limited tissue distribution.
activities at molecular level and to understand how or Although CDNF and MANF have potent effects
if these activities are related. on adult midbrain dopaminergic system (Lindholm et
al., 2007, Voutilainen et al., 2009) and cortical neu-
rons in the ischemia model (Airavaara et al., 2009), it
POTENTIAL THERAPEUTIC PROTEINS is currently unknown which other CNS or PNS neuro-
nal cell types or non-neuronal cells CDNF and
The development of NTF therapy for neurodegenera- MANF may affect in vivo. GDNF was originally
tive diseases is at an early stage. Several growth fac- introduced as a specific NTF for midbrain dopaminer-
tors have been reported as the survival factors for gic neurons (Lin et al., 1993), but it is now clear that
midbrain dopaminergic neurons, but only GDNF and the claim of specificity is not valid. Further preclini-
NRTN have well-established neurorestorative effects cal studies are needed to assess the potency of CDNF
in preclinical models of PD. Direct infusion of GDNF and MANF as therapeutic agents for PD and other
into the putamen of PD patients led to significant neurodegenerative diseases, and their possible side
clinical improvement without serious side-effects effects.
(Gill et al., 2003; Patel et al., 2005; Slevin et al., The biology of novel CDNF/MANF proteins is
2005) and induced sprouting of dopaminergic fibers starting to be revealed. Gene ablation studies will pre-
(Love et al., 2005). Although the results of these sumably elucidate the roles of CDNF and MANF
phase I studies were encouraging, a double blinded in vivo. It will be exciting to see what are the molecu-
phase II study on PD patients showed low clinical ef- lar interactions and signaling pathways related to the
ficacy of GDNF, and the formation of GDNF function potential neurotrophic and cytoprotective activities of
blocking antibodies (Lang et al., 2006). Thus, GDNF CDNF and MANF.
was withdrawn from clinic by Amgen.
Recent press release on May 27, 2009 by Ceregene The authors thank Dr. Veli-Matti Leppänen and Vimal
reported clinically modest positive effects of gene Parkash for providing the original structural images. They
are indebted to Dr. Raimo K. Tuominen, Susanne Bäck,
therapy by NRTN (CERE-120) on PD patients after
and Merja H. Voutilainen for making available unpublished
18 months of treatment. Neurturin expression by data.
CERE-120 was detected in the targeted putamen, but
no evidence was provided for the retrograde axonal
transport of NRTN to the dopaminergic cell bodies in REFERENCES
the SN, evidently reducing bioactivity of the factor.
Diffusion of GDNF and NRTN are known to be lim- Ahn VE, Faull KF, Whitelegge JP, Fluharty AL, Prive GG.
ited in brain tissue due to the binding to the heparan 2003. Crystal structure of saposin B reveals a dimeric
sulfate proteoglycans (Bespalov and Saarma, 2007). shell for lipid binding. Proc Natl Acad Sci USA 100:38–
It is important to search and study novel NTFs for 43.
midbrain dopaminergic neurons that might give new Ahn VE, Leyko P, Alattia JR, Chen L, Prive GG. 2006.
therapeutic approaches in the future for the treatment Crystal structures of saposins A and C. Protein Sci 15:
of neurodegenerative diseases. CDNF and MANF are 1849–1857.
definitely potential novel proteins for the treatment of Airaksinen MS, Holm L, Hätinen T. 2006. Evolution of the
GDNF family ligands and receptors. Brain Behav Evol
PD, since in animal models of PD they can protect
68:181–190.
and repair very efficiently the nigro-striatal dopami-
Airaksinen MS, Saarma M. 2002. The GDNF family: Sig-
nergic neurons. An important additional aspect is that nalling, biological functions and therapeutic value. Nat
MANF readily diffuses in the brain (Voutilainen et Rev Neurosci 3:383–394.
al., 2009), and even more efficiently than GDNF Airavaara M, Shen H, Kuo CC, Peränen J, Saarma M,
[data obtained by Merja H. Voutilainen, Susanne Hoffer B, Wang Y. 2009. Mesencephalic astrocyte-
Bäck, and Raimo K. Tuominen; Fig. 3(E)]. Since derived neurotrophic factor reduces ischemic brain injury
Developmental Neurobiology
Novel CDNF/MANF Family of NTFs 369

and promotes behavioral recovery in rats. J Comp Neurol Harpster MH, Bandyopadhyay S, Thomas DP, Ivanov PS,
515:116–124. Keele JA, Pineguina N, Gao B, et al. 2006. Earliest
Anderson DH, Sawaya MR, Cascio D, Ernst W, Modlin R, changes in the left ventricular transcriptome postmyocar-
Krensky A, Eisenberg D. 2003. Granulysin crystal struc- dial infarction. Mamm Genome 17:701–715.
ture and a structure-derived lytic mechanism. J Mol Biol Hawkins CA, de Alba E, Tjandra N. 2005. Solution struc-
325:355–365. ture of human saposin C in a detergent environment.
Andersson M, Gunne H, Agerberth B, Boman A, Bergman J Mol Biol 346:1381–1392.
T, Sillard R, Jornvall H, et al. 1995. NK-lysin, a novel Heinrich PC, Behrmann I, Haan S, Hermanns HM, Muller-
effector peptide of cytotoxic T and NK cells. Structure Newen G, Schaper F. 2003. Principles of interleukin
and cDNA cloning of the porcine form, induction by (IL)-6-type cytokine signalling and its regulation. Bio-
interleukin 2, antibacterial and antitumour activity. chem J 374:1–20.
EMBO J 14:1615–1625. Hoffer BJ, Hoffman A, Bowenkamp K, Huettl P, Hudson J,
Andressoo JO, Saarma M. 2008. Signalling mechanisms Martin D, Lin LF, et al. 1994. Glial cell line-derived neuro-
underlying development and maintenance of dopamine trophic factor reverses toxin-induced injury to midbrain do-
neurons. Curr Opin Neurobiol 18:297–306. paminergic neurons in vivo. Neurosci Lett 182:107–111.
Apostolou A, Shen Y, Liang Y, Luo J, Fang S. 2008. Hogg PJ. 2003. Disulfide bonds as switches for protein
Armet, a UPR-upregulated protein, inhibits cell prolifera- function. Trends Biochem Sci 28:210–214.
tion and ER stress-induced cell death. Exp Cell Res 314: Holtz WA, Turetzky JM, O’Malley KL. 2005. Microarray
2454–2467. expression profiling identifies early signaling transcripts
Bauer S, Kerr BJ, Patterson PH. 2007. The neuropoietic associated with 6-OHDA-induced dopaminergic cell
cytokine family in development, plasticity, disease and death. Antioxid Redox Signal 7:639–648.
injury. Nat Rev Neurosci 8:221–232. Hovland DN Jr, Boyd RB, Butt MT, Engelhardt JA, Mox-
Belmont PJ, Tadimalla A, Chen WJ, Martindale JJ, Thuer- ness MS, Ma MH, Emery MG, et al. 2007. Six-month
auf DJ, Marcinko M, Gude N, et al. 2008. Coordination continuous intraputamenal infusion toxicity study of
of growth and endoplasmic reticulum stress signaling by recombinant methionyl human glial cell line-derived
regulator of calcineurin 1 (RCAN1), a novel ATF6- neurotrophic factor (r-metHuGDNF) in rhesus monkeys.
inducible gene. J Biol Chem 283:14012–14021. Toxicol Pathol 35:676–692.
Bespalov MM, Saarma M. 2007. GDNF family receptor Howells DW, Porritt MJ, Wong JY, Batchelor PE, Kalnins
complexes are emerging drug targets. Trends Pharmacol R, Hughes AJ, Donnan GA. 2000. Reduced BDNF
Sci 28:68–74. mRNA expression in the Parkinson’s disease substantia
Bruhn H. 2005. A short guided tour through functional and nigra. Exp Neurol 166:127–135.
structural features of saposin-like proteins. Biochem J Huang EJ, Reichardt LF. 2001. Neurotrophins: Roles in
389:249–257. neuronal development and function. Annu Rev Neurosci
Burke RE. 2004. Ontogenic cell death in the nigrostriatal 24:677–736.
system. Cell Tissue Res 318:63–72. Hudson J, Granholm AC, Gerhardt GA, Henry MA, Hoff-
Chauhan NB, Siegel GJ, Lee JM. 2001. Depletion of glial man A, Biddle P, Leela NS, et al. 1995. Glial cell line-
cell line-derived neurotrophic factor in substantia nigra derived neurotrophic factor augments midbrain dopami-
neurons of Parkinson’s disease brain. J Chem Neuroanat nergic circuits in vivo. Brain Res Bull 36:425–432.
21:277–288. Jongstra J, Schall TJ, Dyer BJ, Clayberger C, Jorgensen J,
Ellgaard L, Ruddock LW. 2005. The human protein disul- Davis MM, Krensky AM. 1987. The isolation and
phide isomerase family: Substrate interactions and func- sequence of a novel gene from a human functional T cell
tional properties. EMBO Rep 6:28–32. line. J Exp Med 165:601–614.
Evron E, Cairns P, Halachmi N, Ahrendt SA, Reed AL, Jordan PA, Gibbins JM. 2006. Extracellular disulfide
Sidransky D. 1997. Normal polymorphism in the incom- exchange and the regulation of cellular function. Anti-
plete trinucleotide repeat of the arginine-rich protein oxid Redox Signal 8:312–324.
gene. Cancer Res 57:2888–2889. Jørgensen JR, Thompson L, Fjord-Larsen L, Krabbe C,
Fontan A, Rojo A, Sanchez Pernaute R, Hernandez I, Lopez I, Torp M, Kalkkinen N, Hansen C, et al. 2009. Characteri-
Castilla C, Sanchez Albisua J, et al. 2002. Effects of fibro- zation of meteorin—An evolutionary conserved neuro-
blast growth factor and glial-derived neurotrophic factor on trophic factor. J Mol Neurosci 39:104–116.
akinesia, F-DOPA uptake and dopamine cells in parkinso- Kadota T, Shingo T, Yasuhara T, Tajiri N, Kondo A,
nian primates. Parkinsonism Relat Disord 8:311–323. Morimoto T, Yuan WJ, et al. 2009. Continuous intraven-
Gill SS, Patel NK, Hotton GR, O’sullivan K, McCarter R, tricular infusion of erythropoietin exerts neuroprotective/
Bunnage M, Brooks DJ, et al. 2003. Direct brain infusion rescue effects upon Parkinson’s disease model of rats
of glial cell line-derived neurotrophic factor in Parkinson with enhanced neurogenesis. Brain Res 1254:120–127.
disease. Nat Med 9:589–595. Kotani Y, Matsuda S, Sakanaka M, Kondoh K, Ueno S,
Grothe C, Timmer M. 2007. The physiological and pharma- Sano A. 1996. Prosaposin facilitates sciatic nerve regen-
cological role of basic fibroblast growth factor in the eration in vivo. J Neurochem 66:2019–2025.
dopaminergic nigrostriatal system. Brain Res Rev 54: Krieglstein K. 2004. Factors promoting survival of mesence-
80–91. phalic dopaminergic neurons. Cell Tissue Res 318:73–80.
Developmental Neurobiology
370 Lindholm and Saarma

Lang AE, Gill S, Patel NK, Lozano A, Nutt JG, Penn R, tebrate neurotrophic factor supporting dopaminergic neu-
Brooks DJ, et al. 2006. Randomized controlled trial of rons. Proc Natl Acad Sci USA 106:2429–2434.
intraputamenal glial cell line-derived neurotrophic Parain K, Murer MG, Yan Q, Faucheux B, Agid Y, Hirsch
factor infusion in Parkinson disease. Ann Neurol 59: E, Raisman-Vozari R. 1999. Reduced expression of
459–466. brain-derived neurotrophic factor protein in Parkinson’s
Lee AH, Iwakoshi NN, Glimcher LH. 2003. XBP-1 regu- disease substantia nigra. Neuroreport 10:557–561.
lates a subset of endoplasmic reticulum resident chaper- Parkash V, Lindholm P, Peränen J, Kalkkinen N, Oksanen
one genes in the unfolded protein response. Mol Cell E, Saarma M, Leppänen VM, et al. 2009. The structure
Biol 23:7448–7459. of the conserved neurotrophic factors MANF and CDNF
Levi-Montalcini R. 1987. The nerve growth factor 35 years explains why they are bifunctional. Protein Eng Des Sel
later. Science 237:1154–1162. 22:233–241.
Liepinsh E, Andersson M, Ruysschaert JM, Otting G. 1997. Patel NK, Bunnage M, Plaha P, Svendsen CN, Heywood P,
Saposin fold revealed by the NMR structure of NK-lysin. Gill SS. 2005. Intraputamenal infusion of glial cell line-
Nat Struct Biol 4:793–795. derived neurotrophic factor in PD: A two-year outcome
Lin LF, Doherty DH, Lile JD, Bektesh S, Collins F. 1993. study. Ann Neurol 57:298–302.
GDNF: A glial cell line-derived neurotrophic factor for Pena SV, Hanson DA, Carr BA, Goralski TJ, Krensky AM.
midbrain dopaminergic neurons. Science 260:1130– 1997. Processing, subcellular localization, and function
1132. of 519 (granulysin), a human late T cell activation mole-
Lindholm P, Peränen J, Andressoo JO, Kalkkinen N, cule with homology to small, lytic, granule proteins.
Kokaia Z, Lindvall O, Timmusk T, et al. 2008. MANF is J Immunol 158:2680–2688.
widely expressed in mammalian tissues and differently Petrova P, Raibekas A, Pevsner J, Vigo N, Anafi M, Moore
regulated after ischemic and epileptic insults in rodent MK, Peaire AE, et al. 2003. MANF: A new mesence-
brain. Mol Cell Neurosci 39:356–371. phalic, astrocyte-derived neurotrophic factor with selectiv-
Lindholm P, Voutilainen MH, Laurén J, Peränen J, Leppä- ity for dopaminergic neurons. J Mol Neurosci 20:173–188.
nen VM, Andressoo JO, Lindahl M, et al. 2007. Novel Piepoli A, Gentile A, Valvano MR, Barana D, Oliani
neurotrophic factor CDNF protects and rescues midbrain C, Cotugno R, Quitadamo M, et al. 2006. Lack of
dopamine neurons in vivo. Nature 448:73–77. association between UGT1A7. UGT1A9, ARP,
Liu J, Wang CY, O’Brien JS. 2001. Prosaptide D5, a retro- SPINK1 and CFTR gene polymorphisms and pancre-
inverso 11-mer peptidomimetic, rescued dopaminergic atic cancer in Italian patients. World J Gastroenterol
neurons in a model of Parkinson’s disease. FASEB J 15: 12:6343–6348.
1080–1082. Puskovic V, Wolfe D, Wechuck J, Krisky D, Collins J, Glo-
Love S, Plaha P, Patel NK, Hotton GR, Brooks DJ, Gill SS. rioso JC, Fink DJ, et al. 2006. HSV-mediated delivery of
2005. Glial cell line-derived neurotrophic factor induces erythropoietin restores dopaminergic function in MPTP-
neuronal sprouting in human brain. Nat Med 11:703– treated mice. Mol Ther 14:710–715.
704. Raykhel I, Alanen H, Salo K, Jurvansuu J, Nguyen VD,
Lu B, Pang PT, Woo NH. 2005. The yin and yang of neuro- Latva-Ranta M, Ruddock L. 2007. A molecular specific-
trophin action. Nat Rev Neurosci 6:603–614. ity code for the three mammalian KDEL receptors. J Cell
Mizobuchi N, Hoseki J, Kubota H, Toyokuni S, Nozaki J, Biol 179:1193–1204.
Naitoh M, Koizumi A, et al. 2007. ARMET is a soluble Romero-Ramirez L, Cao H, Nelson D, Hammond E, Lee
ER protein induced by the unfolded protein response via AH, Yoshida H, Mori K, et al. 2004. XBP1 is essential
ERSE-II element. Cell Struct Funct 32:41–50. for survival under hypoxic conditions and is required for
Mogi M, Togari A, Kondo T, Mizuno Y, Komure O, Kuno tumor growth. Cancer Res 64:5943–5947.
S, Ichinose H, et al. 1999. Brain-derived growth factor Rossmann M, Schultz-Heienbrok R, Behlke J, Remmel N,
and nerve growth factor concentrations are decreased in Alings C, Sandhoff K, Saenger W, et al. 2008. Crystal
the substantia nigra in Parkinson’s disease. Neurosci Lett structures of human saposins C and D: Implications for
270:45–48. lipid recognition and membrane interactions. Structure
Nishino J, Yamashita K, Hashiguchi H, Fujii H, Shimazaki 16:809–817.
T, Hamada H. 2004. Meteorin: A secreted protein that Sauer H, Oertel WH. 1994. Progressive degeneration of
regulates glial cell differentiation and promotes axonal nigrostriatal dopamine neurons following intrastriatal
extension. EMBO J 23:1998–2008. terminal lesions with 6-hydroxydopamine: A combined
O’Brien JS, Carson GS, Seo HC, Hiraiwa M, Kishimoto Y. retrograde tracing and immunocytochemical study in the
1994. Identification of prosaposin as a neurotrophic fac- rat. Neuroscience 59:401–415.
tor. Proc Natl Acad Sci USA 91:9593–9596. Schober A. 2004. Classic toxin-induced animal models of
O’Brien JS, Carson GS, Seo HC, Hiraiwa M, Weiler S, Parkinson’s disease: 6-OHDA and MPTP. Cell Tissue
Tomich JM, Barranger JA, et al. 1995. Identification of Res 318:215–224.
the neurotrophic factor sequence of prosaposin. FASEB J Schwertassek U, Balmer Y, Gutscher M, Weingarten L,
9:681–685. Preuss M, Engelhard J, Winkler M, et al. 2007. Selective
Palgi M, Lindström R, Peränen J, Piepponen TP, Saarma redox regulation of cytokine receptor signaling by extrac-
M, Heino TI. 2009. Evidence that DmMANF is an inver- ellular thioredoxin-1. EMBO J 26:3086–3097.
Developmental Neurobiology
Novel CDNF/MANF Family of NTFs 371

Shridhar R, Shridhar V, Rivard S, Siegfried JM, Pietrasz- Tadimalla A, Belmont PJ, Thuerauf DJ, Glassy MS,
kiewicz H, Ensley J, Pauley R, et al. 1996a. Mutations in Martindale JJ, Gude N, Sussman MA, et al. 2008. Mesen-
the arginine-rich protein gene, in lung, breast, and pros- cephalic astrocyte-derived neurotrophic factor is an
tate cancers, and in squamous cell carcinoma of the head ischemia-inducible secreted endoplasmic reticulum
and neck. Cancer Res 56:5576–5578. stress response protein in the heart. Circ Res 103:1249–
Shridhar V, Rivard S, Shridhar R, Mullins C, Bostick L, 1258.
Sakr W, Grignon D, et al. 1996b. A gene from human Tanaka H, Shimada Y, Harada H, Shinoda M, Hatooka S,
chromosomal band 3p21.1 encodes a highly conserved Imamura M, Ishizaki K. 2000. Polymorphic variation of
arginine-rich protein and is mutated in renal cell carcino- the ARP gene on 3p21 in Japanese esophageal cancer
mas. Oncogene 12:1931–1939. patients. Oncol Rep 7:591–593.
Shridhar V, Rivard S, Wang X, Shridhar R, Paisley C, Mul- Uehara T, Nakamura T, Yao D, Shi ZQ, Gu Z, Ma Y, Mas-
lins C, Beirnat L, et al. 1997. Mutations in the arginine- liah E, et al. 2006. S-nitrosylated protein-disulphide
rich protein gene (ARP) in pancreatic cancer. Oncogene isomerase links protein misfolding to neurodegeneration.
14:2213–2216. Nature 441:513–517.
Silva RM, Ries V, Oo TF, Yarygina O, Jackson-Lewis V, Vaccaro AM, Salvioli R, Barca A, Tatti M, Ciaffoni F,
Ryu EJ, Lu PD, et al. 2005. CHOP/GADD153 is a medi- Maras B, Siciliano R, et al. 1995. Structural analysis of
ator of apoptotic death in substantia nigra dopamine neu- saposin C and B. Complete localization of disulfide
rons in an in vivo neurotoxin model of parkinsonism. bridges. J Biol Chem 270:9953–9960.
J Neurochem 95:974–986. Vogel A, Schwarzmann G, Sandhoff K. 1991. Glycosphin-
Slevin JT, Gerhardt GA, Smith CD, Gash DM, Kryscio R, golipid specificity of the human sulfatide activator pro-
Young B. 2005. Improvement of bilateral motor func- tein. Eur J Biochem 200:591–597.
tions in patients with Parkinson disease through the uni- Voutilainen MH, Bäck S, Pörsti E, Toppinen L, Lindgren
lateral intraputaminal infusion of glial cell line-derived L, Lindholm P, Peränen J, et al. 2009. Mesencephalic
neurotrophic factor. J Neurosurg 102:216–222. astrocyte-derived neurotrophic factor is neurorestorative
Sugaya R, Ishimaru S, Hosoya T, Saigo K, Emori Y. 1994. in rat model of Parkinson’s disease. J Neurosci 29:9651–
A Drosophila homolog of human proto-oncogene ret 9659.
transiently expressed in embryonic neuronal precursor Yasuhara T, Shingo T, Muraoka K, Kameda M, Agari T, Wen
cells including neuroblasts and CNS cells. Mech Dev 45: Ji Y, Hayase H, et al. 2005. Neurorescue effects of VEGF on
139–145. a rat model of Parkinson’s disease. Brain Res 1053:10–18.
Szegezdi E, Duffy A, O’Mahoney ME, Logue SE, Mylotte Zacchigna S, Lambrechts D, Carmeliet P. 2008. Neurovas-
LA, O’Brien T, Samali A. 2006a. ER stress contributes cular signalling defects in neurodegeneration. Nat Rev
to ischemia-induced cardiomyocyte apoptosis. Biochem Neurosci 9:169–181.
Biophys Res Commun 349:1406–1411. Zhu B, Pennack JA, McQuilton P, Forero MG, Mizuguchi
Szegezdi E, Logue SE, Gorman AM, Samali A. 2006b. K, Sutcliffe B, Gu CJ, et al. 2008. Drosophila neurotro-
Mediators of endoplasmic reticulum stress-induced apo- phins reveal a common mechanism for nervous system
ptosis. EMBO Rep 7:880–885. formation. PLoS Biol 6:e284.

Developmental Neurobiology

You might also like