ArticlePDF Available

Enhanced autophagic cell death in expanded polyhistidine variants of HOXA1 reduces PBX1-coupled transcriptional activity and inhibits neuronal differentiation

Authors:

Abstract and Figures

HOXA1 is a member of the homeobox gene family and is involved in early brain development. In our previous study, we identified novel variants of polyhistidine repeat tract in HOXA1 gene and showed that ectopic expression of expanded variants led to enhanced intranuclear aggregation and accelerated cell death in a time-dependent manner. Here, we further investigate the implications of polyhistidine variants on HOXA1 function. Aside from intranuclear aggregation, we observed cytosolic aggregates during the early stages of expression. Rapamycin, an autophagy inducer, resulted in decreased protein aggregation and cell death. Here, we also show an interaction between variants of HOXA1 and one of the HOX protein known cofactors, PBX1. Expanded HOXA1 variants exhibited reduced PBX1-coupled transcriptional activity through a regulatory enhancer of HOXB1. Moreover, we demonstrate that both deleted and expanded variants inhibited neurite outgrowth in retinoic acid-induced neuronal differentiation in neuroblastoma cells. These results provide further evidence that expanded polyhistidine repeats in HOXA1 enhance aggregation and cell death, resulting in impaired neuronal differentiation and cooperative binding with PBX1.
Protein aggregations in the cytosol are cleared by an autophagic process. A: Right after transfection, cells were treated with 100 μM z-VAD-fmk and scored after 24 hr postransfection. z-VAD-fmk did not inhibit intranuclear protein aggregation. Mock EGFP served as a negative control. B: Fluorescence images of EGFP and HOXA1-EGFP 12-His variant treated with z-VAD-fmk. C: COS-7 cells were transfected with HOXA1-EGFP constructs at 18 hr posttransfection; cytosolic aggregates could be seen abundantly in cells transfected with HOXA1-EGFP 12-histidine variant. These aggregates were eventually cleared out after 42 hr and were mostly concentrated in the nuclei. D: Graph showing percentage of cells with protein aggregation and dead cells per number of EGFP-positive transfected cells at 18 and 42 hr after transfection. E: Transfected cells were treated with 10 mM 3-methyladenine (3-MA) 15 hr prior to fixation. HOXA1-EGFP 10-His variant-transfected cells were treated with DMSO to serve as a control for cell toxicity. 3-MA increases protein aggregations and cell death in HOXA1-EGFP-transfected COS-7 cells. Dead cells and those exhibiting nuclear and cytosolic aggregation were scored 18 hr after transfection. F: No protein aggregation was detected in mock-EGFP-transfected COS-7 cells treated with 3-MA; however, an increase in cell death was observed. HOXA1-EGFP 12-His variant exhibited cytosolic aggregates around the outer perinuclear periphery, which are denoted by arrows. G: Immediately after transfection, 10-His and 12-His repeat variants of HOXA1-EGFP-transfected COS-7 cells were treated with 2 μg/ml Rapamycin prior to fixation. Rapamycin reduces cytosolic aggregation and cell death significantly in the cells transfected with 12-His repeat variant. Dead cells and protein aggregations were scored 42 hr after transfection. Error bars represent SEM; n = 3. *P < 0.05, **P < 0.01. P values from a paired t-test in all experiments. [Color figure can be viewed in the online issue, which is available at www.interscience.wiley.com.]
… 
Content may be subject to copyright.
Enhanced Autophagic Cell Death in
Expanded Polyhistidine Variants of
HOXA1 Reduces PBX1-Coupled
Transcriptional Activity and Inhibits
Neuronal Differentiation
Rubigilda C. Paraguison,
1
Katsumi Higaki,
1
Kenji Yamamoto,
2
Hideo Matsumoto,
2
Tsukasa Sasaki,
3
Nobumasa Kato,
3
and Eiji Nanba
1
*
1
Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University,
Yonago, Japan
2
Department of Psychiatry and Behavioral Sciences, Tokai University School of Medicine,
Kanagawa, Japan
3
Department of Psychiatry, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
HOXA1 is a member of the homeobox gene family and
is involved in early brain development. In our previous
study, we identified novel variants of polyhistidine re-
peat tract in HOXA1 gene and showed that ectopic
expression of expanded variants led to enhanced intra-
nuclear aggregation and accelerated cell death in a
time-dependent manner. Here, we further investigate the
implications of polyhistidine variants on HOXA1 func-
tion. Aside from intranuclear aggregation, we observed
cytosolic aggregates during the early stages of expres-
sion. Rapamycin, an autophagy inducer, resulted in de-
creased protein aggregation and cell death. Here, we
also show an interaction between variants of HOXA1
and one of the HOX protein known cofactors, PBX1.
Expanded HOXA1 variants exhibited reduced PBX1-
coupled transcriptional activity through a regulatory en-
hancer of HOXB1. Moreover, we demonstrate that both
deleted and expanded variants inhibited neurite out-
growth in retinoic acid-induced neuronal differentiation
in neuroblastoma cells. These results provide further
evidence that expanded polyhistidine repeats in HOXA1
enhance aggregation and cell death, resulting in im-
paired neuronal differentiation and cooperative binding
with PBX1. V
V
C2006 Wiley-Liss, Inc.
Key words: HOXA1; neuronal differentiation; PBX1; poly-
histidine; protein aggregation; autophagy
HOX genes form a subset of the family of home-
obox genes (Pearson et al., 2005). They are involved in
specifying positional identity along the anterior-posterior
axis of all bilaterian animals. In humans, the HOXA–D
clusters comprise 39 HOX genes, located on chromo-
some regions 7p15, 17p21, 12q13, and 2q31 (Grier et al.,
2005). During embryonic development, HOX genes
are expressed sequentially 30to 50along the anterior to
posterior axis. HOX genes contain a 61-amino-acid
helix-turn-helix DNA-binding domain known as the
homeodomain (Gehring et al., 1994). It is well established
that HOX/DNA binding specificity is modified by other
DNA-binding proteins, which act as cofactors. Among
these are the PBX proteins, which are widely expressed
in fetal and adult tissues and interact preferentially with
30HOX proteins (Phelan and Featherstone, 1997). PBX
can modulate the affinity and stability of DNA binding
and regulate transcriptional activity. The cooperative he-
terodimerization is carried out through a conserved
protein motif found N-terminal to the Hox homeodo-
main (Slupsky et al., 2001; Huang et al., 2005). Interac-
tions between PBX and HOX might also be mediated
by residues of the N-terminal arm of HOX proteins
(Shanmugam et al., 1997). It has been reported that
PBX1 and HOXB1 can cooperatively activate the tran-
scription through an autoregulatory element, directing
spatially restricted expression of the HOXB1 gene (b1-
ARE) in the developing hindbrain. However, only lim-
ited kinds of HOX can bind cooperatively with PBX
(HOXA1, HOXB1, and HOXA2; Di Rocco et al.,
Contract grant sponsor: Japanese Ministry of Health, Labor and Welfare;
Contract grant number: H14-kokoro-002; Contract grant sponsor: Japa-
nese Ministry of Education, Culture Sports, Science and Technology;
Contract grant number: 2005, 17659315; Contract grant number: 2004,
16012242.
*Correspondence to: Eiji Nanba, MD, PhD, Division of Functional
Genomics, Research Center for Bioscience and Technology, Tottori
University, 86 Nishi-machi, Yonago 683-8503, Japan.
E-mail: enanba@grape.med.tottori-u.ac.jp
Received 18 April 2006; Revised 4 October 2006; Accepted 5 October
2006
Published online 27 November 2006 in Wiley InterScience (www.
interscience.wiley.com). DOI: 10.1002/jnr.21137
Journal of Neuroscience Research 85:479–487 (2007)
'2006 Wiley-Liss, Inc.
2001). This autoregulatory enhancer is the key regula-
tory element for the normal rhombomere 4 expression
of Hoxb1 in the developing hindbrain, whereas Hoxa1
and Hoxb1 synergize in patterning the hindbrain cranial
nerves and second pharyngeal arch (Gavalas et al., 1998).
HOXA1 is one of the first HOX genes to be
expressed during embryonic development (Pearson et al.,
2005). In mice, its expression starts from 7.5 dpc, and it
is established in the neuroectoderm and mesoderm at
8.0 dpc. (Remacle et al., 2004). HOXA1 gene encodes
two alternatively spliced mRNAs, which appear to be
differentially expressed in the developing embryo. The
homeodomain-containing variant 1 undergoes transcrip-
tional activation during hindbrain development from E7
to E8.5 (Godwin et al., 1998). Functional inactivation of
this gene results in prenatal lethality and numerous mal-
formations (Lufkin et al., 1991; Carpenter et al., 1993).
Hoxa1 null mice exhibit hindbrain segmentation and pa-
tterning defects that cause abnormal development of
cranial nerve, cranial ganglia, and branchial arch deriva-
tives (Chisaka et al., 1992). Furthermore, ectopic expres-
sion of Hoxa1 in transgenic mice leads to abnormalities
of the developing hindbrain and ultimately results in
embryonic death (Zhang et al., 1994).
HOXA1 gene contains a tract of 10-histidine re-
peat. In our previous study, we identified novel variants
of polyhistidine tracts in HOXA1 gene in a Japanese
population, and no homozygous case has been found for
any of these variants (Paraguison et al., 2005). Certain
individuals were heterozygous for deleted 7- and 9-histi-
dine repeats and expanded 11- and 12-histidine repeats.
Expression of expanded polyhistidine variants of HOXA1
proteins resulted in accelerated formation of ubiquitinated
intranuclear aggregates and increased cell death. How-
ever, the mechanism by which this aggregation occurs is
poorly understood. In this study, we showed that expres-
sion of expanded polyhistidine variants of HOXA1 in
human neuroblastoma cell line SK-N-SH and embryonic
carcinoma cell line P19 also caused increased intranuclear
aggregation and cell death. However, there was a signifi-
cant reduction of protein aggregation and cell death
upon Rapamycin treatment, indicating involvement of
the autophagic process. Expanded variants exhibited im-
paired cooperative binding with the cofactor PBX1,
resulting in decreased transcriptional activity. Moreover,
cells overexpressing expanded and deleted variants ex-
hibited impaired neuronal differentiation. These data
provide new insights on the function of polyhistidine
variants of HOXA1 protein in neuronal cells.
MATERIALS AND METHODS
Antibodies and Reagents
The following antibodies were used: polyclonal goat
anti-HOXA1 antibody (Santa Cruz Biotechnology, Santa
Cruz, CA; sc-17146), monoclonal mouse anti-EGFP antibody
(Santa Cruz Biotechnology, sc-9996), polyclonal rabbit anti-
MAP2, H-300 (Santa Cruz Biotechnology, sc-20172), poly-
clonal rabbit anti PBX1, P-20 (Santa Cruz Biotechnology,
sc-889), and polyclonal rabbit anti-b-tubulin H235 (Santa
Cruz Biotechnology, sc-9104) and Alexa Fluor 594-conjugated
anti-goat IgG, Alexa Fluor 555-conjugated anti-rabbit IgG,
and Alexa Fluor 555-conjugated anti-mouse IgG (Molecular
Probes, Eugene, OR). The following reagents were also used
for this study: 10 lM retinoic acid (Sigma, St. Louis, MO;
R2625) immediately after transfection, 10 mM 3-methyladenine
(3-MA; Sigma, M9281) 15 hr prior to fixation, and 100 lM
z-VAD-fmk (Promega, Madison, WI; G7231) and 2 lg/ml
Rapamycin (Sigma, R0395) both right after transfection.
Expression Vectors and Reporter Construct
Human HOXA1 expression vectors were generated as
described previously (Paraguison et al., 2005). The following
primer sets with suitable restriction enzyme recognition sites
(Sac I and Bam HI) were used to generate HOXA1 variant 1:
50- TAGAGCTCACCATGGACAATGCAAGAATGAACT-
CC-30and 50-ATGGATCCGTGTGGGAGGTAGTCAGA-
GTGTCTGA-30. All DNA amplification steps were per-
formed using high fidelity Pfu Ultra DNA polymerase (Strata-
gene, La Jolla, CA) using genomic DNA and cDNA derived
from human normal lymphoblast and confirmed by sequenc-
ing (ABI 3130xl). Expression constructs were derived from
CMV promoter-based expression vector, pCMV-Script (Stra-
tagene) and pEGFP-N1 (Invitrogen, San Diego, CA). The
expression plasmids pCMV-Script and pEGFP-N1 were used
as empty vector controls. The luciferase reporter construct
pAdMLARE containing b1-ARE and the Pbx1 expression
construct pSGPbx1a were generous gifts of Prof. Zappavigna
(Di Rocco et al., 2001).
Cell Culture and Transfection
COS-7 cells, murine P19 embryonal carcinoma (EC)
cells, and the human neuroblastoma cell line SK-N-SH were
maintained in Dulbecco’s modified Eagle’s medium (DMEM;
Sigma; D6429) supplemented with 10% fetal bovine serum
(FBS; HyClone, Logan, UT). Cultures 50–80% confluent
were transfected with Fugene 6 transfection reagent (Roche,
Indianapolis, IN) in accordance with the manufacturer’s rec-
ommendations. For a typical experiment, 1 lg of expression
vector was used in a 35-mm culture dish. For cotransfection
experiments, 1 lg of reporter plasmid (pAdMLARE), 0.5 lg
of HOXA1 expression construct, 1 lg of PBX1 expression
construct, and 0.3 lg of pEGFP as an internal control were
used in a 35-mm dish. For Western blot analyses, cells cultured
in 10-cm dishes were transfected with 10 lg of plasmid con-
struct using Fugene-6 (Roche). To initiate differentiation,
SK-N-SH cells were inoculated and treated with 10 lM
retinoic acid (RA), whereas P19 cells were grown in a serum-
free condition. Dead cells were scored under a fluorescence
microscope (Leica DMIRE2).
Luciferase Assay
After 24 hr posttransfection, the cells were harvested
and lysed in Pica Gene cell culture lysis reagent (Toyo Ink,
Tokyo, Japan). Luciferase assay was carried out by using a
Pica Gene kit (Toyo Ink) in accordance with the manufac-
turer’s protocol. HOXA1 cooperative expression with PBX1
480 Paraguison et al.
Journal of Neuroscience Research DOI 10.1002/jnr
using pAdMLARE reporter construct was assessed by quanti-
tative luciferase assay with Luminescencer-PSN (Bio-Instru-
ment ATTO AB-220). pEGFP-N1 plasmid was used as an in-
ternal control.
Immunostaining, Dead Cell Scoring, and Imaging
The cells transiently expressing HOXA1 constructs were
grown in 35-mm dishes containing glass coverslips. Cells
attached to the glass coverslips were washed with phosphate-
buffered saline (PBS), fixed with 4% paraformaldehyde, and
incubated with antibodies as described previously (Paraguison
et al., 2005). Confocal scanning analysis was performed with a
Leica confocal microscope (Leica TCS-SP2). The degree of
protein accumulation within the cell nuclei and the cytosol of
transfected cells was scored. The cells were counted from
10 randomly selected microscope fields of each sample, 15 in
the case of SK-N-SH cells. The ratio of the number of cells
with protein aggregation over the total number of cells was
then computed. Each experiment was performed independ-
ently in triplicate.
Western Blotting and Immunoprecipitation
Posttransfected cells were harvested and lysed by sonica-
tion in a buffer containing 10 mM Tris-HCl (pH 7.4),
150 mM NaCl, 1 mM EDTA, 1 mM EGTA, and protease
inhibitor cocktail (Roche). Proteins were quantified by using
a protein assay rapid kit (Wako, Osaka, Japan), run on 10% or
12% SDS-PAGE gels, and transferred on PVDF membranes
(Millipore, Bedford, MA; IPVH00010) by a semidry blotter
(Bio-Rad, Hercules, CA). Membranes were incubated with
antibodies as described previously. Protein lysates from cotrans-
fection experiments were immunoprecipitated with anti-GFP
and used for Western blotting with anti-PBX1. Polyclonal
rabbit anti-MAP2 antibody was used for quantifying neuronal
differentiation in SK-N-SH. Signals were detected using ECL
reagent (Amersham-Pharmacia Biotech, Arlington Heights, IL)
on X-ray films (Fuji). For quantification, images were ana-
lyzed in NIH Image software.
RESULTS
Expanded Polyhistidine Variants of HOXA1
Enhanced Intranuclear Protein Aggregation
in Neuronal Cell Lines
We have previously reported that expanded poly-
histidine variants in HOXA1 resulted in early nuclear
protein aggregation and an increased cell death in COS7
cells (Paraguison et al., 2005). Increasing polyhistidine
repeat length coincided with early protein aggregation
and accelerated cell death. In contrast, no significant dif-
ference was observed between cells overexpressing 7-poly-
histidine variant and wild-type 10-polyhistidine repeat
variant.
In the current study, we observed that these nu-
clear aggregations were also detected in both SK-N-SH
cells (Fig. 1A) and P19 cells (Fig. 1B). To rule out the
possibility that these aggregates were caused by enhanced
green fluorescent protein (EGFP) tagging, transfection
with untagged cytomegalovirus (CMV)-driven expres-
sion of HOXA1 variants was performed in COS-7 cells.
Immunostaining with anti-HOXA1/Alexa 594 anti-goat
also exhibited protein aggregates 18 hr posttransfection
(Fig. 1C). After 18–20 hr, cells transfected with EGFP-
tagged HOXA1 proteins were analyzed for Western
blotting. In lysates of cells transfected with 11- and 12-
polyhistidine variants, insoluble high-molecular-weight
proteins remained in the stacking gel (Fig. 1D), indica-
tive of protein complex accumulation.
Rapamycin Cleared Protein Aggregations and
Decreased Cell Death in COS-7 Cells Expressing
Expanded Variants of HOXA1, Whereas 3-MA
Reversed This Effect
We also attempted to determine the type of cell
death occurring as a consequence of expression of
HOXA1 expanded polyhistidine variants. Recently, it
has been reported that autophagic cell death was partially
mediated by caspase activation (Ravikumar et al., 2006).
Thus, we examined whether caspase inhibition could
suppress cell death by using a cell-permeable pan caspase
inhibitor, z-VAD-fmk, that binds to the catalytic site of
caspase proteases and can inhibit induction of apoptosis
(Broustas et al., 2004). However, no significant inhibi-
tion was seen in cells treated with 100 lM z-VAD-fmk
(Fig. 2A,B). Thus, we speculated that classical apoptosis
is not the mechanism involved in HOXA1-related cell
death.
To understand better the mechanism responsible
for clearance of protein aggregates, we examined the
possible role of autophagy in degrading these proteins and
the effect of caspase inhibition on cell death in COS-7
cells. During our time course experiments, we observed
that aggregations were present not only in the nucleus
but also in the cytosol at the early stages of expression
(18 hr after transfection). These cytosolic aggregates
were abundant in cells expressing the expanded 12-histi-
dine variants of HOXA1-EGFP (Fig. 2C). Aggregates
gradually cleared out by the endogenous autophagic
mechanism of the cell and were concentrated in the
nuclei at 42 hr after transfection (Fig. 2C,D). We ex-
plored the possible involvement of the autophagic pro-
cess in the clearing of protein aggregates in these cells.
3-Methyladenine (3-MA), which has an inhibitory ef-
fect on autophagy (Ravikumar et al., 2002), enhanced
nuclear aggregations and cell death (Fig. 2E,F). There is
a decreased rate of clearance of protein aggregates
observed in 3-MA-treated cells overexpressing 12-His
variant. Cytosolic aggregates around the outer perinu-
clear periphery are noticeable in the later stage of expres-
sion, i.e., about 42 hr, where they were supposed to
be cleared out in the 3-MA nontreated transfected cells
(Fig. 2F). Conversely, rapamycin, an autophagy-inducing
chemical (Ravikumar et al., 2006), decreased cytosolic and
nuclear aggregation that eventually reduced cell death
particularly in the expanded 12-histidine variant (Fig. 2G).
This suggested that clearing of HOXA1-EGFP protein
aggregates was mediated by an autophagic mechanism. We
Polyhistidine Variants of HOXA1 in Neurons 481
Journal of Neuroscience Research DOI 10.1002/jnr
further noted that the autophagic clearing system was
more efficient in degrading cytosolic aggregates, since
autophagic vacuoles were directly accessible in the cytosol.
However, the increased production of accumulated intra-
nuclear proteins might have overloaded the autophagic
clearing mechanism, thus resulting in cell death.
Fig. 1. Expression of HOXA1 polyhistidine expansion variants results
in protein aggregation. HOXA1-EGFP construct variants were expressed
in neuronal cell lines. As early as 15–20 hr after transfection, intranuclear
aggregates were already detected in cells transfected with expanded
HOXA1 variant. Images were taken 18 hr after transfection. EGFP fluo-
rescent signals were visualized by confocal microscopy. A: Neuroblas-
toma cells, SK-N-SH. B: Embryonic carcinoma cells, P19. C: COS-7
cells transfected with untagged HOXA1 and immunostained with anti-
HOXA1/Alexa 594 anti-goat (red signals). Fluorescence images were
taken 18 hr posttransfection. D: 18–24-hr posttransfected cell lysates an-
alyzed for Western blotting using anti-GFP. An increased amount of
high-molecular-weight SDS-insoluble protein was detected in the stack-
ing gel in lanes of expanded HOXA1, indicating that the length of histi-
dine repeats is directly proportional with the degree protein accumula-
tions. [Color figure can be viewed in the online issue, which is available
at www.interscience.wiley.com.]
Fig. 2. Protein aggregations in the cytosol are cleared by an autopha-
gic process. A: Right after transfection, cells were treated with
100 lM z-VAD-fmk and scored after 24 hr postransfection. z-VAD-
fmk did not inhibit intranuclear protein aggregation. Mock EGFP
served as a negative control. B: Fluorescence images of EGFP and
HOXA1-EGFP 12-His variant treated with z-VAD-fmk. C: COS-7
cells were transfected with HOXA1-EGFP constructs at 18 hr post-
transfection; cytosolic aggregates could be seen abundantly in cells
transfected with HOXA1-EGFP 12-histidine variant. These aggre-
gates were eventually cleared out after 42 hr and were mostly con-
centrated in the nuclei. D: Graph showing percentage of cells with
protein aggregation and dead cells per number of EGFP-positive
transfected cells at 18 and 42 hr after transfection. E: Transfected cells
were treated with 10 mM 3-methyladenine (3-MA) 15 hr prior to
fixation. HOXA1-EGFP 10-His variant-transfected cells were treated
with DMSO to serve as a control for cell toxicity. 3-MA increases
protein aggregations and cell death in HOXA1-EGFP-transfected
COS-7 cells. Dead cells and those exhibiting nuclear and cytosolic
aggregation were scored 18 hr after transfection. F: No protein
aggregation was detected in mock-EGFP-transfected COS-7 cells
treated with 3-MA; however, an increase in cell death was observed.
HOXA1-EGFP 12-His variant exhibited cytosolic aggregates around
the outer perinuclear periphery, which are denoted by arrows. G:
Immediately after transfection, 10-His and 12-His repeat variants of
HOXA1-EGFP-transfected COS-7 cells were treated with 2 lg/ml
Rapamycin prior to fixation. Rapamycin reduces cytosolic aggrega-
tion and cell death significantly in the cells transfected with 12-His
repeat variant. Dead cells and protein aggregations were scored 42 hr
after transfection. Error bars represent SEM; n ¼3. *P<0.05,
**P<0.01. Pvalues from a paired t-test in all experiments. [Color
figure can be viewed in the online issue, which is available at
www.interscience.wiley.com.]
"
482 Paraguison et al.
Journal of Neuroscience Research DOI 10.1002/jnr
Figure 2.
Polyhistidine Variants of HOXA1 in Neurons 483
Journal of Neuroscience Research DOI 10.1002/jnr
Transcriptional Activities of Polyhistidine
Variants of HOXA1 Coupled With PBX1
We next examined the physiological relevance of
polyhistidine variants of HOXA1 protein in neuronal
cells. PBX1 plays a major role in cooperative transcrip-
tional activation with HOXA1 through an autoregula-
tory element, the b1-ARE (Di Rocco et al., 2001). To
investigate the cooperative transcription of PBX1 with
Fig. 3. Expanded HOXA1 reduces PBX1-coupled transcriptional
activity. Luciferase assay from the different cell lines. Cells were
cotransfected with plasmids containing PBX1, HOXA1, and the
enhancer b1-ARE fused to a luciferase reporter gene (Di Rocco et al.,
2001). EGFP vector was used as an internal control. A: COS-7. B:
Neuroblastoma cell line SK-N-SH. C: Embryonic carcinoma cell line
P19 grown in serum-free medium. Bars represent the mean 6SE of
at least three independent experiments. *P<0.05, **P<0.01.
Fig. 4. HOXA1 and PBX1 interaction is impaired in expanded
variant of HOXA1. A: Immunofluorescence images of COS-7
cotransfected with PBX1, an enhancer bi-ARE, and 7-His, 10-His, or
12-His repeat variants of HOXA1-EGFP. Fluorescence images were
taken 24 hr posttransfection. Red signals: anti-PBX1. Note the absence
of PBX1 signal in protein aggregates of the 12-His variant. B: Immu-
noprecipitation with anti-GFP and Western blotted with anti-PBX1
and anti-HOXA1. PBX1 protein shows decreased levels in both
expanded and deleted variants of HOXA1-EGFP transfected to COS-
7cells.C: Quantification of bound PBX1 protein levels per HOXA1
protein from Western blot analysis. [Color figure can be viewed in the
online issue, which is available at www.interscience.wiley.com.]
484 Paraguison et al.
Journal of Neuroscience Research DOI 10.1002/jnr
the different variants of HOXA1 through b1-ARE, we
simultaneously expressed HOXA1, PBX1, EGFP (an
internal control), and b1-ARE luciferase reporter con-
structs in COS7, SK-N-SH, and P19 cells. Luciferase
assay were performed 24 hr after transfection. The P19
embryonic cell line was grown under serum-free condi-
tions. Growth in serum-free media itself committed EC
cells to neural differentiation (Darmon et al., 1981) and
addition of retinoic acid (RA) intensified this effect
(Tanaka et al., 1992). However, nonneural cell types arise
after treatment with RA under serum-containing condi-
tions. Differentiation in serum-free media alone is accom-
panied by expression of only neuroectodermal/neural
mRNAs, but treatment with RA invariably induces the
cells to express both neuroectodermal/neural and endo-
dermal mRNAs (Pachernik et al., 2005), so the serum-
free medium condition was used in this experiment.
The transcriptional activities of expanded variants
were significantly reduced in all cell lines, whereas the
activities of deleted variant with 7-polyhistidine were
enhanced in P19 cells compared with the activity of
10-polyhistidine variant (Fig. 3A–C). Immunofluores-
cence images revealed the inability of aggregated forms
of expanded HOXA1 protein to bind efficiently with
PBX1 protein (Fig. 4A). Inefficient interaction between
expanded HOXA1 and PBX1 is clearly indicated by
low levels of PBX1 protein in immunoprecipitation assay
with anti-GFP (Fig. 4B,C).
Expanded and Deleted Polyhistidine Variants in
HOXA1 Inhibited Neuronal Differentiation
Because HOXA1 is one of the neurodevelopmental
genes, we examined whether polyhistidine variants had
any effect on the process of neuronal differentiation. To
accomplish this, RA-induced neuronal differentiation
was performed in SK-N-SH cells. Phase microscopy
shows the effect of RA treatment on the neurites of these
cells (Fig. 5A). Immunocytochemistry with antibody
against the neuron-specific protein MAP2 revealed
neurite outgrowth in cells expressing the 10-histidine
HOXA1-GFP, 42 hr after transfection (Fig. 5B). In con-
trast, a greater number of cells expressing expanded and
deleted forms of HOXA1-GFP failed to initiate neurite
outgrowth. Total levels of MAP2 protein in cells over-
expressing expanded and deleted variants were also
significantly reduced (Fig. 5C,D). The appearance of
Fig. 5. Expanded and deleted polyhistidine repeats in HOXA1 in-
hibit neuronal differentiation. A: Retinoic acid (RA)-induced neuro-
nal differentiation in SK-N-SH cells Phase images of SK-N-SH cells
treated with (right) or without (left) 10 lM RA for 42 hr. B:
HOXA1-EGFP-transfected SK-N-SH treated with 10 lM RA for
42 hr. Immunofluorescence images where taken 42 hr after transfec-
tion. Expanded 11- and 12-His repeat variants as well as deleted
7-His repeat variants inhibit neuronal differentiation. Note the neurite
outgrowth immunostained with MAP2 (red signal). C: Western blot
analysis shows decreased MAP2 expression levels in the expanded and
deleted forms. D: Quantification of MAP2 expression levels normal-
ized with b-tubulin. E: Quantification of the percentage of EGFP-
MAP2-double-positive cells. Error bars represent SEM; n ¼3. *P<
0.05, **P<0.01. Pvalues from a paired t-test in all experiments.
[Color figure can be viewed in the online issue, which is available at
www.interscience.wiley.com.]
"
Polyhistidine Variants of HOXA1 in Neurons 485
Journal of Neuroscience Research DOI 10.1002/jnr
differentiated cells was also examined by scoring MAP2-
positive transformants expressing the HOXA1-EGFP
variants. EGFP-MAP2 double-positive cells quantifica-
tion also confirmed that there is a significant decrease
in the percentage of MAP2 levels particularly in the
expanded variants.
DISCUSSION
In this current study, we have shown that poly-
histidine repeat tract variants compromised HOXA1
function in transcription and neuronal differentiation.
Moreover, we have presented evidence that cell death
resulting from polyhistidine repeat expansion is mediated
primarily by authophagy and not by a caspase-dependent
mechanism. Authophagy is a type of cell death involving
bulk degradation of cytoplasmic proteins or organelles in
the lytic compartment. Inhibition of autophagy enhances
protein aggregation and cell death (Ravikumar et al.,
2002). Our data showed that 3-MA accelerates protein
accumulations and enhances cell death, particularly in
the expanded variants (Fig. 2E,F). Conversely, Rapamy-
cin, an inducer of the autophagic process, decreases pro-
tein aggregates and reduces cell death (Fig. 2G). Taken
together, our findings indicate that autophagy is involved
in the degradation and clearance of aggregations of
expanded HOXA1 variants.
HOXA1 splice variant 1 is reported to be active in
E7–E8.5 and functions in the presumptive rhombomere
3 and 4 regions of the developing hindbrain (Zhang
et al., 1994). However, the nonhomeodomain-contain-
ing variant is expressed in the endodermal derivative
after E8.5 to the adult stage (Godwin et al., 1998).
Therefore, we speculate that splice variant 1 may be
active only in early stages of embryonic neurodevelop-
ment. As one of the DNA-binding proteins and tran-
scription factors in the HOX gene family, HOXA1 also
relies on the activity of transcriptional cofactors aside
from its DNA-binding properties (Pearson et al., 2005).
A previous study has reported that HOXB1 and PBX1
cooperatively activate transcription under the control of
b1-ARE (autoregulatory enhancer). Moreover, HOXA1
and HOXA2 are also able to activate transcription by
b1-ARE in cooperation with PBX1 (Di Rocco et al.,
2001). We examined the binding ability of our HOXA1
variants to PBX1 and b1-ARE by evaluating the coop-
erative transcriptional activation using a luciferase assay.
Expanded variants transfected into COS-7 as well as SK-
N-SH neuroblastoma and P19 embryonic carcinoma
cells (EC) that were induced to undergo neuronal differ-
entiation indeed showed significantly reduced activation
compared with the wild type, whereas the deleted
variant expressed in P19 cell line showed an enhanced
activation (Fig. 3). However, the reason for increased
transcriptional activity observed in the deleted variant in
this cell line is as yet unknown. Immunofluorescence
images and immunoprecipitation assay clearly confirmed
the inability of aggregated forms of expanded HOXA1
protein to bind efficiently with PBX1 protein. The co-
operative interaction between Hox and Pbx is mediated
by a conserved hexapeptide sequence located toward the
N-terminal region from Hox homeodomain (Phelan and
Fetherstone, 1997; Remacle et al., 2004). Our results
suggest that polyhistidine variants in HOXA1 might also
affect the binding efficiency of its homeodomain to
certain cofactors and/or other target genes or proteins.
Human neuroblastoma SK-N-SH cells, from a ma-
lignant pediatric tumor derived from the neural crest,
retains its ability to differentiate into the neuronal lineage
when exposed to RA (Wainwright et al., 2001).
HOXA1 is the first target gene activated by RA, fol-
lowed by a sequential activation of other HOX genes
(Simeone et al., 1990; Martinez-Ceballos et al., 2005).
We showed an inhibition of neuronal differentiation
by not only the extended but also the deleted variants
(Fig. 5). Our study provides novel insights on the patho-
logical implications of the polyhistidine tract in HOXA1
and leaves us with the intriguing possibility that polyhis-
tidine repeat expansions and deletions may cause aberra-
tions in neuronal morphogenesis or differentiation in
general.
EC cells differentiate into various lineages depend-
ing on the presence of activators in the culture medium
(Pachernik et al., 2005). Growth under serum-free con-
ditions committed EC cells to neural differentiation.
Differentiation of EC cells into endodermal-like cells is
induced by serum. Interestingly, we found that, in EC
cells committed to neural differentiation, transcriptional
activity inversely coincided with polyhistidine repeat
length. These results further strengthen our theory that
polyhistidine length affects neurodevelopment. In agree-
ment with our results, another study has proposed that
HOXA1 may function as a stimulator of neuroectoder-
mal and mesodermal differentiation and a repressor
of embryonic endoderm formation (Martinez-Ceballos
et al., 2005) and that aberration in HOXA1 could lead
to increased expression of endodermal genes by RA
and would lead to repression of neuroectodermal and
mesodermal markers. Premature death of HOXA1-
expressing cells may impair transcription and neuronal
differentiation.
Recently, Tischfield and colleagues (2005) reported
that patients with Bosley-Salih-Alohrainy syndrome were
homozygous for HOXA1 truncating mutations, whereas
heterozygotes had normal phenotypes. This mutation
resulted in abnormal development of the central nervous
system in the brainstem. Distinguishing phenotypes
include horizontal gaze abnormalities, mental retardation,
and autism spectrum disorder. Even low levels of Hoxa1
expression in Hoxa1
+/–
cells are sufficient for normal
activation of the Hoxa1 pathway and may explain why
mice heterozygous for HOXA1 mutations appear normal
(Pasqualetti et al., 2001; Martinez-Ceballos et al., 2005).
In our previous report, we identified heterozygous poly-
histidine repeat variants in HOXA1 gene from a
Japanese population comprising normal and autistic indi-
viduals. No individuals homozygous for these mutations
have been found. We speculate that there is a possibility
486 Paraguison et al.
Journal of Neuroscience Research DOI 10.1002/jnr
that phenotypic aberrations may exist in homozygous
individuals. Future in vivo studies are essential to exam-
ine the physiological functions of the polyhistidine var-
iants of HOXA1.
ACKNOWLEDGMENTS
We thank Prof. Vincenzo Zappavigna for provid-
ing the PBX1 and pAdMLARE reporter plasmids.
R.C.P. is the recipient of a research scholarship from the
Japanese Ministry of Education, Culture, Sports, Science
and Technology.
REFERENCES
Broustas CG, Gokhale PC, Rahman A, Dritschlo A, Ahmad I, Kasid U.
2004. BRCC2, a novel BH3-like domain-containing protein, induces
apoptosis in a caspase-dependent manner. J Biol Chem 279:26780–
26788.
Carpenter EM, Goddard JM, Chisaka O, Manley NR, Capecchi MR.
1993. Loss of Hoxa1 (Hox-1.6) function results in the reorganization of
the murine hindbrain. Development 118:1063–1075.
Chisaka O, Musci TS, Cappecchi MR. 1992. Developmental defects of
the ear, cranial nerves and hindbrain resulting from targeted disruption
of the mouse homeobox gene, Hox-1.6. Nature 355:516–520.
Darmon M, Bottenstein J, Sato G. 1981. Neural differentiation following
culture of embryonal carcinoma cells in a serum-free defined medium.
Dev Biol 85:463–473.
Di Rocco G, Gavalas A, Popperl H, Krumlauf R, Mavilio F, Zappavigna V.
2001. The recruitment of SOX/OCT complexes and the differential acti-
vity of HOXA1 and HOXB1 modulate the Hoxb1 auto-regulatory
enhancer function. J Biol Chem 276:20506–20515.
Gavalas A, Studer M, Lumsden A, Rijli FM, Krumlauf R, Chambon P.
1998. Hoxa1 and Hoxb1 synergize in patterning the hindbrain, cranial
nerves and second pharyngeal arch. Development 125:1123–1136.
Gehring WJ, Affolter M, Burglin T. 1994. Homeodomain proteins.
Annu Rev Biochem 63:487–526.
Godwin AR, Stadler HS, Nakamura K, Cappecchi MR. 1998. Detection
of targeted GFP-Hox gene fusions during mouse embryogenesis. Proc
Natl Acad Sci U S A 95:13042–13047.
Grier DG, Thompson A, Kwasniewska A, McGonigle GJ, Halliday HL,
Lappin TR. 2005. The pathophysiology of HOX genes and their role
in cancer. J Pathol 205:154–171.
Huang H, Rastegar M, Bodner C, Goh SL, Rambaldi I, Featherstone M.
2005. MEIS C termini harbor transcriptional activation domains that
respond to cell signaling. J Biol Chem 280:10119–10127.
Lufkin T, Dierich A, LeMeur M, Chambon M. 1991. Disruption of the
Hox-1.6 homeobox gene results in defects in a region corresponding to
its rostral domain of expression. Cell 66:1105–1119.
Martinez-Ceballos E, Chambon P, Gudas LJ. 2005. Differences in gene
expression between wild type and Hoxa1 knockout embryonic stem
cells after retinoic acid treatment or LIF removal. J Biol Chem 280:
16484–16498.
Pachernik J, Bryja V, Esner M, Kubala L, Dvorak P, Hampl A. 2005.
Neural differentiation of pluripotent mouse embryonic carninoma cells
by retinoic acid: inhibitory effects of serum. Physiol J 54:115–122.
Paraguison RC, Higaki K, Sakamoto Y, Hashimoto O, Miyake N,
Matsumoto H, Yamamoto K, Sasaki T, Kato N, Nanba E. 2005. Poly-
histidine tract expansions in HOXA1 result in intranuclear aggregation
and increased cell death. Biochem Biophys Res Commun 336:1033–
1039.
Pasqualetti M, Neun R, Davenne M, Rijli FM. 2001. Retinoic acid
rescues inner ear defects in Hoxa1 deficient mice. Nat Genet 1:34–39.
Pearson JC, Lemons D, McGinnis W. 2005. Modulating hox gene func-
tions during animal body patterning. Nat Rev Neurosci 6:893–904.
Phelan ML, Featherstone MS. 1997. Distinct HOX N-terminal arm
residues are responsible for specificity of DNA recognition by HOX
monomers and HOX-PBX heteromers. J Biol Chem 272:8635–8643.
Ravikumar B, Duden R, Rubinsztein DC. 2002. Aggregate-prone
proteins with polyglutamine and polyalanine expansions are degraded
by autophagy. Hum Mol Genet 11:1107–1117.
Ravikumar B, Berger Z, Vacher C, O’Kane CJ, Rubinszten DC. 2006.
Rapamycin pre-treatment protects against apoptosis. Hum Mol Genet
15:1209–1216.
Remacle S, Abbas L, De Backer O, Pacico N, Gavalas A, Gofflot F,
Picard JJRezsohazy R. 2004. Loss of function but no gain of function
caused by amino acid substitutions in the hexapeptide of Hoxa1 in vivo.
Mol Cell Biol 24:8567–8575.
Shanmugam K, Featherstone MS, Saragovi HU. 1997. Residues flanking
the HOX YPWM motif contribute to cooperative interactions with
PBX. J Biol Chem 272:19081–19087.
Simeone A, Acampora D, Arcioni L, Andrews PW, Boncinelli E,
Mavilio F. 1990. Sequential activation of HOX 2 homeobox genes by reti-
noic acid in human embryonal carcinoma cells. Nature 346:763–766.
Slupsky CM, Sykes DB, Gay GL, Sykes BD. 2001. The HoxB1 hexa-
peptide is a prefolded domain: implications for the Pbx1/Hox interac-
tion. Prot Sci 10:1244–1253.
Tanaka Y, Kawahata K, Nakata T, Hirokawa A. 1992. Chronological
expression of microtubule-associated proteins (MAPs) in EC cell P19
after neural induction by retinoic acid. Brain Res 596:269–278.
Tischfield MA, Bosley TM, Salih MAM, Alorainy IA, Sener EC, Nester
MJ, Oystreck DT, Chan WM, Andrews C, Erickson RP, Engle EC.
2005. Homozygous HOXA1 mutations disrupt human brainstem,
inner ear, cardiovascular and cognitive development. Nat Genet 37:
1035–1037.
Wainwright LJ, Lasorella A, Iavarone A. 2001. Distinct mechanisms of
cell cycle arrest control the decision between differentiation and senes-
cence in human neuroblastoma cells. Proc Natl Acad Sci U S A 98:
9396–9400.
Zhang M, Kim HJ, Marshall H, Gendron-Maguire M, Lucas DA, Baron A,
Gudas T, Gridley LJ, Krumlauf R, Grippo JF. 1994. Ectopic Hoxa-1
induces rhombomere transformation in mouse hindbrain. Development
120:2431–2442.
Polyhistidine Variants of HOXA1 in Neurons 487
Journal of Neuroscience Research DOI 10.1002/jnr
... The development of these 45 tools in the last few years facilitated the interpretation of RNA-Seq data . 46 In this study, we seek to develop a web application with substantially enhanced functionality with (1) 47 automatic gene ID conversion with broad coverage, (2) comprehensive gene annotation and pathway 48 database for both plant and animals, (3) several methods for in-depth EDA and pathway analysis, (4) access 49 to web services such as KEGG [19] and STRING-db [21] via application programming interface (API), 50 and (5) improved reproducibility by generating R scripts for stand-alone analysis. 51 ...
... Figure 6). This unexpected result is consistent with Hoxa1's role in neuronal 203 differentiation [48,49]. Polymorphisms of this gene are associated with cerebellar volume in humans [50]. ...
... DEGs are also enriched with 430 genes related to neuron parts, synapse, as well as neurodegenerative diseases. This is consistent with reports 431 of Hoxa1's role in neuron differentiation [48][49][50]. Hoxa1 knockdown induces expression of genes 432 associated with the cytokine-cytokine interaction, lysosome, and cell migration, probably in response to the 433 injected siRNAs. These genes are overrepresented with target genes of NF-κB, known to be involved in 434 immune response. ...
Preprint
Full-text available
The analysis and interpretation of the RNA-Seq data can be time-consuming and challenging. We aim to streamline the bioinformatic analyses of gene-level data by developing a user-friendly web application for exploratory data analysis, differential expression, and pathway analysis. iDEP (integrated Differential Expression and Pathway analysis) seamlessly connects 63 R/Bioconductor packages, 208 annotation databases for plant and animal species, and 2 web services. The workflow can be reproduced by downloading customized R code and related files. As demonstrated by two examples, iDEP ( http://ge-lab.org/idep/ ) democratizes access to bioinformatics resources and empowers biologists to easily gain actionable insights from transcriptomic data.
... 49 Although not all IDP proteins are aggregation-prone, many HOX transcription factors clearly are susceptible. [50][51][52] PolyAla expansion in HOX proteins also leads to aggregation and retention of HOXD13 in cytoplasm. 53 Wild-type HOXD13 contains a 15 amino acid polyalanine repeat (Fig. 3). ...
... 53 Such, HOX aggregates can lead to cell death. 50 Nonsense mutations that lead to hox mRNA degradation create a haploinsufficiency, resulting in a mild synpolydactyly phenotype. 58,59 Fig. 3 Amino acid sequence schematics of HOX proteins linked to human developmental abnormalities. ...
Chapter
During animal development, HOX transcription factors determine the fate of developing tissues to generate diverse organs and appendages. The power of these proteins is striking: mis-expressing a HOX protein causes homeotic transformation of one body part into another. During development, HOX proteins interpret their cellular context through protein interactions, alternative splicing, and post-translational modifications to regulate cell proliferation, cell death, cell migration, cell differentiation, and angiogenesis. Although mutation and/or mis-expression of HOX proteins during development can be lethal, changes in HOX proteins that do not pattern vital organs can result in survivable malformations. In adults, mutation and/or mis-expression of HOX proteins disrupts their gene regulatory networks, deregulating cell behaviors and leading to arthritis and cancer. On the molecular level, HOX proteins are composed of DNA binding homeodomain, and large regions of unstructured, or intrinsically disordered, protein sequence. The primary roles of HOX proteins in arthritis and cancer suggest that mutations associated with these diseases in both the structured and disordered regions of HOX proteins can have substantial functional effects. These insights lead to new questions critical for understanding and manipulating HOX function in physiological and pathological conditions.
... Hoxa1 knockdown also induces genes related to plasma membrane, neurons and synapses (Additional file 3: Figure S7). This unexpected result is consistent with Hoxa1's role in neuronal differentiation [36,37]. Polymorphisms of this gene are associated with cerebellar volume in humans [38]. ...
... DEGs are also enriched with genes related to neuron parts, synapse, as well as neurodegenerative diseases. This is consistent with reports of Hoxa1's role in neuron differentiation [36][37][38]. Hoxa1 knockdown induces expression of genes associated with the cytokine-cytokine interaction, lysosome, and cell migration, probably in response to the injected siRNAs. These genes are overrepresented with target genes of NF-κB, known to be involved in immune response. ...
Article
Full-text available
Background RNA-seq is widely used for transcriptomic profiling, but the bioinformatics analysis of resultant data can be time-consuming and challenging, especially for biologists. We aim to streamline the bioinformatic analyses of gene-level data by developing a user-friendly, interactive web application for exploratory data analysis, differential expression, and pathway analysis. Results iDEP (integrated Differential Expression and Pathway analysis) seamlessly connects 63 R/Bioconductor packages, 2 web services, and comprehensive annotation and pathway databases for 220 plant and animal species. The workflow can be reproduced by downloading customized R code and related pathway files. As an example, we analyzed an RNA-Seq dataset of lung fibroblasts with Hoxa1 knockdown and revealed the possible roles of SP1 and E2F1 and their target genes, including microRNAs, in blocking G1/S transition. In another example, our analysis shows that in mouse B cells without functional p53, ionizing radiation activates the MYC pathway and its downstream genes involved in cell proliferation, ribosome biogenesis, and non-coding RNA metabolism. In wildtype B cells, radiation induces p53-mediated apoptosis and DNA repair while suppressing the target genes of MYC and E2F1, and leads to growth and cell cycle arrest. iDEP helps unveil the multifaceted functions of p53 and the possible involvement of several microRNAs such as miR-92a, miR-504, and miR-30a. In both examples, we validated known molecular pathways and generated novel, testable hypotheses. Conclusions Combining comprehensive analytic functionalities with massive annotation databases, iDEP (http://ge-lab.org/idep/) enables biologists to easily translate transcriptomic and proteomic data into actionable insights. Electronic supplementary material The online version of this article (10.1186/s12859-018-2486-6) contains supplementary material, which is available to authorized users.
... MEOX2 présente aussi une région riche en histidine / glutamine qui permet l'activation de la cycline kinase p21 (CIP1 / WAF1), protéine inhibitrice du cycle cellulaire, dans les cellules endothéliales vasculaires(Chen et al., 2007). Ce motif polyhistidine / polyglutamine n'est pas spécifique de MEOX2 et se retrouve dans d'autres protéines HOX(Paraguison et al., 2007). ...
Thesis
L’expression de facteurs de transcription (FT), aux rôles clés dans l’embryogénèse, semble être une caractéristique importante des gliomes et particulièrement des glioblastomes (GBM). Le FT MEOX2 n’a jamais été étudié dans les gliomes/glioblastomes et encore moins dans les cellules souches de glioblastomes (CSG) bien que ces dernières soient au cœur du caractère récidivant des gliomes. Les objectifs de mon travail de thèse étaient d’explorer les bases de données publiques pour trouver un ou plusieurs FT clés impliqués dans la survie des patients porteurs d’un gliome, pour ensuite confirmer l’impact diagnostique et pronostique du FT trouvé, MEOX2, à partir d’une cohorte locale. Le rôle de ce FT a ensuite été étudié dans les cellules souches de GBM. Parmi les gènes différentiellement exprimés entre les gliomes de grade II ou III mutés IDH1/2 et codélétés 1p/19q, ou mutés IDH1/2 non-codélétés, ou IDH1/2 sauvages, et les GBM IDH sauvages, MEOX2 était un FT négativement corrélé à la survie globale et à la survie sans progression. L’étude des bases de données publiques, et d’une cohorte interne de 112 gliomes a permis de montrer que MEOX2 était enrichi dans les gliomes les plus agressifs (GBM et gliomes de grade II ou III IDH sauvage) et était associé au statut IDH sauvage, indépendamment du statut de codélétion 1p19q. Son expression serait sous la dépendance de son nombre de copie chromosomique et de l’état de méthylation de son promoteur. L’analyse des courbes de survie a montré que la forte expression de MEOX2 était associée à un mauvais pronostic pour les patients. Concernant son rôle biologique, l’expression de MEOX2 était nucléaire et hétérogène entre les CSG. MEOX2 avait un effet anti-prolifératif via la voie des MAPK et sa perte d’expression corrélait avec la différenciation des CSG et l’acquisition de caractéristiques du lignage neuronal. L’inhibition de MEOX2 régulait un réseau de gènes restreint comprenant la cadhérine CDH10. Plus particulièrement, l’inhibition de MEOX2 corrélait avec une augmentation d’un acteur de la voie des cadhérines, CDH10, et avec une diminution de plusieurs acteurs de la voie des intégrines. MEOX2 est encore un acteur très peu connu et étudié dans la carcinogénèse et notre travail apporte une contribution à la compréhension de son rôle dans les gliomes et dans les CSG.
... Gavalas et al (34) reported that HoXa1 or HoXB1 null mutants cause developmental disorders in the hindbrain. Subsequently, in 2007, Paraguison et al (35) demonstrated that polyhistidine variants of HoXa1 lead to decreased PBX homeobox 1 activity and inhibition of neuronal differentiation. in 2008, Martinez-ceballos and Gudas (36) highlighted the necessity of HoXa1 in retinoic acid-induced embryonic stem cell differentiation to neurons. ...
Article
Full-text available
Cognitive impairment is one of the primary features of vascular dementia (VD). However, the specific mechanism underlying the regulation of cognition function in VD is not completely understood. The present study aimed to explore the effects of microRNA (miR)‑150 on VD. To determine the effects of miR‑150 on cognitive function and hippocampal neurons in VD model rats, rats were subjected to intracerebroventricular injections of miR‑150 antagomiR. The Morris water maze test results demonstrated that spatial learning ability was impaired in VD model rats compared with control rats. Moreover, compared with antagomiR negative control (NC), miR‑150 antagomiR alleviated cognitive impairment and enhanced memory ability in VD model rats. The triphenyltetrazolium chloride, Nissl staining and immunohistochemistry results further demonstrated that miR‑150 knockdown improved the activity of hippocampal neurons in VD model rats compared with the antagomiR NC group. To validate the role of miR‑150 in neurons in vitro, the PC12 cell line was used. The flow cytometry and Hoechst 33342/PI double staining results indicated that miR‑150 overexpression significantly increased cell apoptosis compared with the mimic NC group. Moreover, the dual‑luciferase reporter gene assay results indicated that miR‑150 targeted HOXA1 and negatively regulated HOXA1 expression. Therefore, the present study indicated that miR‑150 knockdown ameliorated VD symptoms by upregulating HOXA1 expression in vivo and in vitro.
... Many HOX genes function in the development of embryonic brain 18 . The HOXA1 gene has been linked to brainstem and cerebellum development 19,20 . Furthermore, miR-339-5p has been predicted to be a downstream miRNA of long non-coding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1) through in silico analysis. ...
Article
Full-text available
Hypoxic-ischemic brain damage (HIBD) is a major cause of fatality and morbidity in neonates. However, current treatment approaches to alleviate HIBD are not effective. Various studies have highlighted the role of microRNAs (miRNAs) in various biological functions in multiple diseases. This study investigated the role of miR-339-5p in HIBD progression. Neonatal HIBD mouse model was induced by ligation of the right common carotid artery. Neuronal cell model exposed to oxygen-glucose deprivation (OGD) was also established. The miR-339-5p expression in mouse brain tissues and neuronal cells was quantified and the effects of miR-339-5p on neuronal cell activity and apoptosis induced by hypoxia-ischemia were explored. The overexpression or knockdown of long non-coding RNA (lncRNA) nuclear-enriched abundant transcript 1 (NEAT1) in hippocampal neurons was used to determine the effect of lncRNA NEAT1 on the expression of miR-339-5p and homeobox A1 (HOXA1) and apoptosis. Short hairpin RNA targeting lncRNA NEAT1 and miR-339-5p antagomir were used in neonatal HIBD mice to identify their roles in HIBD. Our results revealed that miR-339-5p was downregulated in neonatal HIBD mice and neuronal cells exposed to OGD. Downregulated miR-339-5p promoted neuronal cell viability and suppressed apoptosis during hypoxia-ischemia. Moreover, lncRNA NEAT1 competitively bind to miR-339-5p to increase HOXA1 expression and inhibited neuronal cell apoptosis under hypoxic-ischemic conditions. The key observations of the current study present evidence demonstrating that lncRNA NEAT1 upregulated HOXA1 to alleviate HIBD in mice by binding to miR-339-5p.
... Beside the HD and HX regions, a specific feature of HOXA1 resides in two stretches of histidines (11-His and 5-His repeats) at positions 60-70 and 138-142 of the HOXA1 sequence, respectively (Fig. 1B). Sequence or length modification in the 11-His repeat was associated to different diseases [36,37] and we have recently shown that the 11-His motif is important for the transcriptionindependent HOXA1-mediated activation of NF-κB [25]. Six deletion derivatives of HOXA1 were generated (Fig. 1B) to identify protein parts required for the interaction with PRDM14: HOXA1 ΔNter was deleted of its 40 N-terminal residues, HOXA1 Δ11His was deleted of its 11-His repeat, HOXA1 Δ11-5His was deleted from the 11-His to the 5-His motifs, HOXA1 Δ71-199 lacks the central region extending from the 11-His repeat to the hexapeptide, HOXA1 HD+Cter only contains the homeodomain and the C-terminal extension of the protein, and HOXA1 ΔHD was deleted of the complete homeodomain. ...
Article
Understanding how the activity of transcription factors like HOX proteins is regulated remains a widely open question. In a recent screen for proteins interacting with HOXA1, we identified a PRDM protein family member, PRDM14, which is known to be transiently co-expressed with HOXA1 in epiblast cells before their specification towards somatic versus germ cell fate. Here, we confirm PRDM14 is an interactor of HOXA1 and we identify the homeodomain of HOXA1 as well as the PR domain and Zinc fingers of PRDM14 to be required for the interaction. An 11-His repeat of HOXA1 previously highlighted to contribute to HOXA1-mediated protein-protein interactions is also involved. At a functional level, we provide evidence that HOXA1 displays an unexpectedly long half-life and demonstrate that PRDM14 can reduce the stability and affect the transcriptional activity of HOXA1.
... Previous studies showed that neuronal development in mice required high expression of Pbx1a rather than Pbx1b (Paraguison et al. 2007;Golonzhka et al. 2015;Linares et al. 2015), supporting the finding of no PBX1b expression in NPCs. Additionally, we did not observe significantly different expression of the total PBX1 and three other PBX family members across cell types ( Figure 5H, fold change < 2). ...
Article
Full-text available
Understanding embryonic stem cell (ESC) fate decision between self-renewal and proper differentiation is important for developmental biology and regenerative medicine. Attentions have focused on underlying mechanisms involving histone modifications (HMs), alternative pre-mRNA splicing (AS), and cell-cycle progression. However, their intricate interrelations and joint contributions to ESC fate decision remain unclear. We performed integrative analyses on transcriptomic and epigenomic data derived from human ESC (hESC) and five types of differentiated cells. We identified thousands of AS exons and revealed their development- and lineage-dependent characterizations. Following the observation that dynamic HM changes predominantly occur in AS exons upon hESCs differentiation, we identified 3 of 16 investigated HMs (H3K36me3, H3K27ac, and H4K8ac) that are strongly associated with 52.8% of hESC differentiation-related AS events. Further analyses showed that the HM-associated AS genes predominantly function in G2/M phases and ATM/ATR-mediated DNA damage response pathway for cell differentiation, whereas HM-unassociated AS genes enrich in G1 phase and pathways for self-renewal. These results imply a potential epigenetic mechanism by which some HMs contribute to ESC fate decision through the AS regulation of specific pathways and cell-cycle genes. We exemplified the potential mechanism by a cell cycle-related transcription factor, PBX1, which regulates the pluripotency regulatory network by binding to NANOG. We found that the isoform switch between PBX1a and PBX1b is strongly associated with H3K36me3 alteration and implicated in hESC fate determination. Supported by extended dataset from Roadmap/ENCODE projects, we identified the alternative splicing of PBX1 as a novel candidate linking H3K36me3 to embryonic stem cell fate decision.
... These repeats are conserved among HOXA1 orthologs, although the size of the longest His repeat varies, the mouse protein containing 11 and 5 His tracts, respectively. The function of these His repeats remains unexplored, but His repeat length variations have been reported in cases of autism in the Japanese population and in vitro expression of expanded His-repeat variants of HOXA1 resulted in a loss of cooperative binding with PBX1, leading to nuclear protein aggregation, impaired neuronal differentiation and cell death (17)(18)(19). His deletion within the HOXA1 sequence has also been found in ventricular septal defect patients (20). Together, these results suggest that these His repeats critically influence the function of the HOXA1 protein. ...
Article
Full-text available
HOX proteins define a family of key transcription factors regulating animal embryogenesis. HOX genes have also been linked to oncogenesis and HOXA1 has been described to be active in several cancers, including breast cancer. Through a proteome-wide interaction screening, we previously identified the TNFR-associated proteins RBCK1/HOIL-1 and TRAF2 as HOXA1 interactors suggesting that HOXA1 is functionally linked to the TNF/NF-κB signaling pathway. Here, we reveal a strong positive correlation between expression of HOXA1 and of members of the TNF/NF-κB pathway in breast tumor datasets. Functionally, we demonstrate that HOXA1 can activate NF-κB and operates upstream of the NF-κB inhibitor IκB. Consistently, we next demonstrate that the HOXA1-mediated activation of NF-κB is non-transcriptional and that RBCK1 and TRAF2 influences on NF-κB are epistatic to HOXA1. We also identify an 11 Histidine repeat and the homeodomain of HOXA1 to be required both for RBCK1 and TRAF2 interaction and NF-κB stimulation. Finally, we highlight that activation of NF-κB is crucial for HOXA1 oncogenic activity.
Article
HOXA1 belongs to the HOX family of transcription factors which are key regulators of animal development. Little is known about the molecular pathways controlling HOXA1. Recent data from our group revealed distinct partner proteins interacting with HOXA1. Among them, OGT is an O-linked N-acetylglucosamine (O-GlcNAc) transferase modifying a variety of proteins involved in different cellular processes including transcription. Here, we confirm OGT as a HOXA1 interactor, we characterise which domains of HOXA1 and OGT are required for the interaction, and we provide evidence that OGT post-translationally modifies HOXA1. Mass spectrometry experiments indeed reveal that HOXA1 can be phosphorylated on the AGGTVGSPQYIHHSY peptide and that upon OGT expression, the phosphate adduct is replaced by an O-GlcNAc group. This article is protected by copyright. All rights reserved.
Article
Full-text available
The analysis of Hoxa1 and Hoxb1 null mutants suggested that these genes are involved in distinct aspects of hindbrain segmentation and specification. Here we investigate the possible functional synergy of the two genes. The generation of Hoxa1(3′RARE)/Hoxb1(3′RARE) compound mutants resulted in mild facial motor nerve defects reminiscent of those present in the Hoxb1 null mutants. Strong genetic interactions between Hoxa1 and Hoxb1 were uncovered by introducing the Hoxb1(3′RARE) and Hoxb1 null mutations into the Hoxa1 null genetic background. Hoxa1(null)/Hoxb1(3′RARE) and Hoxa1(null)/Hoxb1(null)double homozygous embryos showed additional patterning defects in the r4-r6 region but maintained a molecularly distinct r4-like territory. Neurofilament staining and retrograde labelling of motor neurons indicated that Hoxa1 and Hoxb1 synergise in patterning the VIIth through XIth cranial nerves. The second arch expression of neural crest cell markers was abolished or dramatically reduced, suggesting a defect in this cell population. Strikingly, the second arch of the double mutant embryos involuted by 10.5 dpc and this resulted in loss of all second arch-derived elements and complete disruption of external and middle ear development. Additional defects, most notably the lack of tympanic ring, were found in first arch-derived elements, suggesting that interactions between first and second arch take place during development. Taken together, our results unveil an extensive functional synergy between Hoxa1 and Hoxb1 that was not anticipated from the phenotypes of the simple null mutants.
Article
Full-text available
The Hox-1.6 gene disrupted in embryonic stem cells by homologous recombination was introduced into the mouse germline. Heterozygous mice were normal, but homozygous mice died at birth from anoxia and had numerous defects that were centered at the level of rhombomeres 4 to 7 and included delayed hindbrain neural tube closure, absence of certain cranial nerves and ganglia, and malformed inner ears and bones of the skull. Thus, Hox-1.6 is involved in regional specification along the rostrocaudal axis, but only in its most rostral domain of expression. Hox-1.6 appears to specify neurogenic neural crest cells prior to specification of mesenchymal neural crest cells by Hox-1.5. Thus, within the same region of the presumptive hindbrain, two HOX-1 genes are involved in the patterning of two different populations of neural crest cells. The implication of these results for the function of the Hox network during mouse embryogenesis is discussed.
Article
Full-text available
Targeted disruption of the murine hox-A1 gene results in severe defects in the formation of the hindbrain and associated cranial ganglia and nerves. Carbocyanine dye injections were used to trace afferent and efferent projections to and from the hindbrain in hox-A1-/hox-A1- mutant mice. Defects were observed in the position of efferent neurons in the hindbrain and in their projection patterns. In situ hybridization was used to analyze the transcription pattern of genes expressed within specific rhombomeres. Krox-20, int-2 (fgf-3), and hox-B1 all display aberrant patterns of expression in hox-A1- mutant embryos. The observed morphological and molecular defects suggest that there are changes in the formation of the hindbrain extending from rhombomere 3 through rhombomere 8 including the absence of rhombomere 5. Also, motor neurons identified by their axon projection patterns which would normally be present in the missing rhombomere appear to be respecified to or migrate into adjacent rhombomeres, suggesting a role for hox-A1 in the specification of cell identity and/or cell migration in the hindbrain.
Article
Pluripotent murine embryonal carcinoma (EC) P19 cells are induced at a high rate into neural cells using retinoic acid and serum-free medium. EM observation revealed great increase of microtubules (MTs) after neuronal induction. To study the expression of microtubule-associated proteins (MAPs), immunoblotting and immunocytochemistry were performed with phosphorylated MAP1B (pMAP1B)-, MAP2-, and MAP1A-specific monoclonal antibodies. They did not stain undifferentiated cells. Early MAPs (pMAP1B and MAP2C) appeared 12 h after the neuronal induction, changing to late MAPs (MAP1A and MAP2A/B) at 3–5 days. These expression patterns are quite similar to those of neural cells in vivo. Anti-pMAP1B stained not only neurites but also the cell body and varicosities. But after extraction of the soluble component by permeabilization, pMAP1B was found in only MT-domains of the neurites at LM and EM levels, indicating that some part of pMAP1B is a structural component of neurite MTs and others exist in a soluble form. After culturing for more than 5 days, pMAP1B disappeared from the soma, but still remained in the distal ends of neurites. Here we showed that P19 is a good model system for studying the expression of MAPs on the continuous course of neuronal differentiation.
Article
The ability to use a vital cell marker to study mouse embryogenesis will open new avenues of experimental research. Recently, the use of transgenic mice, containing multiple copies of the jellyfish gene encoding the green fluorescent protein (GFP), has begun to realize this potential. Here, we show that the fluorescent signals produced by single-copy, targeted GFP in-frame fusions with two different murine Hox genes, Hoxa1 and Hoxc13, are readily detectable by using confocal microscopy. Since Hoxa1 is expressed early and Hoxc13 is expressed late in mouse embryogenesis, this study shows that single-copy GFP gene fusions can be used through most of mouse embryogenesis. Previously, targeted lacZ gene fusions have been very useful for analyzing mouse mutants. Use of GFP gene fusions extends the benefits of targeted lacZ gene fusions by providing the additional utility of a vital marker. Our analysis of the Hoxc13(GFPneo) embryos reveals GFP expression in each of the sites expected from analysis of Hoxc13(lacZneo) embryos. Similarly, Hoxa1(GFPneo) expression was detected in all of the sites predicted from RNA in situ analysis. GFP expression in the foregut pocket of Hoxa1(GFPneo) embryos suggests a role for Hoxa1 in foregut-mediated differentiation of the cardiogenic mesoderm. journal article
Article
Gene targeting in mouse embryo-derived stem cells has been used to generate mice with a disruption in the homeobox gene Hox-1.6. Mice heterozygous at the Hox-1.6 locus appear normal, whereas Hox-1.6-/Hox-1.6- mice die at or shortly after birth. These homozygotes exhibit profound defects in the formation of the external, middle and inner ears as well as in specific hindbrain nuclei, and in cranial nerves and ganglia. The affected tissues lie within a narrow region along the anteroposterior axis of the mouse but are of diverse embryonic origin. The set of defects associated with the disruption of Hox-1.6 is distinct from and nonoverlapping with that of the closely linked Hox-1.5 gene. But both mutations cause loss, rather than homeotic transformation, of tissues and structures.
Article
Morphogens, i.e., endogenous, diffusible molecules which induce pathways of differentiation through a gradient of concentration, play a key role in vertebrate development. Retinoic acid (RA) is a natural morphogen in chicken development, where it specifies the limb anteroposterior (AP) axis (Tickle et al. 1982; Thaller and Eichele 1987, 1988; Brockes 1989), and possibly in frogs, where alteration of intraembryonic RA levels dramatically affects the AP polarity of the developing CNS (Durston et al., 1989). The identification of three nuclear RA receptors in mouse and man (Petkovich et al. 1987; Giguere et al. 1987; Brand et al. 1988; Zelent et al. 1989) supports the concept that the morphogens act as intracellular signal molecules with direct gene control functions. Although the genes involved in transducing signals provided by morphogens are still unknown, homeobox genes, which specify positional information in Drosophila and possibly in vertebrate embryogenesis (Gehring 1987; Ingham 1988; Holland and Hogan 1988), are among the suggested candidates (Brockes 1989; de Robertis et al. 1989).
Article
RETINOIC acid had been implicated as a natural morphogen in chicken and frog embryogenesis, and is presumed to act through the gene regulatory activity of a family of nuclear receptors. Homeobox genes, which specify positional information in Drosophila and possibly in vertebrate embryogenesis, are among the candidate responsive genes. We previously reported that retinoic acid specifically induces human homeobox gene (HOX) expression in the embryonal carcinoma cell line NT2/D1. We now show that the nine genes of the HOX2 cluster are differentially activated in NT2/D1 cells exposed to retinoic acid concentrations ranging from 10(-8) to 10(-5) M. Genes located in the 3' half of the cluster are induced at peak levels by 10(-8) M retinoic acid, whereas a concentration of 10(-6) to 10(-5) M is required to fully activate 5' genes. At both high and low retinoic acid concentrations, HOX2 genes are sequentially activated in embryonal carcinoma cells in the 3' to 5' direction.
Article
The embryonal carcinoma line C17-S1 clone 1003 is multipotential in vivo. When the cells are grown in vitro in serum-containing medium most of them remain undifferentiated, while a few differentiate into a unique morphologic type of epithelioid cell. If 1003 cells are passaged into a defined medium containing insulin, transferrin, selenium, and fibronectin they grow for six to eight generations at the same rate as in serum-containing medium. During this time, all the cells of the culture differentiate into a limited number of phenotypes with neuroepithelial and neuronal cells predominating. Differentiation could be obtained in the defined medium at relatively low cell densities. Exogenous fibronectin is required for cell attachment to the substratum, and when absent the cells form aggregates in which differentiation still occurs. Low amounts of serum added to the defined medium allow multiplication and maintenance of cells of undifferentiated phenotype and prevent differentiation into neuronal cells.