Accepted Manuscript TDP-43 proteolysis is associated with astrocyte reactivity after traumatic brain injury in rodents
Chih-Yuan Huang, Yi-Che Lee, Ping-Chia Li, Po-Chou Liliang, Kang Lu, Kuo-Wei Wang, Li-Ching Chang, Li-Yen Shiu, MingFeng Chen, Yuan-Ting Sun, Hao-kuang Wang PII: DOI: Reference:
S0165-5728(17)30394-6 doi:10.1016/j.jneuroim.2017.10.011 JNI 476647
To appear in:
Journal of Neuroimmunology
Received date: Revised date: Accepted date:
30 August 2017 14 October 2017 16 October 2017
Please cite this article as: Chih-Yuan Huang, Yi-Che Lee, Ping-Chia Li, Po-Chou Liliang, Kang Lu, Kuo-Wei Wang, Li-Ching Chang, Li-Yen Shiu, Ming-Feng Chen, Yuan-Ting Sun, Hao-kuang Wang , TDP-43 proteolysis is associated with astrocyte reactivity after traumatic brain injury in rodents. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Jni(2017), doi:10.1016/ j.jneuroim.2017.10.011
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT
Research report TDP-43 proteolysis is associated with astrocyte reactivity after traumatic brain injury in rodents
PT
Abbreviated title: TDP-43 proteolysis is associated with reactive astrocyte
Chih-Yuan Huang,1 Yi-Che Lee,2 Ping-Chia Li,3 Po-Chou Liliang,3,4 Kang Lu,3,4
RI
Kuo-Wei Wang,3,4 Li-Ching Chang,3 Li-Yen Shiu,5 Ming-Feng Chen,5 Yuan-Ting Sun,6
Neurosurgical Service, Department of Surgery, National Cheng Kung University
NU
1
SC
and Hao-kuang Wang3,4*
MA
Hospital, Tainan, Taiwan
Department of Nephrology, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
3
School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan.
4
Department of Neurosurgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
5
Department of Medical Research, Cell Therapy and Research Center, E-Da Hospital,
EP T
ED
2
I-shou University, Kaohsiung, Taiwan. Department of Neurology, National Cheng Kung University Hospital, College of
AC C
6
Medicine, National Cheng Kung University, Tainan, Taiwan
Address correspondence to:
Hao kuang Wang, MD, PhD Department of Neurosurgery, E-Da Hospital/ I-Shou University, Taiwan No.1, Yida Road, Jiaosu Village, Yanchao District, Kaohsiung City 82445, Taiwan
ACCEPTED MANUSCRIPT
Tel: +866-975106080 E-mail:
[email protected],
[email protected]
Word count for abstract: 162, Number of words: 3089
PT
Number of references: 27, Number of figures: 6
RI
Keywords: astrocyte; astrogliosis; traumatic brain injury; transactivation response
AC C
EP T
ED
MA
NU
SC
DNA-binding protein 43.
ACCEPTED MANUSCRIPT
Abbreviations: Amyotrophic lateral sclerosis
ANOVA
A two-way analysis of variance
FTLD GFAP
Frontotemporal lobar degeneration Glial fibrillary acidic protein
TBI
Traumatic brain injury
TDP-43 TX
Transactivation response DNA-binding protein 43 High-salt Triton X-100
SARK
Sarkosyl
z-DEVD-fmk
N-benzyloxycarbonyl-Asp-Glu-Val-Asp- fluoromethyl ketone
AC C
EP T
ED
MA
NU
SC
RI
PT
ALS
ACCEPTED MANUSCRIPT
Abstract The aggregation and deposition of transactivation response DNA-binding protein 43 (TDP-43) in neurons and astrocytes is characteristic in a number of neurodegenerative diseases including Alzheimer ’s disease, frontotemporal lobar degeneration, and
PT
amyotrophic lateral sclerosis. Nevertheless, the exact role of TDP-43 in astrocytes is unknown. Recently, TDP-43 was identified in neurons but not astrocytes after traumatic
RI
brain injury (TBI) in humans. In the present study, we evaluated TDP-43 expression and
SC
proteolysis in astrocytes in a rat model of TBI. We assessed TDP-43 fragment expression, astrocyte morphology, neuronal population numbers, and motor function
NU
after TBI with or without intracerebroventricular administration of a caspase-3 inhibitor.
MA
Motor dysfunction was observed after TBI in potential association astrocytic TDP-43 short fragment mislocalization and accumulation, astrogliosis, and neuronal loss.
ED
Notably, caspase-3 inhibition prevented these changes after TBI. Our findings suggest that TDP-43 proteolysis in astrocytes is related to astrogliosis and subsequent neuronal
EP T
loss in TBI, and that TDP-43 may be an important therapeutic target for preventing
AC C
motor dysfunction after TBI.
ACCEPTED MANUSCRIPT
1. Introduction Transactive response DNA binding protein (TDP-43) is a heterogeneous nuclear RNA-binding protein that regulates gene expression, transcription, and multiple aspects of RNA processing and functions including splicing, stability, transport, translation, and
PT
microRNA maturation (Sieben et al., 2012; Al-Chalabi et al., 2012; Gendron et al., 2013). Under physiological conditions, TDP-43 is enriched in the nucleus; however,
RI
TDP-43 can be cleaved by caspase-3 to generate short 25-kDa and 35-kDa C-terminal
SC
fragments, resulting in the formation of ubiquitin-positive cytoplasmic inclusions, cellular toxicity, and ultimately death (Zhang et al., 2009; Barmada et al., 2010; Che et
NU
al., 2011). TDP-43 was first identified in neuronal cytoplasmic inclusions in
MA
frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS) in 2006 (Sieben et al., 2012; Al-Chalabi et al., 2012). Recently pathological TDP-43
ED
inclusions were also identified in Alzheimer’s disease (Fang et al., 2014). Thus, TDP-43 accumulation and deposition is presumed to play a role in the pathogenesis of
EP T
neurodegenerative diseases.
Traumatic brain injury (TBI) is a leading cause of death and disability that has a
AC C
limited number of clinical treatment options (Maas et al., 2008). TBI leads to the activation of multiple inflammatory pathways including astrocyte activation (Blennow et al., 2012). Whereas astrocytes play neurotrophic and supportive roles in nervous tissues under physiological conditions (Pekny and Pekna, 2014; Pekny and Wilhelmsson, 2014), astrocytes are subject to hypertrophy and proliferation after TBI that can lead to a number of positive (e.g., neural protection and repair) and negative (e.g., glial scarring and alterations in plasticity) effects; yet, the role of astrocytes after TBI is not well understood. Moreover, few studies have investigated a role for TDP-43 in TBI.
ACCEPTED MANUSCRIPT
Nuclear depletion and cytoplasmic aggregation of TDP-43 has been documented in astrocytes; however, the toxicity of TDP-43 fragments in astrocytes is different from that observed in neurons (Haidet-Phillips et al., 2013; Serio et al., 2013; Tong et al., 2013; Rojas et al., 2014). Haidet-Phillips et al. showed that TDP-43 overexpression or
PT
knockout in astrocytes did not produce a cytotoxic phenotype (Haidet-Phillips et al., 2013). Additionally, it is not clear as to whether pathogenic TDP-43 accumulation in
RI
astrocytes causes non-cell-autonomous neuronal death (Haidet-Phillips et al., 2013;
SC
Serio et al., 2013; Tong et al., 2013; Rojas et al., 2014); whereas some evidence suggests that astrocytes expressing mutated forms of TDP43 can cause the
NU
non-cell-autonomous death of motor neurons (Tong et al., 2013; Rojas et al., 2014),
MA
other evidence contradicts these findings (Haidet-Phillips et al., 2013; Serio et al., 2013).
ED
In the present study, we investigated whether TBI was associated with alterations in TDP-43 expression and cleavage in astrocytes using a rodent model. Rats were
EP T
treated with a caspase-3 inhibitor or vehicle immediately after TBI and TDP-43 fragment expression, astrocyte morphology, neuronal population numbers, and motor
AC C
function were examined at up to 7 days post-injury.
ACCEPTED MANUSCRIPT
2. Materials and methods 2.1 Ethical statement All procedures were reviewed and approved by the Institution Animal Care and
PT
Use Committee(IACUC) of E-Da hospital (IACUC), and conformed to the guidelines of
SC
RI
the Taiwan Council for Animal Care. The number of IACUC is 104004.
NU
2.2 Study design
Adult male Sprague-Dawley rats weighing 300–350 g were purchased from
MA
Charles River Laboratories. Then those rats were maintained at constant temperature (22 ± 2°C) and a 12 hour light/dark cycle with food and water available ad libitum for 1
ED
week before procedures. Details on the injury device, TBI model, and behavioral tests
EP T
were previously reported (Wang et al., 2015). Table 1 showing the time-line diagram of TBI with experimental procedure. Finally, animals were divided into three groups to examine whether TBI induced the cleavage and aggregation of TDP-43 species in lesion
AC C
sites and specifically in astrocytes.
2.3 Experimental animals 2.3.1 Traumatic brain injury model. TBI was induced using a weight drop device as previously established in the Feeney weight-drop model (Feeney et al., 1981, Liliang et al., 2010; Wang et al., 2015).
ACCEPTED MANUSCRIPT
Briefly, rats were anesthetized, the skull was exposed, and a left partial craniotomy was performed. A 40-g steel rod with a flat end was freely dropped from 30 cm onto strike a transducer rod (diameter, 4.5 mm) that was oriented with its tip placed directly onto the rat’s skull, posterior to bregma (Feeney et al., 1981, Liliang et al., 2010; Wang et al.,
PT
2015). Rats were anesthetized and underwent left parietal craniotomy without cortical contusion injury. After postoperative procedures, rats were returned to their home cages
SC
RI
and monitored carefully.
2.3.2 Intracerebroventricular administration of a caspase-3 inhibitor.
NU
Rats were placed in a stereotaxic apparatus for intracerebroventricular
MA
administration. The cell-permeable and irreversible caspase-3 inhibitor N-benzyloxycarbonyl-Asp-Glu-Val- Asp- fluoromethyl ketone (EMD Millipore, Billerica, MA) was dissolved in 0.2% DMSO (500 ng in 1 μL of DMSO) (Clark et al., 2000;
ED
Knoblach et al., 2004). The caspase-3 inhibitor (2 μL) or vehicle (2 μL) was
EP T
administered by intracerebroventricular injection ipsilateral to the lesion site at 10 min
AC C
post-TBI via a micro-osmotic pump and infusion cannula.
2.4 Experimental procedures 2.4.1 Western blotting.
To evaluate protein expression levels in the cerebral cortical regions surrounding TBI lesions, protein extracts were obtained by homogenization of cortical tissues in RIPA lysis buffer (Clark et al., 2000; Knoblach et al., 2004; Wang et al., 2012), samples were electrophoresed and transferred to blotting membranes, and proteins were probed
ACCEPTED MANUSCRIPT
with the following primary antibodies: anti-TDP-43 (1:5000; Proteintech), anti-caspase-3 (1:2000; Cell Signaling), and anti-actin (1:15,000; Millipore). The anti-TDP-43 antibody used in this study recognized not only the full- length TDP-43 product but also 25–35 kDa cleavage products and phosphorylated forms of TDP-43.
PT
The anti-caspase-3 antibody recognized the activated form caspase-3 resulting from
expressed as the mean ± standard error of the mean.
RI
cleavage at Asp175. The relative intensities of bands were normalized to actin and
SC
For the analysis of TDP-43 in cellular inclusions, brain tissues were dissected and
NU
proteins were extracted with specialized buffers (Wang et al., 2012). Low-salt buffer, high-salt Triton X-100 buffer, myelin floatation buffer, and sarkosyl buffer were used to
MA
extract proteins from tissues at 5 mL/g (volume/weight). Sarkosyl-insoluble materials were further extracted in 0.25 mL/g urea buffer. Urea-soluble proteins were analyzed by
ED
Western blot analysis with the above primary antibodies.
EP T
2.4.2 Immunohistochemistry.
For immunohistostaining, brain sections were stained with anti–TDP-43 (1:50;
AC C
Gene Tex), glial fibrillary acidic protein (GFAP) (1:100, Abcam),anti–caspase-3 (1:100;Cell Signaling) or antineuron-specific nuclear protein (NeuN) (Millipore), and then with biotin-conjugated secondary antibodies (Millipore) followed by detection with 3,3′ diaminobenzidine (DAB) (Millipore) (Wang et al., 2012). For immunofluorescence staining, coronal brain sections were stained with anti-LC3 antibody (1:200, Novus Biologicals). DAPI staining was used to locate the nuclei. 2.4.3 Motor function assessments.
ACCEPTED MANUSCRIPT
We used the beam-walking test to evaluate the recovery of motor function after TBI (Loane et al. 2009; Wang et al. 2012). A basal level of competence (< 10 faults over 50 steps) was established before surgery. Then, the beam walk task was performed at 1, 3, and 7 days post-injury.
PT
The rotarod test was used to assess the motor coordination after TBI (Loane et al.,
RI
2009; Wang et al., 2014). Each rat received a training session on the rotarod apparatus at a constant speed of 8 rpm before surgery. Test trials were conducted at 1, 3, and 7 days
SC
post-injury.
NU
2.4.4 Flow cytometry
MA
For quantifying the survival of neuronal number, whole ipsilateral semi-brain images were acquired by TissueFAXS and analyzed by TissueQuest (TissueGnostics,
ED
Viena, Austria) as previously described (Wang et al., 2012). Stereologic evaluation of ipsilateral semi-brain was conducted on every 12th section throughout the forebrain
EP T
with the damaged region by the Cavalieri principle. Flow cytometry- like data (dot-plot) were analyzed and organized by TissueQuest, with each dot on the plot representing
AC C
intensity of NeuN and Dapi from a single cell. The plot accumulated the values of all cells from the semi-brain, and then the positive signal number could be identified by gating the immunoreactive intensity. Neuronal cell counts were comparable between the sham and caspase-3 inhibitor-treated TBI groups.
2.5 Housing and husbandry Rats were housed in the Animal Center of I-Shou University, Kaohsiung, Taiwan
ACCEPTED MANUSCRIPT
and maintained on a 12:12-h light/dark cycle (lights on at 07:00) in a room with controlled temperature (22 ± 2°C) and humidity (60 ± 5%), with ad libitum access to food and water. Rats were allowed to acclimate for at least 1 week before the
PT
experimental sessions.
RI
2.6 Sample size
SC
Adult male Sprague-Dawley rats weighing 300–350 g were purchased from
NU
Charles River Laboratories (Wang et al., 2015). Rats were randomly divided into 3 groups: TBI with vehicle treatment (TBI, n = 27), TBI with caspase-3 inhibitor
MA
treatment (TBI + CI, n = 12), and sham group (sham, n = 5) (Figure 1A). In TBI group, an equal amount of saline was injected. In sham group, no procedure was performed.
EP T
ED
All the experiments were conducted and analyzed in blinded manner.
2.7 Allocating animals to experimental groups
AC C
Cage randomization was used in our study. On the day of surgery, a cage will be selected randomly from the pool of all cages containing animals eligible for inclusion in the study, regardless of arrival date. Primary randomization is conducted by an individual other than the surgeon. On the basis of their position on the rack, cages are given a temporary numerical designation for the purpose of primary randomization.
2.8 Experimental outcomes
ACCEPTED MANUSCRIPT
The primary endpoints of the present study were the cleavage and aggregation of TDP-43 species in lesion sites and specifically in astrocytes. Then we assessed TDP-43 fragment expression, astrocyte morphology, neuronal population numbers, and motor function after TBI with or without intracerebroventricular administration of a caspase-3
RI
PT
inhibitor.
SC
2.9 Statistical methods
NU
Results are reported as the mean ± standard error of the mean. To examine whether the data is normally distributed, D`Agostino and Pearson omnibus normality test was
MA
used before single mean or multiple mean comparison analysis. All data were confirmed that were normally distributed. Statistical differences were analyzed using a 2-way
ED
analysis of variance (ANOVA) followed by post hoc Bonferroni tests for multiple comparisons. For 2-group comparisons, data were analyzed using unpaired t-tests.
AC C
EP T
Differences were considered to be statistically significant when P < 0.05.
ACCEPTED MANUSCRIPT
3. Results 3.1 Cleavage products of TDP-43 are increased in astrocytes after TBI Whole-cell extracts from lesion sites were immunoblotted to investigate changes in TDP-43 expression post-injury. Levels of the 25-kDa and 35-kDa fragments of TDP-43
PT
were increased whereas full- length TDP-43 (43 kDa) was decreased at 7 days post-injury (Figure 2A). Additionally, glial fibrillary acidic protein (GFAP) was
RI
co-localized with TDP-43 and its cleavage products in morphologically activated
SC
astrocytes at 7 days post-injury (Figure 2B). Next, we examined urea-soluble fractions of forebrain extracts to investigate the presence of cytoplasmic TDP-43 inclusions.
NU
Cleavage products including the 25-kDa and 35-kDa fragments of TDP-43 were
MA
upregulated at 7 days post-injury (Figure 2C). These data suggested that TBI induced the cleavage and aggregation of TDP-43 species in lesion sites and specifically in
ED
astrocytes.
EP T
3.2 Caspase-3 is activated in astrocytes after TBI Next, we performed immunoblotting to quantify the expression levels of activated
AC C
caspase-3 in cortical extracts post-injury. Activated caspase-3 expression was increased in injured cortex extracts at 3 and 7 days post-injury (Figure 3A). Additionally, GFAP was co-localized with activated caspase-3 in morphologically activated astrocytes at 7 days post-injury (Figure 3B). Thus, it was concluded that TBI induced caspase-3 activation in astrocytes.
3.3 Caspase-3 inhibitor treatment decreases TDP-43 cleavage and astrocyte activation after TBI
ACCEPTED MANUSCRIPT
Intracerebroventricular administration of a caspase-3 inhibitor immediately after TBI resulted in a 50% reduction in expression of the 35-kDa fragment of TDP-43 (Figure 4A) and a 30% reduction in expression of activated caspase-3 in injured cortex extracts at 7 days post-injury (Figure 4B); however, there was no obvious effect on the
PT
25-kDa fragment of TDP-43. Moreover, caspase-3 inhibitor treatment prevented the development of astrogliosis after TBI: non-reactive astrocyte morphology was observed
SC
RI
in inhibitor-treated animals at 7 days post-injury (Figure 4C).
3.4 Motor dysfunction after TBI is sensitive to caspase-3 inhibition
NU
To assess motor function, rotarod testing and beam walk testing were performed at
MA
1, 3, and 7 days post-injury. Rotarod test latencies (Figure 5A) and beam walking latencies (Figure 5B) were significantly shorter in TBI rats compared to rats in the sham
ED
group or TBI rats treated with the caspase-3 inhibitor. Moreover, we analyzed foot placement accuracy (number of errors) and found that TBI rats treated with the
EP T
caspase-3 inhibitor performed significantly better on the right (injury) side than did untreated TBI rats (Figure 5C). However, no between-group differences were observed
AC C
for the left side. These results suggested that caspase-3 inhibition prevented the short-term impairment of motor coordination and balance after TBI.
3.5 Caspase-3 inhibition promotes neuronal survival after TBI To investigate whether neuronal death after TBI was sensitive to caspase-3 inhibition and potentially related to the presence of TDP-43 cleavage products in reactive astrocytes, we conducted immunohistochemistry staining for NeuN and Dapi and quantified the neuronal number by a flow cytometry- like analysis system
ACCEPTED MANUSCRIPT
(TissueFAX and TissueQuest) in the ipsilateral semi-brain at 7 days after TBI dmage. Neuronal cell counts were comparable between the sham and caspase-3 inhibitor-treated TBI groups by TissueQuest-based dot plot (Figure 6A). The quantified result indicated that TBI significantly leaded neuronal loss in the ipsilateral semi-brain at 7 days after
PT
TBI, but caspase-3 inhibitor improved the neuronal survival of TBI rats, a higher level of survived neuron were observed in caspase-3 inhibitor treated TBI rats (n = 5 rats per
AC C
EP T
ED
MA
NU
SC
RI
group).
ACCEPTED MANUSCRIPT
4. Discussion The present study demonstrates highlights a potential relationship between TDP-43 and astrogliosis after TBI. First, we found that TDP-43 cleavage products were increased after TBI (potentially owing to caspase-3 activation) in a manner associated
PT
with astrocytic hypertrophy. Second, rats developed severe, short-term impairments in motor coordination and balance after TBI. These behavioral impairments were possibly
RI
associated with TDP-43 short fragment mislocalization and accumulation in astrocytes.
SC
Third, intracerebroventricular treatment with a caspase-3 inhibitor immediately after TBI decreased the abundance of TDP-43 cleavage products, prevented astrocytic
NU
hypertrophy as well as motor impairments, and promoted neuronal survival. These
MA
results suggest that TDP-43 cleavage and short fragment mislocalization are pathogenic components of TBI, and that TDP-43 proteolysis is possibly related to astrogliosis after
ED
TBI.
TDP-43 plays a role in the repression of gene transcription and translation as well
EP T
as multiple aspects of RNA processing and function. In pathological contexts, TDP-43 can be cleaved, phosphorylated, and ubiquitinated (Sieben et al., 2012; Al-Chalabi et al.,
AC C
2012; Gendron et al., 2013; Wang et al., 2014), and finally transported outside of the nucleus to the cytoplasm (Zhang et al., 2009; Barmada et al., 2010; Che et al., 2011; Sieben et al., 2012; Al-Chalabi et al., 2012; Gendron et al., 2013). Zhang et al. showed that accumulation of the 25-kDa fragment of TDP-43 led to the formation of toxic, insoluble, and ubiquitin- and phospho-positive cytoplasmic inclusions within cells (Zhang et al., 2009). In another study, Che et al. reported that abundance of the 35-kDa fragment of TDP-43 led to cellular redistribution, inclusion body formation, and altered RNA processing (Che et al., 2011). Therefore, short C-terminal fragments of TDP-43
ACCEPTED MANUSCRIPT
produce neuronal toxicity and cell death by forming ubiquitin-positive cytoplasmic inclusions within cells (Zhang et al., 2009; Barmada et al., 2010; Che et al., 2011; Sieben et al., 2012; Al-Chalabi et al., 2012; Gendron et al., 2013). Neuronal TDP-43 aggregation is accordingly observed in a number of neurological disorders including
PT
FTLD and ALS (Sieben et al., 2012, Al-Chalabi et al., 2012, Gendron et al., 2013). TDP-43 aggregation and nuclear depletion also occur in astrocytes, but TDP-43 toxicity
RI
is not well studied in this context. In the present study, levels of the 25- and 35-kDa
SC
fragments of TDP-43 were increased and full-length TDP-43 (43 kDa) was decreased after TBI in a manner associated with neuronal loss and motor impairment. Moreover,
NU
TDP-43 cleavage products were co- localized with GFAP in reactive astrocytes after TBI.
MA
These findings suggest that TBI may induce TDP-43 proteolysis in astrocytes as a process related to astrocyte activation and downstream functional consequences of TBI.
ED
The exact role of TDP-43 in astrocytes and its relationship with neuronal death is not fully understood. Astrocytes are the most abundant cell type in the brain and serve a
EP T
variety of supportive functions (Pekny et al., 2014). Astrogliosis or astrocyte reactivity refers to morphological and functional changes in astrocytes that occur in response to
AC C
insult or injury. Reactive astrocytes permit the isolation and sequestration of injured tissue, limiting the lesion size and its influence on surrounding tissues; however, if left unresolved, reactive gliosis can affect neuroplasticity and CNS regeneration. Indeed, neurons cannot survive in the brain without close interaction with astrocytes (Pekny et al., 2014). Excitotoxicity is another common mechanism of secondary brain injury after TBI. Neurons are highly vulnerable to excitotoxicity after TBI. Therefore, neuronal cell counts were significantly lower in the TBI group.
ACCEPTED MANUSCRIPT
Some studies have shown that astrocytes expressing mutations in TDP43 can induce motor neuron cell death (Tong et al., 2013; Rojas et al., 2014). In contrast, other studies have reported that TDP-43 proteinopathies do not produce astrocyte non-cell-autonomous neuronal death in cell culture (Haidet-Phillips et al., 2013; Serio et
PT
al., 2013). In the present study, local administration of a caspase-3 inhibitor reduced TDP-43 cleavage, prevented astrocyte activation, and increased neuronal survival. The
RI
observed neuron-sparing effect may have been related to the preservation of healthy
SC
astrocyte-neuron interactions after TBI.
TDP-43 has been previously implicated in TBI (Moisse et al., 2009; McKee et al.,
NU
2010; Johnson et al., 2011; Yang et al., 2014). Previous studies have suggested that
MA
TDP-43 proteinopathy associated with repetitive head trauma is similar to that found in FTLD; indeed, the increased expression of TDP-43 cleavage products, protein
ED
redistribution to the cytoplasm, and inclusion formation have been reported in TBI models (McKee et al., 2010, Yang et al., 2014). In contrast, a recent study found no
EP T
association between a history of TBI and the presence of abnormally phosphorylated TDP-43 inclusions (Johnson et al., 2011). The present study findings supplement the
AC C
gaps in these previous studies by defining a more clear relationship between TBI and TDP-43 proteolysis in astrocytes. Most TBI studies use saline injection as sham control groups. In those studies, they want to prove the effect of treatment. In our study, rats were randomly divided into 3 groups in our study: TBI with vehicle treatment (TBI), TBI with caspase-3 inhibitor treatment (TBI + CI), and sham group (sham). In our sham group, no procedure was performed. We focus on the TDP-43 proteolysis in astrocytes after TBI. Therefore, we used normal rats as sham group.
ACCEPTED MANUSCRIPT
Caspases 3 play an essential role during apoptotic cell death. Recent studies suggest that caspase-3 also functions as a regulatory molecule in neurogenesis and synaptic activity (Clark et al., 2000; D'Amelio et al., 2010). Therefore, active caspase 3 could be detect at sham group. After TBI, caspase-3 immunoreactivity represents active
PT
caspase-3 is increased and caspase 3 inhibitor treatment reduces active caspase-3 activity. This study demonstrates caspase-3 cleavage and bioactivity after TBI in rats.
RI
Local posttreatment with caspase 3 inhibitor reduces TDP-43 cleavage, caspase-3
SC
activity and neuron loss after injury.
Our study had several limitations. First, we did not use an in vitro model to
NU
confirm the influence of TDP-43 cleavage in astrocytes on astrocyte-neuron interactions.
MA
Thus, we cannot speculate as to the exact mechanism of neurotoxicity involved. Second, it would be useful to induce TBI in transgenic animals overexpressing wild-type human
ED
or mouse TDP-43. Future studies using this strategy can provide information about whether TDP-43 overexpression leads to astrocyte activation. Third, we did not
EP T
specifically inhibit TDP-43 cleavage in our study. Currently, there is no drug available that specifically targets TDP-43 cleavage. Future molecular research is necessary to
AC C
answer additional questions about the exact role of TDP-43 processing in neurological and neurodegenerative disease.
ACCEPTED MANUSCRIPT
5. Conclusions Our study demonstrates that short fragments of TDP-43 are generated in astrocytes local to the lesion site after TBI in rats. Moreover, our findings suggest that early management of TDP-43 proteolysis can effectively reduce astrogliosis and improve
PT
functional outcomes post-injury. TDP-43, its proteolysis, and its cleavage products in astrocytes may be an important therapeutic target for motor dysfunction after TBI.
RI
Further molecular research is necessary to identify a specific inhibitor of TDP-43
AC C
EP T
ED
MA
NU
SC
proteolysis and confirm its utility in TBI and other brain disorders.
ACCEPTED MANUSCRIPT
ACKNOWLEDGEMENTS This study was supported in part by grants from the Ministry of Science and Technology, Taiwan (103-2314-B-214 -007 -MY2) and the E-da hospital, Kaohsiung,
PT
Taiwan (EDAHP-101026, EDAHP-103012, EDPJ103075 and EDPJ104072).
SC
RI
DISCLOSURE
The authors report no conflict of interest concerning the materials or methods used
MA
NU
in this study or the findings specified in this paper.
CONTRIBUTORSHIP STATEMENT
ED
Chih-Yuan Huang, Yi-Che Lee, Ping-Chia Li, Po-Chou Liliang, and Hao-kuang
EP T
Wang designed research; Kang Lu, Kuo-Wei Wang, Li-Ching Chang, Li- Yen Shiu, Ming-Feng Chen, Yuan- Ting Sun, and Hao-kuang Wang analyzed data; H.-K. Wang
AC C
wrote the paper.
ACCEPTED MANUSCRIPT
References Al-Chalabi A, Jones A, Troakes C, King A, Al-Sarraj S, van den Berg LH, 2012. The genetics and neuropathology of amyotrophic lateral sclerosis. Acta Neuropathol. 124:339-52.
PT
Barmada SJ, Skibinski G, Korb E, Rao EJ, Wu JY, Finkbeiner S, 2010. Cytoplasmic mislocalization of TDP-43 is toxic to neurons and enhanced by a mutation
RI
associated with familial amyotrophic lateral sclerosis. J Neurosci. 30:639-49
traumatic brain injury. Neuron. 76:886-99.
SC
Blennow K, Hardy J, Zetterberg H, 2012. The neuropathology and neurobiology of
NU
Che MX, Jiang YJ, Xie YY, Jiang LL, Hu HY, 2011. Aggregation of the 35-kDa
MA
fragment of TDP-43 causes formation of cytoplasmic inclusions and alteration of RNA processing. FASEB J. 25:2344-53.
ED
Clark RS, Kochanek PM, Watkins SC, Chen M, Dixon CE, Seidberg NA, Melick J, Loeffert JE, Nathaniel PD, Jin KL, Graham SH, 2000. Caspase-3 mediated neuronal
EP T
death after traumatic brain injury in rats. J Neurochem. 74; 740–753. D'Amelio M, Cavallucci V, Cecconi F. 2010. Neuronal caspase-3 signaling: not only cell
AC C
death. Cell Death Differ, 17(7):1104-14. Fang YS, Tsai KJ, Chang YJ, Kao P, Woods R, Kuo PH, Wu CC, Liao JY, Chou SC, Lin V, Jin LW, Yuan HS, Cheng IH, Tu PH, Chen YR, 2014. Full- length TDP-43 forms toxic amyloid oligomers that are present in frontotemporal lobar dementia-TDP patients. Nat Commun. 5:4824. Feeney DM, Boyeson MG, Linn RT, Murray HM, and Dail WG, 1981. Responses to cortical injury: I. Methodology and local effects of contusions in the rat. Brain Res. 211: 67–77.
ACCEPTED MANUSCRIPT
Gendron TF, Rademakers R, Petrucelli L, 2013. TARDBP mutation analysis in TDP-43 proteinopathies and deciphering the toxicity of mutant TDP-43. J Alzheimers Dis. 33 Suppl 1:S35-45 Haidet-Phillips AM, Gross SK, Williams T, Tuteja A, Sherman A, Ko M, Jeong YH,
PT
Wong PC, Maragakis NJ, 2013. Altered astrocytic expression of TDP-43 does not influence motor neuron survival. Exp Neurol. Dec 250:250-9.
RI
Johnson VE, Stewart W, Trojanowski JQ, Smith DH, 2011. Acute and chronically
SC
increased immunoreactivity to phosphorylation- independent but not pathological TDP-43 after a single traumatic brain injury in humans. Acta Neuropathol.
NU
122:715-26.
MA
Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG, 2010. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal
ED
research. PLoS Biol. 8(6):e1000412.
Knoblach SM, Alroy DA, Nikolaeva M, Cernak I, Stoica BA, Faden AI, 2004. Caspase
EP T
inhibitor z-DEVD-fmk attenuates calpain and necrotic cell death in vitro and after traumatic brain injury. J Cereb Blood Flow Metab. 24: 1119–1132.
AC C
Liliang PC, Liang CL, Lu K, Wang KW, Weng HC, Hsieh CH, Tsai YD, Chen HJ, 2010. Relationship between injury severity and serum tau protein levels in traumatic brain injured rats. Resuscitation. 81:1205-8. Loane DJ, Pocivavsek A, Moussa CE, Thompson R, Matsuoka Y, Faden AI, Rebeck GW, and Burns MP, 2009. Amyloid precursor protein secretases as therapeutic targets for traumatic brain injury. Nat Med. 15: 377–379. Maas AI, Stocchetti N, Bullock R, 2008. Moderate and severe traumatic brain injury in adults. Lancet Neurol. 7:728-41.
ACCEPTED MANUSCRIPT
McKee AC, Gavett BE, Stern RA, Nowinski CJ, Cantu RC, Kowall NW, Perl DP, Hedley-Whyte ET, Price B, Sullivan C, Morin P, Lee HS, Kubilus CA, Daneshvar DH, Wulff M, Budson AE, 2010. TDP-43 proteinopathy and motor neuron disease in chronic traumatic encephalopathy. J Neuropathol Exp Neurol. 69:918-29.
PT
Moisse K, Mepham J, Volkening K, Welch I, Hill T, Strong MJ, 2009. Cytosolic TDP-43 expression following axotomy is associated with caspase 3 activation in NFL-/-
RI
mice: support for a role for TDP-43 in the physiological response to neuronal injury.
SC
Brain Res. 1296:176-86.
benefits. Physiol Rev. 94:1077-98.
NU
Pekny M, Pekna M, 2014. Astrocyte reactivity and reactive astrogliosis: costs and
MA
Pekny M, Wilhelmsson U, Pekna M, 2014. The dual role of astrocyte activation and reactive gliosis. Neurosci Lett. 565:30-8.
ED
Rojas F, Cortes N, Abarzua S, Dyrda A, van Zundert B, 2014. Astrocytes expressing mutant SOD1 and TDP43 trigger motoneuron death that is mediated via sodium
EP T
channels and nitroxidative stress. Front Cell Neurosci. 8:24. Serio A, Bilican B, Barmada SJ, Ando DM, Zhao C, Siller R, Burr K, Haghi G, Story D,
AC C
Nishimura AL, Carrasco MA, Phatnani HP, Shum C, Wilmut I, Maniatis T, Shaw CE, Finkbeiner S, Chandran S, 2013. Astrocyte pathology and the absence of non-cell autonomy in an induced pluripotent stem cell model of TDP-43 proteinopathy. Proc Natl Acad Sci U S A. 110:4697-702. Sieben A, Van Langenhove T, Engelborghs S, Martin JJ, Boon P, Cras P, De Deyn PP, Santens P, Van Broeckhoven C, Cruts M, 2012. The genetics and neuropathology of frontotemporal lobar degeneration. Acta Neuropathol. 124:353-72. Tong J, Huang C, Bi F, Wu Q, Huang B, Liu X, Li F, Zhou H, Xia XG, 2013. Expression
ACCEPTED MANUSCRIPT
of ALS-linked TDP-43 mutant in astrocytes causes non-cell-autonomous motor neuron death in rats. EMBO J. 32:1917-26. Wang IF, Guo BS, Liu YC, Wu CC, Yang CH, Tsai KJ, and Shen CK, 2012. Autophagy activators rescue and alleviate pathogenesis of a mouse model with proteinopathies
PT
of the TAR DNA-binding protein 43. Proc Natl Acad Sci U S A. 109: Wang HK, Lee YC, Huang CY, Liliang PC, Lu K, Chen HJ, Li YC, Tsai KJ, 2015.
RI
Traumatic brain injury causes frontotemporal dementia and TDP-43 proteolysis.
SC
Neuroscience. 300:94-103.
Yang Z, Lin F, Robertson CS, Wang KK, 2014. Dual vulnerability of TDP-43 to calpain
NU
and caspase-3 proteolysis after neurotoxic conditions and traumatic brain injury. J
MA
Cereb Blood Flow Metab. 34:1444-52.
Zhang YJ, Xu YF, Cook C, Gendron TF, Roettges P, Link CD, Lin WL, Tong J,
ED
Castanedes-Casey M, Ash P, Gass J, Rangachari V, Buratti E, Baralle F, Golde TE, Dickson DW, Petrucelli L, 2009. Aberrant cleavage of TDP-43 enhances
AC C
EP T
aggregation and cellular toxicity. Proc Natl Acad Sci U S A. 106:7607-12.
ACCEPTED MANUSCRIPT
Figure Legends Figure 1. (A) Flowchart showing the animal model of TBI with caspase-3 inhibitor treatment. Animals were divided into three groups: sham surgery with no treatment (sham, n = 5), TBI with vehicle (0.2% DMSO) treatment (TBI, n = 27), and TBI with
PT
caspase-3 inhibitor treatment (TBI + CI, n = 12).
RI
Figure 2. TBI induces TDP-43 proteolysis in astrocyte. (A) Western blot analysis of
SC
TDP-43 levels in extracts of the cortex obtained from sham, TBI Day 1, Day 3 and Day 7 rats. The amounts of the 35-kDa TDP-43 fragments were increased 3 days, and 7 days
NU
post-TBI, and the 25- kDa TDP-43 fragments were increased 7 days post-TBI. Full-
MA
length TDP-43 decreased at 1, 3, and 7 days post-TBI (n = 5 rats per group). Blotting patterns are shown in the upper panel, and the statistical analysis is shown in the bottom
ED
panel. Results are the mean ± SEM of three independent experiments. *P < 0.05; **P < 0.005; ***P < 0.001. (B) Representative photomicrographs showing TDP-43
EP T
immunoreactivity in brain sections from sham, and 7 days post- TBI rats. TDP-43 (green)/ Glial fibrillary acidic protein (GFAP; red)/ 4',6-diamidino-2-phenylindole
AC C
(DAPI; blue) triple labeling in the sham or contusion cortices. TDP-43 were co-localized with GFAP. (n = 5 rats per group). (C) Representative TDP-43 protein bands in the urea-soluble fraction of brain extracts. The arrow points to the unmodified form of TDP-43 on the gel (n = 7 rats per group).
Figure 3. TBI increases active form of caspase-3. (A) Western blot analysis of the level of the active form of caspase-3 in extracts of the cortex obtained from sham, TBI Day 1, Day 3, and D7 rats (n = 5 rats per group). *P < 0.05; **P < 0.005 (B) Representative
ACCEPTED MANUSCRIPT
photomicrographs showing TDP-43 immunoreactivity in brain sections from sham, and 7 days post- TBI rats. GFAP (red)/ Caspase-3 (green)/ 4',6-diamidino-2-phenylindole (DAPI; blue) triple labeling in the sham or contusion cortices. Caspase-3 were
PT
co-localized with GFAP. (n = 5 rats per group).
Figure 4. Caspase 3 inhibitor blocked cytoplasmic redistribution of TDP-43 in the
RI
astrocyte. (A) Representative protein bands of TDP-43, TDP-35, and TDP-25 in the
SC
lesion from rat brains treated with caspase-3 inhibitor or vehicle. C-terminal fragment of TDP-43 generated after TBI and was rescued by caspase-3 inhibitor treatment. (n = 5
NU
rats per group). *P < 0.05 (B) Western blot analysis of the level of the active form of
MA
caspase-3 in extracts of the cortex obtained from TBI with and without caspase-3 inhibitor (n = 5 rats per group). *P < 0.05 (C) TDP-43 (green)/ Glial fibrillary acidic
ED
protein (GFAP; red)/ 4',6-diamidino-2-phenylindole (DAPI; blue) triple labeling in the contusion cortices with caspase-3 inhibitor. The morphology of reactive astrocyte
EP T
become less hypertrophy.
AC C
Figure 5. TBI induce motor dysfunction. (A) The latencies of the rotarod test were significantly shorter in rats TBI rats. (B, C, D) Beam walking performance of sham, TBI Day 7, and TBI + CI Day 7 rats. The latencies, the total faults of the right hind legs, and the total faults of the left hind legs of the three groups are shown. *P < 0.05; **P < 0.005; ***P < 0.001.
Figure 6. Early treatment of caspase-3 inhibitor can improve neuron survival. (A) The dot plot from FACS-like analysis system-based neuronal counting in
ACCEPTED MANUSCRIPT
immunocytochemistry staining for NeuN and Dapi. (B) Quantification of survived
AC C
EP T
ED
MA
NU
SC
RI
PT
neuronal number by FACS-like analysis system (n = 5 rats per group). *P < 0.05.
PT
ACCEPTED MANUSCRIPT
AC C
EP T
ED
MA
NU
SC
RI
Fig. 1
EP T AC C
Fig. 2
ED
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC C
EP T
ED
Fig. 3
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC C
EP T
ED
Fig. 4
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC C
EP T
ED
MA
Fig. 5
PT
ACCEPTED MANUSCRIPT
AC C
EP T
ED
MA
NU
SC
RI
Fig. 6
MA
NU
SC
RI
PT
ACCEPTED MANUSCRIPT
AC C
EP T
ED
Graphical abstract
ACCEPTED MANUSCRIPT
HIGHLIGHTS
TBI increased TDP-43 proteolysis in astrocyte, and induced astrocyte hypertrophy.
TBI induced impaired behaviors that was associated with TDP-43 and its fragments
in astrocyte. Early management of TBI is effective in reducing TDP-43-associated impaired
AC C
EP T
ED
MA
NU
SC
RI
PT
behaviors