Serum and amygdala microRNA signatures of posttraumatic stress: Fear correlation and biomarker potential

June 7, 2017 | Autor: N. Balakathiresan | Categoria: Fear, Animals, Male, Amygdala, microRNAs, Rats, Biological markers, Psychiatric, Rats, Biological markers, Psychiatric
Share Embed


Descrição do Produto

Journal of Psychiatric Research 57 (2014) 65e73

Contents lists available at ScienceDirect

Journal of Psychiatric Research journal homepage: www.elsevier.com/locate/psychires

Serum and amygdala microRNA signatures of posttraumatic stress: Fear correlation and biomarker potential Nagaraja S. Balakathiresan a, 1, Raghavendar Chandran a, b, 1, Manish Bhomia a, Min Jia c, He Li c, Radha K. Maheshwari a, * a b c

Department of Pathology, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814, USA Biological Sciences Group, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA

a r t i c l e i n f o

a b s t r a c t

Article history: Received 11 December 2013 Received in revised form 21 May 2014 Accepted 29 May 2014

Exposure to acute traumatic stress can cause permanent changes in neurological circuitry and may lead to the development of an anxiety disorder known as posttraumatic stress disorder (PTSD). Current diagnosis of PTSD is based on clinical or behavioral symptom assessment, however, these are not definitive due to overlapping symptoms with other psychiatric disorders or mild traumatic brain injury (mTBI). No FDA approved diagnostic tests or biomarkers are currently available for diagnosis of PTSD. Recently, circulating miRNAs have emerged as novel biomarkers of many diseases. In this study, we have examined the altered expression of serum and amygdala miRNAs in an animal model of PTSD. Differentially expressed and statistically significant miRNAs in serum were validated for their presence in amygdala of corresponding animals. A panel of nine stress-responsive miRNAs viz., miR-142-5p, miR-19b, miR-1928, miR-223-3p, miR-322*, miR-324, miR-421-3p and miR-463* and miR-674* were identified, and may have potential as biomarker(s) for PTSD. Further validations by bioinformatics and system biology approaches indicate that five miRNAs such as miR-142-5p, miR-19b, miR-1928, miR-223 and miR-421-3p may play a potential role in the regulation of genes associated with delayed and exaggerated fear. To the best of our knowledge, this is the first report demonstrating the plausibility of using circulating miRNAs as biomarkers of PTSD. Published by Elsevier Ltd.

Keywords: Traumatic stress Biomarker microRNA Serum Amygdala PTSD

1. Introduction Psychological distress that may follow after witnessing a traumatic event usually subsides in most cases, however, for some individuals, this distress develops into a chronic disorder which is termed as posttraumatic stress disorder (PTSD) (Sones et al., 2011). According to recent studies, it is estimated that approximately 7.3% of the global civilian population suffer from PTSD (Malan-Muller et al., 2013). PTSD is also a major psychological health issue in armed forces veterans who were previously involved in combat activities. Study shows that 10e30% of US veterans suffer from PTSD or other stress related disorders (Zhang et al., 2011). The clinical symptoms of PTSD may include feeling of helplessness, hypervigilance, irritability, exaggerated fear response or trauma-

* Corresponding author. Tel.: þ1 (301) 295 3394; fax: þ1 (301) 295 1640. E-mail address: [email protected] (R.K. Maheshwari). 1 Authors contributed equally in this study. http://dx.doi.org/10.1016/j.jpsychires.2014.05.020 0022-3956/Published by Elsevier Ltd.

specific reenactment (Berna et al., 2012). Diagnosis of PTSD is currently based on symptoms determined from the patient's clinical history, examination of mental status, duration of symptoms and clinical symptom checklists or the patient self-report (Weathers et al., 2001). Several promising diagnosing methods and candidate biomarkers such as imaging (i.e., magnetic resonance imaging (MRI), functional MRI, single photon emission computed tomography etc.), psychological, endocrine and molecular (DNA/SNP, mRNA, protein) are currently in various phases of development (Schmidt et al., 2013). However, to date, none of these methods is in clinical use due to lack of reliability, specificity and cost efficacy (Schmidt et al., 2013). MiRNAs are small (~22 nucleotide), endogenous, evolutionarily conserved non-coding RNAs and are posttranscriptional gene regulators of diverse biological processes. MiRNAs in circulation are considered as good biomarkers because they are highly stable in serum due to their ability to withstand repeated freeze thaw, enzymatic degradation and extreme pH conditions (Scholer et al., 2010). Circulatory miRNAs have recently shown great promise as

66

N.S. Balakathiresan et al. / Journal of Psychiatric Research 57 (2014) 65e73

non-invasive and reliable diagnostic biomarkers for different diseases and disorders such as traumatic brain injury (TBI), bipolar disorder and schizophrenia (Lai et al., 2011; Rong et al., 2011; Balakathiresan et al., 2012; Redell et al., 2010). For instance, miRNAs are implicated as plasma biomarkers in clinical samples of mild cognitive impairment (MCI) which is usually associated with the early stages of many neurodegenerative disorders such as Alzheimer's disease, vascular and frontotemporal dementia, and Parkinson's disease. It was reported that miR-132 and miR-134 families could accurately diagnose MCI in majority of patients at asymptomatic phase (Sheinerman et al., 2012). In schizophrenia, miR-34a was shown to be an accurate serum biomarker for diagnosis (Lai et al., 2011). However, currently, there are no reports on the use of circulatory miRNAs as non-invasive biomarkers for the diagnosis of PTSD. In this study, we have used a rat model of learned helplessness stress to identify significantly modulated miRNAs in serum after traumatic stress. Our animal model mimics the pathophysiology similar to those found in PTSD subjects such as delayed and exaggerated startle response, enhanced plasma corticosterone (CORT) levels and retarded body weight gain several days after the cessation of stress (Jia et al., 2012). Among them, the delayed appearance of exaggerated acoustic startle response (ASR) corresponds to compressed time scale of the rat's life compared to a human such as 1e3 months delay in development of symptoms in PTSD patients (Jiang et al., 2011; Jia et al., 2012). The three major regions in limbic system such as amygdala, hippocampus and the pre-frontal cortex (PFC) are mainly altered in PTSD (Mahan and Ressler, 2012). Among them, hyperactive amygdala in PTSD plays a central role in fear dysregulation in response to trauma related cues (Schmidt et al., 2013). Mahan and Ressler (2012) suggested that many of the molecular tools that have been developed to study behavior in rodents could be applied to study the mechanism(s) of fear dysregulation. In this study, we have identified the significantly modulated miRNAs in serum after traumatic stress to develop a novel non-invasive PTSD diagnostic biomarker and validated their expression in amygdala since it is involved in regulating fear response under stress (Morey et al., 2012). Using in silico and system biology approaches, we have elucidated the regulatory role of modulated miRNAs in PTSD pathophysiology and in particular the fear response. 2. Methods and materials 2.1. Animals and stress protocol Twenty four male albino Sprague Dawley rats (Taconic Farms, Germantown, NY, USA) weighing 76e100 g and aged between 4 and 6 weeks old were used for this study. These animals were kept for acclimation for a week and then the rats were grouped into two groups of 12 animals each for stress and control. Young animals were used for this study to give sufficient time for simulating PTSD progression as seen in the battlefield scenario. Development of PTSD like symptoms may take at least two weeks after the cessation of stressors in the animal model and hormonal changes occur immediately after stress exposure as compared to the molecular level changes (Servatius et al. 1995). Hence, young animals were used to give sufficient time for studying the molecular level changes like protein or gene expression during PTSD development. Housing conditions, acclimation of rats and the stress protocol were followed as previously described (Jia et al., 2012). The stress protocol consisted of a 2 h per day session of immobilization along with tail shocks for three consecutive days. These animals were restrained and exposed to 40 electric shocks (2 mA, 3 s duration) at varying intervals of 150e210 s. Control groups were handled

similar to stress group such as acclimation and housing except for the stress protocol. The Institutional Animal Care and Use Committee of the USUHS approved all the experimental procedures. 2.2. Samples collection Animals from both the groups of control and post stress were sacrificed immediately (day 0; N ¼ 6 each) and day 14 (N ¼ 6 each) after the last stress exposure and the samples were collected between 1100 and 1200 h. Trunk blood was collected in 15 ml centrifuge tubes (VWR International, Radnor, PA, USA) and was left to clot at room temperature for 30 min for serum extraction. Blood was centrifuged (Allegra 6R centrifuge, Beckman Coulter) at 3500 rpm for 30 min at 4  C and supernatants were harvested in clean tubes. The supernatant was again centrifuged at 3500 rpm for 10 min at 4  C to pellet down remaining cellular fraction. The serum obtained was aliquoted into 1.5 ml microfuge tubes and stored at 80  C until further use. Brain dissection and subsequent collection of amygdala was carried out as described previously (Jia et al., 2012). Amygdala tissues were immediately submerged into RNAlater-RNA stabilization reagent (Qiagen, Valencia, USA) in microfuge tubes and then stored at 80  C until further use. 2.3. RNA isolation, quantity and quality check Total RNA including miRNA was isolated from the serum samples using the miRNeasy Serum/Plasma Kit (Qiagen, Valencia, USA) according to the manufacturer's protocol. QIAzol lysis reagent (1 ml) was added to the serum sample (200 mL) and vortexed. After incubating at room temperature for 5 min, 200 mL of chloroform was added and the samples were incubated at room temperature for 2e3 min and centrifuged for 15 min at 12,000g at 4  C. The aqueous phase obtained after centrifugation was mixed with 1.5 volume of 100% ethanol and loaded into an RNeasy MiniElute spin column in a 2 ml collection tube. The flow through after centrifugation was discarded and the column was washed with 700 mL of Buffer RWT, 500 mL of Buffer RPE, 500 mL of 80% ethanol and then finally eluted with 14 mL of RNAse-free water. Total RNA was isolated from the amygdala tissue by combining a protocol of TRIzol reagent (Ambion/Life Technologies, Carlsbad, CA, USA) and the mirVana miRNA isolation kit (Ambion/Life Technologies, Carlsbad, CA, USA) according to the manufacturer's protocol. Briefly, 2 volumes of TRIzol were added to the samples along with 1 volume of chloroform. After centrifugation, the aqueous layer was collected and mixed with 1.25 volume of absolute ethanol and passed through the RNAqueous micro kit cartridge and RNA eluted in TE buffer. Quality and quantity of small RNA for both serum and amygdala samples were analyzed using Agilent Small RNA kit (Agilent Technologies, Santa Clara, CA, USA) in Agilent 2100 Bioanalyzer. Bioanalyzer data indicated the presence of good quality miRNA in total serum RNA extractions. However, the miRNA quantity in serum was an average of 15 ng/ml (Fig. S1). This is expected since miRNAs are reported to be present in serum at low concentration and most of them are secreted out of the cells (Sayed et al., 2013). miRNA concentrations of 30 ng of serum and 5 ng of amygdala miRNAs were used for the PCR reactions. 2.4. Reverse transcription, pre-amplification and real-time quantitative PCR Reverse transcription (RT) was performed with TaqMan miRNA RT Kit (Life Technologies, Carlsbad, CA, USA) as described with slight modifications (Balakathiresan et al., 2012). miRNA quantity was measured from the total RNA of bioanalyzer data and used as template RNA (5 ng-brain miRNA; 30 ng-serum miRNA) for RT

N.S. Balakathiresan et al. / Journal of Psychiatric Research 57 (2014) 65e73

67

Table 1 Posttraumatic stress exposure altered miRNAs in serum. S#

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72

Serum Day 0

Serum Day 14

Detector

RQ_Stress-Control

P value_Stress-Control

Detector

mmu-miR-188-5p mmu-miR-511 hsa-miR-183* mmu-miR-224 mmu-miR-130a hsa-miR-27a* mmu-miR-328 mmu-miR-128a mmu-miR-148a mmu-miR-29c mmu-miR-28 mmu-miR-26b mmu-miR-31 mmu-miR-151-3p mmu-miR-223 hsa-miR-425 hsa-miR-26b* mmu-miR-652 mmu-miR-31* rno-miR-7* hsa-miR-223 mmu-miR-210 mmu-miR-339-5p mmu-miR-26a mmu-miR-191 mmu-miR-872* mmu-miR-200a mmu-miR-30d mmu-miR-138 mmu-miR-331-3p mmu-miR-191*

7.90 3.61 2.64 2.56 6.34 5.83 5.00 4.70 4.65 4.58 4.42 3.82 3.78 3.71 3.62 3.47 3.42 3.37 3.17 3.11 3.03 3.02 3.01 2.81 2.73 2.71 2.67 2.46 2.33 2.30 2.21

0.04 0.01 0.02 0.02 0.03 0.00 0.01 0.02 0.00 0.01 0.00 0.03 0.00 0.00 0.01 0.02 0.00 0.05 0.01 0.00 0.01 0.04 0.02 0.04 0.01 0.03 0.02 0.04 0.04 0.03 0.05

mmu-miR-673 mmu-miR-1928 mmu-miR-463* mmu-miR-326 rno-miR-148b-5p hsa-miR-106b* rno-miR-7a* hsa-miR-27a* mmu-miR-101b hsa-miR-26b* mmu-miR-17* mmu-miR-328 hsa-miR-338-5P mmu-miR-128a mmu-miR-24-2* mmu-miR-877* mmu-miR-16* mmu-miR-345 mmu-miR-18a* mmu-miR-652 mmu-miR-181c hsa-miR-425 hsa-miR-9* mmu-miR-301b mmu-miR-92a rno-miR-339-3p mmu-miR-301a mmu-miR-223 mmu-miR-494 mmu-miR-19a hsa-miR-421 mmu-miR-350 mmu-miR-130b mmu-miR-191* mmu-miR-342-3p mmu-miR-18a mmu-miR-20b mmu-miR-106a mmu-miR-872 hsa-miR-423-3P mmu-miR-339-5p rno-miR-17-3p mmu-miR-20a mmu-miR-191 mmu-miR-17 mmu-miR-324-5p mmu-miR-29a mmu-miR-19b hsa-miR-223 mmu-miR-24 mmu-miR-142-5p mmu-miR-138* mmu-miR-331-3p mmu-miR-872* mmu-miR-186 mmu-miR-27a hsa-miR-23a* mmu-miR-148a mmu-miR-340-3p mmu-miR-142-3p rno-miR-664 hsa-miR-22* mmu-let-7a* mmu-miR-744 mmu-miR-26a mmu-miR-210 mmu-miR-151-3p mmu-miR-320 mmu-miR-93 mmu-miR-674* mmu-miR-193b mmu-miR-322*

RQ_Stress-Control 12.60 11.23 9.97 9.94 8.38 7.36 6.74 6.35 6.12 6.05 5.59 5.41 5.36 5.36 5.11 4.94 4.84 4.80 4.73 4.66 4.62 4.55 4.53 4.43 4.35 4.34 4.27 4.25 4.20 4.16 3.96 3.90 3.90 3.88 3.86 3.84 3.82 3.55 3.51 3.51 3.42 3.37 3.31 3.27 3.27 3.26 3.17 3.13 3.11 3.04 2.95 2.92 2.90 2.88 2.80 2.74 2.71 2.71 2.70 2.70 2.69 2.68 2.67 2.60 2.53 2.51 2.49 2.37 2.37 2.30 2.26 2.25

P value_Stress-Control 0.00 0.00 0.01 0.00 0.00 0.00 0.00 0.00 0.00 0.00 0.01 0.00 0.00 0.00 0.00 0.00 0.01 0.00 0.00 0.00 0.00 0.01 0.04 0.02 0.00 0.00 0.01 0.00 0.02 0.00 0.00 0.01 0.01 0.00 0.00 0.01 0.01 0.01 0.04 0.00 0.00 0.03 0.01 0.00 0.01 0.01 0.01 0.02 0.02 0.00 0.03 0.01 0.00 0.01 0.03 0.03 0.04 0.02 0.05 0.05 0.00 0.02 0.04 0.00 0.04 0.02 0.01 0.00 0.02 0.04 0.01 0.05 (continued on next page)

68

N.S. Balakathiresan et al. / Journal of Psychiatric Research 57 (2014) 65e73

Table 1 (continued ) S#

Serum Day 0 Detector

Serum Day 14 RQ_Stress-Control

P value_Stress-Control

73 74 75 76 77 78 79 80 81 82

Detector

RQ_Stress-Control

P value_Stress-Control

hsa-miR-671-5p rno-miR-7* mmu-miR-484 hsa-miR-324-3p mmu-miR-324-3p mmu-miR-720-001629 rno-miR-190b mmu-let-7i rno-miR-224 mmu-miR-125b-5p

2.25 2.23 2.12 2.11 2.06 2.01 27.95 4.41 2.29 2.11

0.00 0.05 0.02 0.03 0.01 0.02 0.02 0.03 0.02 0.01

miRNAs marked in bold letters are common between day 0 and day 14.

reactions (Fig. S1). Briefly, RT reaction mixture contained 0.8 ml Megaplex RT primers Rodent Pool A/B (v3.0), 0.2 ml 100 mM dNTPs (with dTTP), 1.5 ml Multiscribe reverse transcriptase (50 U/ml), 0.8 ml 10 RT Buffer, 0.9 ml MgCl2 (25 mM), 0.1 ml RNAse inhibitor (20 U/ ml), RNA template and nuclease free water to a final volume of 7.5 ml. RT reaction was carried out on Veriti 96-Well Thermal Cycler (Life Technologies, Carlsbad, CA, USA) according to manufacturer's recommended thermal-cycling conditions. Pre-amplification of RT products, cycles and conditions was followed according to the manufacturer's protocol (Life Technologies, Carlsbad, CA, USA). The undiluted pre-amplification products were used for the miRNA profiling using TaqMan Low-Density Rodent microRNAs Array (TLDA) Set v3.0 (Applied Biosystems, Inc) containing 692 rodent miRNAs. The quantitative PCR (qPCR) reaction was carried out at default thermal-cycling conditions in ABI 7900HT Fast Real-Time PCR System (Applied Biosystems, Life Technologies, Foster City, CA).

2.5. TaqMan miRNA assay TaqMan miRNA assays (Applied Biosystems, Life Technologies, Foster City, CA) were carried out to validate the changes in the expression of selected miRNAs in serum and amygdala. RT was performed as per manufacturer's protocol using miRNA specific RT primers and mammalian U6 small nuclear RNA (U6 snRNA) was used as an endogenous control for the validation of all selected miRNAs. RT and RT-qPCR reactions were carried out as described previously from our laboratory by Balakathiresan et al. (2012). TaqMan miRNA assays were carried out in triplicate. For relative quantification, each miRNA was calibrated to the expression of U6 snRNA, which then gave a delta CT (DCt) value for each miRNA (miRNA Ct valueeU6 Ct value). The fold changes were calculated using the comparative Ct method (2DDCt).

2.6. miRNA expression analysis MiRNA expression profiles for Ct values were analyzed using real-time StatMiner® software (Integromics Inc) to identify significantly modulated stress-responsive miRNAs. For relative quantification of miRNAs between control and traumatic stress exposed animals, the following steps were performed in the StatMiner® software suite: quality control of biological replicates, selection of U6 snRNA as an optimal endogenous control, filtering of miRNAs expression having Ct values below 35 cycles and the detection of expression in all biological replicates of calibrator and target. For our study, we used Benjamini Hochberg False Discovery Rate (FDR) adjustment to create a DDCt value showing any up or downregulation along with unadjusted and adjusted P values. Statistically significant miRNAs were selected based on P values less than 0.2 for FDR and for the unadjusted P value lower than 0.05.

2.7. miRNA-target prediction and analysis Predicted targets of differentially expressed serum and amygdala miRNAs downloaded from miRWalk, a target prediction algorithm, were analyzed. MiRWalk is a combinatorial miRNA-target prediction tool and able to identify both predicted and validated targets (Dweep et al., 2011). Both functional and network analysis of altered miRNA and their gene targets associated with fear responses were performed using Ingenuity Pathway Analysis (IPA) program (Ingenuity Systems Inc, Redwood City, CA).

3. Results 3.1. Analysis of miRNA signatures in serum and correlation with amygdala miRNAs following exposure to traumatic stress The miRNA expression profiling identified 82 miRNAs, which were differentially expressed at day 14 after traumatic stress, whereas only 18 miRNAs were modulated in serum at day 0 after the cessation of stress (Table 1). Our primary objective of this study was to identify miRNA candidates in serum to diagnose PTSD, therefore, we also evaluated the miRNAs expression in amygdala due to its critical role in fear conditioning (Morey et al., 2012). A comparison of miRNAs expression profile in amygdala at day 0 and day 14 with serum miRNAs indicated a similar miRNA modulation pattern (Table 2). Fourteen miRNAs were modulated at day 0 whereas 60 miRNAs were modulated at day 14 after the cessation of stress. We also observed that most of the modulated miRNAs at day 0 were significantly downregulated in both serum (27 out of 31) and amygdala (8 out of 14). However, this trend of miRNA downregulation at day 0 was reversed at day 14 post stress where 78 out of 82 miRNAs were upregulated in serum and all 60 significantly modulated miRNAs were upregulated in amygdala. No common miRNAs were found between all the four groups. However, comparison of serum and amygdala profiles showed 9 common miRNAs at day 14. No similar miRNAs between serum and amygdala were observed at day 0. Comparison of miRNAs in serum samples at day 0 and 14 showed 18 common miRNAs whereas only 4 miRNAs were common in amygdala profiling data at day 0 and day 14 (Fig. 1). The symptoms and pathophysiology of PTSD in our model has been previously reported to develop at day 14 after stress exposure, which also correlates with the changes in the miRNA expression profile. Moreover, PTSD in humans has been shown to develop over a period of time after the traumatic stress (Jia et al., 2012). Therefore, we compared miRNA profiles of day 14 serum and amygdala to diagnose PTSD in our stress animal model and identified 9 upregulated miRNAs as common viz., miR-142-5p, miR-19b, miR-1928, miR-223-3p, miR-322*, miR-324, miR-421-3p and miR-463* and miR-674* (Table 3). However, this panel of miRNAs represents a small subset of miRNAs, and it is possible that

N.S. Balakathiresan et al. / Journal of Psychiatric Research 57 (2014) 65e73

69

Table 2 Posttraumatic stress exposure altered miRNAs in amygdala. S#

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Amygdala Day 0

Amygdala Day 14

Detector

RQ_Stress-Control

P value_Stress-Control

Detector

RQ_Stress-Control

P value_Stress-Control

mmu-miR-429 mmu-miR-29b mmu-miR-205 mmu-miR-130b* mmu-miR-690 mmu-miR-186 mmu-miR-449a mmu-miR-331-5p rno-miR-632 mmu-miR-342-3p mmu-miR-376a* mmu-miR-467b mmu-miR-16 hsa-miR-27b*

2.02 2.58 2.31 2.25 2.16 3.02 2.84 2.44 25.01 2.24 2.16 2.07 2.00 2.00

0.01 0.05 0.01 0.04 0.05 0.00 0.01 0.01 0.03 0.02 0.02 0.01 0.02 0.01

rno-miR-632 hsa-miR-190b mmu-miR-1928 hsa-miR-124* mmu-miR-141 mmu-miR-706 mmu-miR-291a-3p mmu-miR-1982.2 rno-miR-673 mmu-miR-1896 hsa-miR-653 mmu-miR-362-5p mmu-miR-463* rno-miR-547 rno-miR-219-1-3p mmu-miR-146b mmu-miR-204 mmu-miR-300* mmu-miR-1188 mmu-miR-433-5p mmu-miR-200c mmu-miR-487b rno-miR-345-3p mmu-miR-130b* mmu-miR-363 rno-miR-409-3P mmu-miR-10a mmu-miR-342-3p mmu-miR-199b mmu-miR-28* mmu-miR-19b hsa-miR-28-3p hsa-miR-136* mmu-miR-124 mmu-miR-125b* mmu-miR-217 hsa-miR-412 hsa-miR-875-5p mmu-miR-674* mmu-miR-103 mmu-miR-671-3p hsa-miR-30e-3p mmu-miR-134 mmu-miR-223 rno-miR-146B mmu-miR-467b hsa-miR-421 mmu-miR-142-5p hsa-miR-151-5P hsa-miR-455 mmu-miR-9 mmu-miR-216b mmu-miR-99a rno-miR-344-3p hsa-miR-340 mmu-miR-383 mmu-miR-140 mmu-miR-188-5p hsa-miR-189 mmu-miR-322*

742.43 14.49 7.85 4.93 4.47 3.73 3.67 3.49 3.43 3.32 3.29 3.28 3.16 3.05 3.01 2.93 2.85 2.84 2.83 2.80 2.79 2.77 2.59 2.58 2.51 2.49 2.45 2.42 2.41 2.38 2.37 2.36 2.35 2.35 2.32 2.25 2.23 2.23 2.22 2.21 2.19 2.18 2.17 2.16 2.14 2.12 2.10 2.10 2.09 2.07 2.06 2.05 2.05 2.04 2.04 2.03 2.02 2.01 2.00 2.00

0.01 0.00 0.00 0.00 0.00 0.00 0.00 0.00 0.01 0.00 0.03 0.01 0.01 0.01 0.02 0.00 0.03 0.00 0.01 0.00 0.00 0.00 0.00 0.00 0.00 0.00 0.00 0.01 0.00 0.01 0.00 0.02 0.05 0.00 0.03 0.02 0.01 0.01 0.02 0.00 0.00 0.00 0.02 0.03 0.01 0.00 0.01 0.00 0.02 0.00 0.00 0.00 0.03 0.00 0.00 0.01 0.00 0.02 0.02 0.01

the other serum miRNAs may also be good biomarker of the traumatic stress. Therefore, in this study, we have only focused on miRNAs which possibly have a source from amygdala and may be involved in exaggerated fear. 3.2. Validation of differential expression in TaqMan miRNA assay Global miRNA screening platforms can introduce bias in the miRNA profiling which can occur because of the reproducibility of

the platform used, pre-amplification step due to low serum concentration and stable endogenous controls. All these factors may contribute and lead to an identification of false positive (Balakathiresan et al., 2012). Therefore, we validated the miRNA profiling data obtained from low-density array platform by performing individual miRNA assay. We selected miR-223 for validation as it was expressed in both serum and amygdala. miRNA-223 was found to be upregulated by two fold both in TLDA and individual miRNA assay. MiR-223 is reported to be enriched in

70

N.S. Balakathiresan et al. / Journal of Psychiatric Research 57 (2014) 65e73

gene targets by IPA program suggested cell death and survival as one of the top most biofunctions in the molecular and cellular functional category (Fig. S2). In canonical pathways, glucocorticoid receptor signaling pathway was among the top five pathways which is regulated by miRNAs (Fig. S3). Molecular functional network was constructed using fear related genes and molecules suggest that miR-223, miR-1928 (miR-221) may have direct role in STMN1 regulation (Fig. 3). Taken together, these data suggest that the selected nine miRNAs may have a potential role in PTSD development as their modulation is observed in both serum and amygdala and makes them an ideal biomarker candidate(s). 4. Discussion

Fig. 1. Overlapping miRNAs data analysis for the significantly modulated miRNAs among the four traumatic stress groups was done using the online Venn diagram generation tool (Oliveros, 2007).

hippocampus, midbrain, and cortex (Harraz et al., 2012). MiR-223 is also implicated in studies related to brain injury and stroke. MiR223 was reported to be prevalent in the relatively large vessellike structures scattered throughout the brain after TBI (Redell et al., 2009). In stroke animal model, miR-223 overexpression in hippocampus showed the neuroprotective effect by regulating the expression of glutamate receptor subunits, GluR2 and NR2B (Harraz et al., 2012). In our validation assays, we used U6 snRNA as an endogenous normalization control. Since, our criteria for selection for miRNAs was based on p value 1.5, we randomly selected one additional miRNA, miR-128a to confirm the TLDA data. The singleplex PCR assay for miR-128a and miR-223 confirmed and validated its expression for the same set of animals from the multiplex platform strengthening the validity of the candidate identified from the multiplex platform (Fig. 2, Fig. S4).

3.3. Prediction of traumatic stress altered miRNA targets and their pathway analysis In an effort to understand the role of 9 miRNAs which are common to both serum and amygdala in PTSD pathophysiology, we performed bioinformatics analysis to identify their gene targets. Analysis in MiRWalk database showed 331 experimentally validated gene targets (Table S1). Among these genes, we found that genes involved in anxiety regulation or developments are among the targets of the modulated miRNAs. Two genes stathmin (STMN1) and aquaporin 4 (AQP4) were identified and the role of these two genes is well studied in anxiety disorder. Moreover, they were identified as direct target of miR-223. Pathway analysis of validated

In this study, we used an animal model of PTSD which involves a restraint and tail shock for three continuous days. We performed a global miRNA expression profiling at day 0 and day 14 of serum and amygdala samples after the traumatic stress exposure to identify potential biomarker candidates in serum as well as the involvement of modulated miRNAs in fear memory formation and consolidation in the amygdala. The miRNA expression at day 0 immediately after the cessation of stress showed that most of the miRNAs were found to be downregulated in amygdala and this may be due to the “de novo protein synthesis” that supports long-lasting functional and structural plasticity which is a molecular requirement for new memory formation. (Griggs et al., 2013). The downregulated miRNAs were also shown to regulate memory formation in amygdala by repressing actin-regulating proteins that are involved in plasticity and memory (Griggs et al., 2013). Furthermore, the global reduction of several miRNAs expression in rodents forebrain such as amygdala, hippocampus and cortex were shown to regulate learning and memory (Gao et al., 2010; Konopka et al., 2010; Lin et al., 2011; Griggs et al., 2013). Much evidence indicates that the newly formed fear memories are being consolidated into stable long-term memories in the amygdala which are believed to be the site of fear memory storage (Fanselow and LeDoux, 1999; Nader et al., 2000). In order to identify miRNAs that are involved in consolidation and long-term stability of fear memories, we performed miRNA profiling in amygdala at day 14 after the cessation of traumatic stress. Analysis of day 14 miRNAs in amygdala revealed a substantial alteration of the posttranscriptional machinery characterized by a global increase in miRNA expression. This change could indicate the development and ongoing pathophysiology of the PTSD, as each miRNA is able to regulate the expression of several target genes (Beveridge et al., 2010). For example, we observed two fold upregulation of miR124, which has been shown to directly target mineralocorticoid receptor (MR) which regulates CORT secretion (Mannironi et al., 2013). Interestingly, Jia et al. (2012) demonstrated the downregulation of MR in amygdala enhanced the secretion of CORT for

Table 3 Posttraumatic stress altered day 14 common miRNAs in serum and amygdala. S#

1 2 3 4 5 6 7 8 9

TLDA ID

mmu-miR-142-5p-002248 mmu-miR-19b-000396 mmu-miR-1928-121164_mat mmu-miR-223-002295 mmu-miR-322#-002506 mmu-miR-324-3p-002509 hsa-miR-421-002700 mmu-miR-463#-002582 mmu-miR-674#-001956

MicroRNA Symbol

rno-miR-142-5p rno-miR-19b-3p rno-miR-221-3p rno-miR-223-3p rno-miR-322-3p rno-miR-324-3p rno-miR-421-3p rno-miR-463-5p rno-miR-674-3p

MirBase ID

MIMAT0000847 MIMAT0000788 MIMAT0000890 MIMAT0000892 MIMAT0000547 MIMAT0000554 MIMAT0017175 MIMAT0017309 MIMAT0005330

Mature Sequence

cauaaaguagaaagcacuacu ugugcaaauccaugcaaaacuga agcuacauugucugcuggguuuc ugucaguuugucaaauacccc aaacaugaagcgcugcaaca ccacugccccaggugcugcugg aucaacagacauuaauuggg uaccuaauuuguuguccauca cacagcucccaucucagaacaa

Serum Day 14

Amygdala Day 14

Fold change

P value

Fold change

P value

2.95 3.13 11.23 4.25 2.25 2.06 3.96 9.97 2.30

0.029 0.018 0.001 0.001 0.048 0.015 0.001 0.006 0.037

2.10 2.37 7.85 2.16 2.00 2.42 2.10 3.16 2.22

0.001 0.000 0.000 0.033 0.013 0.007 0.009 0.010 0.016

N.S. Balakathiresan et al. / Journal of Psychiatric Research 57 (2014) 65e73

Fig. 2. Validation of miR-223 expression in amygdala and serum samples of day 14. The levels of miRNA were normalized by the level of U6 snRNA endogenous control, and all reactions were performed in triplicate.

several days and the development of anxiety. Due to the alteration of large number of miRNAs (60 miRNAs; >2 fold) in day 14 amygdala, we selected only those miRNAs which were common (9 miRNAs) between serum and amygdala of day 14 for further analysis such as correlation with fear related genes. Network analysis of these 9 miRNAs with their fear related gene targets that are available in IPA showed only 5 of them were correlated with fear related genes (Fig. 3). For instance, cAMP responsive element binding protein 1 (Creb1) was identified as a direct target of miR-142-3p. Creb1 was recently reported to be downregulated in rat brain exposed to repeated inescapable shock (Smalheiser et al., 2011). This suggests that miR-142-3p may regulate the expression of Creb1 and may play an important role in stress related response (Fig. 3). Further, miR-221 and miR-223 were also found to regulate the expression of stathmin1 (STMN1), an important amygdala molecule involved in fear conditioning (Shumyatsky et al., 2005).

71

IPA analysis suggested an involvement of five miRNAs viz., miR142-5p, miR-19b, miR-1928, miR-223 and miR-421-3p in the regulation of genes associated with delayed and exaggerated fear. We further explored these five miRNAs for their brain specificity and/or their functions related to any neurological conditions. MiR142-5p was found to be enriched in microglia and has been shown to be upregulated after brain injury (Lei et al., 2009; Wu et al., 2011; Lau et al., 2013). Further, auditory fear training in Rattus norvegicus, downregulated the expression of miR-142-5p in lateral amygdala of naïve animals suggesting its involvement in memory formation dysfunction (Griggs et al., 2013). MiR-19b-3p that copurify with polyribosomes in mammalian neurons showed significantly higher expression in 6-hydroxydopamine-injured MN9D cells, indicating its role in neurodegenerative diseases by contributing to dopaminergic neuronal apoptosis (Li et al., 2013). MiR-221-3p expression was also upregulated in distal axons of superior cervical ganglia (SCG) after spinal cord injury (Liu et al., 2009, Wu et al., 2011). MiR223 and miR-19 are also enriched in glial cells and were shown to inhibit aberrant glial expression of neuronal proteins and phenotypes (Jovi ci c et al., 2013). The miR-421 was first identified in neocortex and hippocampus from developing rat brain and predicted to play a role in neurodegenerative disorders (Miska et al., 2004; Taguchi 2013). Recent studies also suggest participation of miR-421 in the regulation of plasminogen activator Inhibitor-1 (PAI-1) which is known to induce neuronal apoptosis, disrupt the blood brain barrier (BBB) and contribute to neurotoxicity in ischemic brain damage after stroke (Abu Fanne et al., 2010; Marchand et al., 2012). For biomarker identification, we selected only day 14 serum miRNA profiles for the analysis since the day 14 animals showed delayed and exaggerated startle response, enhanced plasma CORT and retarded body weight gain after several days (10e21 days) of posttraumatic stress in rats (Jia et al., 2012). Modulation of miRNAs in serum can occur either because of the change in the miRNAs expression in the regions of the brain which controls the stress

Fig. 3. Network analysis of posttraumatic stress altered day 14 serum and amygdala common miRNAs and their fear related gene targets based on published literatures and available in Ingenuity Pathway Analysis (IPA) software (Ingenuity Systems, www.ingenuity.com). The network correlation between miRNAs and their targets relevant to fear response were custom-built using “my pathway” tool in IPA. Molecular functional network suggests that miR-223, miR-1928 (miR-221) may have direct role in STMN1 regulation. Red color indicates the upregulated expression of miRNAs.

72

N.S. Balakathiresan et al. / Journal of Psychiatric Research 57 (2014) 65e73

response. These miRNAs can leach out in the serum by different ways as previously described (Andrews and Neises 2012). However, there is a possibility that serum miRNA modulation may occur due to a bystander effect of the stress on other organs which can potentially alter the serum miRNA expression profile. Such miRNAs can be a marker for organ stress but cannot be used as marker for psychological stress. To identify the true candidates biomarkers, we performed miRNA profiling of amygdala which is believed to play a critical role in regulation of fear conditioning in this animal model (Andero et al., 2013). Therefore, 9 miRNAs which are found to be upregulated in both amygdala and in serum were selected and analyzed for their correlation with PTSD pathophysiology by computational analysis to validate their potential as diagnostic biomarkers of PTSD. Since miRNA regulates the cell physiology by targeting the mRNA and altering the corresponding protein expression, the validated gene targets of 9 candidate miRNAs were identified using miRWalk program. These gene targets were used to identify the pathways involved using IPA. Interestingly, stress related glucocorticoid receptor signaling pathway appeared as one of the major canonical pathway which was predicted to be regulated by the 9 miRNAs. These computational analysis suggest that the candidate biomarkers of PTSD may have an important role in stress response and hence are good candidates for further biomarker validation studies. In conclusion, our data suggest that traumatic stress associated with a global decrease in day 0 and global increase in day 14 in miRNA expression in amygdala could have profound psychopathological implications in the context of PTSD development by influencing genes involved in fear memory formation and consolidation. A panel of dysregulated miRNAs present in both serum and amygdala after exposure to traumatic stress and their correlation with PTSD pathophysiology suggested them as promising candidates for biomarkers. To the best of our knowledge, this is the first study, where we have identified the serum miRNAs which may have potential for the diagnosis of PTSD. Further studies with human clinical samples will be required to identify the best biomarker candidate for their use as both diagnostic biomarkers and therapeutic targets for PTSD. Author Disclosure Statement The opinions expressed here are those of the authors and should not be construed as official or reflecting the views of the Uniformed Services University of Health Sciences, Bethesda, Maryland, Department of Defense, USA and the Birla Institute of Technology and Science, Pilani, Rajasthan, India. No competing financial interests exist. Contributors Nagaraja Balakathiresan contributed to the study design, experimental work, data analysis and preparation of the manuscript. Raghavendar Chandran contributed to the experimental works and literary search. Manish Bhomia contributed to the experimental work and the preparation of the manuscript. Min Jia conducted the animal experiment and contributed to the collection of blood and brain samples. He Li contributed to the animal experiment study design and effective suggestion for this study. Radha K Maheshwari contributed to the overall study design, manuscript preparation and editing and obtained the funding for the work carried out in this paper.

Role of the Funding Source This work was financially supported by a grant (# D10_I_AR_J6_855) from the Defense Medical Research and Development Program to Dr. Radha K Maheshwari. Conflicts of Interest The authors declare no conflict of interest. Acknowledgments The authors are grateful to Dr. Anuj Sharma and Mr. Kyle Thumar for the constructive suggestions and review of the manuscript. Appendix A. Supplementary data Supplementary data related to this article can be found at http:// dx.doi.org/10.1016/j.jpsychires.2014.05.020. References Abu Fanne R, Nassar T, Yarovoi S, Rayan A, Lamensdorf I, Karakoveski M, et al. Bloodbrain barrier permeability and tPA-mediated neurotoxicity. Neuropharmacology 2010;58(7):972e80. Andero R, Brothers SP, Jovanovic T, Chen YT, Salah-Uddin H, Cameron M, et al. Amygdala-dependent fear is regulated by Oprl1 in mice and humans with PTSD. Science Translation Medicine 2013;5(188):188ra73. Andrews JA, Neises KD. Cells, biomarkers, and post-traumatic stress disorder: evidence for peripheral involvement in a central disease. Journal of Neurochemistry 2012;120(1):26e36. Balakathiresan N, Bhomia M, Chandran R, Chavko M, McCarron RM, Maheshwari RK. MicroRNA let-7i is a promising serum biomarker for blastinduced traumatic brain injury. Journal of Neurotrauma 2012;29(7):1379e87. Berna G, Vaiva G, Ducrocq F, Duhem S, Nandrino JL. Categorical and dimensional study of the predictive factors of the development of a psychotrauma in victims of car accidents. Journal of Anxiety Disorders 2012;26(1):239e45. Beveridge NJ, Gardiner E, Carroll AP, Tooney PA, Cairns MJ. Schizophrenia is associated with an increase in cortical microRNA biogenesis. Molecular Psychiatry 2010;15(12):1176e89. Dweep H, Sticht C, Pandey P, Gretz N. miRWalkedatabase: prediction of possible miRNA binding sites by “walking” the genes of three genomes. Journal of Biomedical Informatics 2011;44(5):839e47. Fanselow MS, LeDoux JE. Why we think plasticity underlying Pavlovian fear conditioning occurs in the basolateral amygdala. Neuron 1999;23(2):229e32. Gao J, Wang WY, Mao YW, Graff J, Guan JS, Pan L, et al. A novel pathway regulates memory and plasticity via SIRT1 and miR-134. Nature 2010;466(7310):1105e9. Griggs EM, Young EJ, Rumbaugh G, Miller CA. MicroRNA-182 regulates amygdaladependent memory formation. Journal of Neuroscience 2013;33(4):1734e40. Harraz MM, Eacker SM, Wang X, Dawson TM, Dawson VL. MicroRNA-223 is neuroprotective by targeting glutamate receptors. Proceedings of the National Academy of Sciences of the United States of America 2012;109(46):18962e7. Jia M, Meng F, Smerin SE, Xing G, Zhang L, Su DM, et al. Biomarkers in an animal model for revealing neural, hematologic, and behavioral correlates of PTSD. Journal of Visualized Experiments 2012;68. Jiang X, Zhang ZJ, Zhang S, Gamble EH, Jia M, Ursano RJ, et al. 5-HT2A receptor antagonism by MDL 11,939 during inescapable stress prevents subsequent exaggeration of acoustic startle response and reduced body weight in rats. Journal of Psychopharmacology 2011;25(2):289e97. Jovi ci c A, Roshan R, Moisoi N, Pradervand S, Moser R, Pillai B, et al. Comprehensive expression analyses of neural cell-type-specific miRNAs identify new determinants of the specification and maintenance of neuronal phenotypes. Journal of Neuroscience 2013;33(12):5127e37. Konopka W, Kiryk A, Novak M, Herwerth M, Parkitna JR, Wawrzyniak M, et al. MicroRNA loss enhances learning and memory in mice. Journal of Neuroscience 2010;30(44):14835e42. Lai CY, Yu SL, Hsieh MH, Chen CH, Chen HY, Wen CC, et al. MicroRNA expression aberration as potential peripheral blood biomarkers for schizophrenia. PLoS One 2011;6(6):e21635. Lau P, Bossers K, Janky R, Salta E, Frigerio CS, Barbash S, et al. Alteration of the microRNA network during the progression of Alzheimer's disease. EMBO Molecular Medicine 2013;5(10):1613e34. Lei P, Li Y, Chen X, Yang S, Zhang J. Microarray based analysis of microRNA expression in rat cerebral cortex after traumatic brain injury. Brain Research 2009;1284:191e201. Li L, Chen HZ, Chen FF, Li F, Wang M, Wang L, et al. Global microRNA expression profiling reveals differential expression of target genes in 6-hydroxydopamineinjured MN9D cells. NeuroMolecular Medicine 2013;15:593e604.

N.S. Balakathiresan et al. / Journal of Psychiatric Research 57 (2014) 65e73 Lin Q, Wei W, Coelho CM, Li X, Baker-Andresen D, Dudley K, et al. The brain-specific microRNA miR-128b regulates the formation of fear-extinction memory. Nature Neuroscience 2011;14(9):1115e7. Liu NK, Wang X-F, Lu QB, Xu X-M. Altered microRNA expression following traumatic spinal cord injury. Experimental Neurology 2009;219:424e9. Mahan AL, Ressler KJ. Fear conditioning, synaptic plasticity and the amygdala: implications for posttraumatic stress disorder. Trends in Neurosciences 2012;35(1):24e35. Malan-Muller S, Hemmings SM, Seedat S. Big effects of small RNAs: a review of microRNAs in anxiety. Molecular Neurobiology 2013;47(2):726e39. Mannironi C, Camon J, De Vito F, Biundo A, De Stefano ME, Persiconi I, et al. Acute stress alters amygdala microRNA miR-135a and miR-124 expression: inferences for corticosteroid dependent stress response. PLoS One 2013;8(9):e73385.  AM, Tre goue €t DA. miR-421 and miR-30c Marchand A, Proust C, Morange PE, Lompre inhibit SERPINE 1 gene expression in human endothelial cells. PLoS One 2012;7(8):e44532. Miska EA, Alvarez-Saavedra E, Townsend M, Yoshii A, Sestan N, Rakic P, et al. Microarray analysis of microRNA expression in the developing mammalian brain. Genome Biology 2004;5(9):R68. Morey RA, Gold AL, LaBar KS, Beall SK, Brown VM, Haswell CC, et al. Amygdala volume changes in posttraumatic stress disorder in a large case-controlled veterans group. Archives of General Psychiatry 2012;69(11):1169e78. Nader K, Schafe GE, Le Doux JE. Fear memories require protein synthesis in the amygdala for reconsolidation after retrieval. Nature 2000;406(6797):722e6. Oliveros JC. VENNY. An interactive tool for comparing lists with Venn Diagrams. http://bioinfogp.cnb.csic.es/tools/venny/index.html; 2007. Redell JB, Liu Y, Dash PK. Traumatic brain injury alters expression of hippocampal microRNAs: potential regulators of multiple pathophysiological processes. Journal of Neuroscience Research 2009;87(6):1435e48. Redell JB, Moore AN, Ward 3rd NH, Hergenroeder GW, Dash PK. Human traumatic brain injury alters plasma microRNA levels. Journal of Neurotrauma 2010;27(12):2147e56. Rong H, Liu TB, Yang KJ, Yang HC, Wu DH, Liao CP, et al. MicroRNA-134 plasma levels before and after treatment for bipolar mania. Journal of Psychiatric Research 2011;45(1):92e5.

73

Servatius RJ, Ottenweller JE, Natelson BH. Delayed startle sensitization distinguishes rats exposed to one or three stress sessions: further evidence toward an animal model of PTSD. Biological Psychiatry 1995;38:539e46. Sayed AS, Xia K, Yang TL, Peng J. Circulating microRNAs: a potential role in diagnosis and prognosis of acute myocardial infarction. Disease Markers 2013;35(5):561e6. Schmidt U, Kaltwasser SF, Wotjak CT. Biomarkers in posttraumatic stress disorder: overview and implications for future research. Disease Markers 2013;35(1):43e54. Scholer N, Langer C, Dohner H, Buske C, Kuchenbauer F. Serum microRNAs as a novel class of biomarkers: a comprehensive review of the literature. Experimental Hematology 2010;38(12):1126e30. Sheinerman KS, Tsivinsky VG, Crawford F, Mullan MJ, Abdullah L, Umansky SR. Plasma microRNA biomarkers for detection of mild cognitive impairment. Aging (Albany NY) 2012;4(9):590e605. Shumyatsky GP, Malleret G, Shin RM, Takizawa S, Tully K, Tsvetkov E, et al. stathmin, a gene enriched in the amygdala, controls both learned and innate fear. Cell 2005;123(4):697e709. Smalheiser NR, Lugli G, Rizavi HS, Zhang H, Torvik VI, Pandey GN, et al. MicroRNA expression in rat brain exposed to repeated inescapable shock: differential alterations in learned helplessness vs. non-learned helplessness. International Journal of Neuropsychopharmacology 2011;14(10):1315e25. Sones HM, Thorp SR, Raskind M. Prevention of posttraumatic stress disorder. Psychiatric Clinics of North America 2011;34(1):79e94. Taguchi YH. MicroRNA-mediated regulation of target genes in several brain regions is correlated to both microRNA-targeting-specific promoter methylation and differential microRNA expression. BioData Mining 2013;6(1):11. Weathers FW, Keane TM, Davidson JR. Clinician-administered PTSD scale: a review of the first ten years of research. Depression and Anxiety 2001;13(3):132e56. Wu D, Raafat M, Pak E, Hammond S, Murashov AK. MicroRNA machinery responds to peripheral nerve lesion in an injury-regulated pattern. Neuroscience 2011;190:386e97. Zhang L, Hu X, Li X, Li H, Ursano R. PTSD and current translational research. In: Kalinin V, editor. Anxiety disorders. InTech; 2011. pp. 69e96. Available from: http://www.intechopen.com/books/anxiety-disorders/ptsd-and-currenttranslational-research.

Lihat lebih banyak...

Comentários

Copyright © 2017 DADOSPDF Inc.