Transcriptome Alterations in Posttraumatic Stress Disorder

Transcriptome Alterations in Posttraumatic Stress Disorder

Accepted Manuscript Transcriptome Alterations in Post-traumatic Stress Disorder Matthew J. Girgenti, Ronald S. Duman PII: S0006-3223(17)32045-0 DOI:...

1MB Sizes 0 Downloads 126 Views

Accepted Manuscript Transcriptome Alterations in Post-traumatic Stress Disorder Matthew J. Girgenti, Ronald S. Duman PII:

S0006-3223(17)32045-0

DOI:

10.1016/j.biopsych.2017.09.023

Reference:

BPS 13336

To appear in:

Biological Psychiatry

Received Date: 21 April 2017 Revised Date:

5 September 2017

Accepted Date: 17 September 2017

Please cite this article as: Girgenti M.J. & Duman R.S., Transcriptome Alterations in Post-traumatic Stress Disorder, Biological Psychiatry (2017), doi: 10.1016/j.biopsych.2017.09.023. 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

Transcriptome Alterations in Post-traumatic Stress Disorder Matthew J. Girgenti and Ronald S. DumanƢ

RI PT

Laboratory of Molecular Psychiatry, Center for Genes and Behavior, Department of Psychiatry, Yale University School of Medicine, New Haven, CT., USA, 06508

SC

Words in Abstract: 140 Words in Text: 4000 Number of Tables: 2 Number of Figures: 3 Number of Supplemental Figures: zero

Short Title: Transcriptomic Approaches to Characterize PTSD

M AN U

Key Words: PTSD; stress; transcriptomics; RNA-sequencing; genomics; prefrontal cortex

EP

TE D

Ƣ Address all correspondences to: Dr. Ronald S. Duman Elizabeth Mears and House Jameson Professor of Psychiatry and Pharmacology Director, Abraham Ribicoff Research Facilities Yale University School of Medicine Phone: (203) 974-7726 E-mail: [email protected]

AC C

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

ABSTRACT

1

ACCEPTED MANUSCRIPT

Post-traumatic stress disorder (PTSD) is a debilitating psychiatric disorder with a life-time prevalence of nearly 8% in the general population. While the underlying molecular and cellular mechanisms of PTSD remain unknown, recent studies indicate that PTSD is associated with aberrant gene expression in brain as well as peripheral blood cells. The advent of next generation sequencing technologies will allow us to elucidate the gene expression changes occurring in both brain and blood of patients with PTSD. RNA sequencing allows for analysis of the amount of transcript being made as well as alternative splicing, novel transcript identification, micro-RNA, and noncoding RNA quantification. Here we provide an overview of the different types of transcriptomic technologies, the gene expression studies performed in human peripheral blood and animal models of PTSD, and review the human PTSD post-mortem brain gene profiling studies performed to date.

91

development remain largely unknown (1). The ability to integrate genetic

AC C

EP

TE D

M AN U

SC

RI PT

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 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90

INTRODUCTION The pathophysiology of PTSD and the molecular changes underlying it’s

2

ACCEPTED MANUSCRIPT

information with disease states has been at the forefront of biomedical science

93

research with the hope of identifying gene mutations that underlie psychiatric, as

94

well as other types of illnesses. However, the possibility that a single mutation, or

95

even a small number of mutations are responsible for a complex illness like

96

schizophrenia (2), depression (3) or PTSD (4) has largely been disproved.

RI PT

92

An assortment of molecular tools has been developed in the intervening

98

years to analyze these genetic changes. Perhaps most notable is the rise of

99

technologies to analyze the output of the genetic code: the transcriptional

100

profiles. The advent of large-scale massively parallel sequencing technologies

101

(deep sequencing) has revolutionized our understanding of the molecular

102

underpinnings of disease states. The ability to interrogate a complete read-out of

103

the genomic, transcriptomic, and epigenomic landscape of a tissue or even

104

individual cells of a particular tissue will give unparalleled insight into the

105

molecular causes of many diseases. In addition, these studies hold the promise

106

of identifying core biological features and signaling pathways that could be

107

influenced by multiple gene mutations that converge to contribute to the neuronal

108

abnormalities that underlie PTSD.

TE D

M AN U

SC

97

PTSD generally develops from single or repeated exposure to traumatic

110

events, and is characterized by intrusive memories, avoidance of stimuli that are

111

reminders of the event, recurrent nightmares, and persistent hyperarousal (5).

112

Interestingly, the largest GWAS of PTSD individuals revealed significantly higher

113

genetic risk overlap with schizophrenia than with MDD or bipolar depression (6).

114

Clinical and preclinical studies have demonstrated three principle brain regions

115

involved in fear learning and modulation. The amygdala, a brain region involved

116

in fear memory (Figure 1) (7-11) is hyperactive in patients with PTSD (12). In

117

addition, the hippocampus (13; 14) and medial prefrontal cortex (mPFC) (15-19)

118

have been shown to directly modulate fear responsiveness of the amygdala in

119

humans as well as in preclinical models. Imaging studies have shown

120

dysregulation of both the hippocampus and mPFC in patients with PTSD(20).

121

Studies of alterations of gene expression in these regions and the epigenetic

122

mechanisms underlying these changes will help to elucidate fundamental

AC C

EP

109

3

ACCEPTED MANUSCRIPT

neurobiological processes of PTSD. It is possible that even though multiple gene

124

mutations may contribute to PTSD in different individuals, there may be

125

convergent underlying neurobiological gene expression changes that contribute

126

to PTSD symptoms. In addition, because of the inherent difficulty of examining

127

human brain tissue in patients, studies of gene expression in peripheral blood are

128

a necessary proxy, though there are obvious limitations and additional studies of

129

post-mortem brain are needed.

RI PT

123

Here we will focus on the approaches for investigating disease-related

131

changes in gene expression at the messenger RNA (mRNA) level. We will begin

132

by discussing the current technologies available for measuring alterations of

133

gene expression. We then summarize the recent publications describing gene

134

expression changes in the periphery of patients with PTSD and how they may

135

impact our understanding of the disorder.

136

differential gene expression studies of preclinical rodent models of PTSD and

137

how these results inform and correlate with transcript changes observed in blood.

138

Examination of these topics will set the stage for a discussion of studies done in

139

post-mortem brains of PTSD subjects and development of a large PTSD brain

140

bank to further pursue this work.

142

M AN U

We will then examine current

TE D

141

SC

130

Gene Expression Profiling of Neuropsychiatric Disorders The development and maturation of neurons and their connections and

144

optimization of circuit formation is critical for fine-tuning all aspects of brain

145

function, including cognition, information processing, and complex behaviors.

146

One of the most important processes in the development and maintenance of

147

these circuits is proper control of gene expression. The significance and impact

148

of gene expression on all aspects of brain function has driven the need for

149

technical and conceptual breakthroughs that allow for characterization of the

150

specific complement of mRNA transcripts.

AC C

EP

143

151

Recent advances in our knowledge of the genomic structure of humans

152

and other mammals have directed the emergence of sequencing approaches to

153

characterize and quantify gene expression products. Next generation sequencing

4

ACCEPTED MANUSCRIPT

(NGS) provides three to four magnitudes more sequencing information than

155

previous systems. The key feature of these systems is combining traditional

156

DNA sequencing technologies with microfluidics, to allow for optimal spatial

157

arrangement of each transcript and efficient scanning of individual base pairs on

158

a small flow cell.

RI PT

154

159

NGS has been applied to many different types of DNA and RNA sequence

160

analysis: genome sequencing (DNA-seq)(21), mRNA profiling (RNA-seq) (22;

161

23),

162

characterizations (26). RNA-seq uses the latest NGS technology to sequence the

163

transcriptome and generate gene expression profiles from many types of

164

samples. RNA is extracted from the tissue of interest and cDNA libraries are

165

generated. Adapter sequences are added to the 3’ and 5’ ends. These linker

166

adapters allow the cDNA to attach to the flow cell where sequencing occurs.

167

Each cDNA fragment (75-100 bp) is sequenced.

168

“reads” are aligned to a known reference genome or can be assembled without

169

reference (Figure 1B). This allows for unprecedented analysis of not only what

170

is expressed, but how it is expressed (i.e. alternative splicing, exonic SNP

171

identification, and novel transcript prediction). RNA-seq has been used to identify

172

the transcript changes in schizophrenia (27), major depressive disorder (28; 29),

173

bipolar (29), and drug dependence (30). There are currently few studies using

174

RNA-seq on human PTSD samples (31; 32). This is due in large part to the lack

175

of an organized repository for post-mortem PTSD brain tissue, a limitation

176

currently being addressed with the development of a new VA repository

177

dedicated to the collection of high quality tissue from PTSD subjects (33).

(ChIP-seq)

(24;

25)

and

epigenomic

M AN U

SC

interactions

EP

TE D

These resulting sequence

AC C

178

DNA-protein

179

Transcript changes in blood of PTSD patients: dysfunction of immune

180

response and glucocorticoid signaling

181

Studies to identify biomarkers of PTSD have focused largely on changes

182

in gene expression in blood of patients with PTSD (Table 1).

183

commonly used cell source for examining gene expression in humans because

184

of its easy availability. Although there are obvious limitations for brain disorders,

5

Blood is a

ACCEPTED MANUSCRIPT

185

peripheral blood likely reflects some changes occurring in the rest of body and

186

can act as proxy for detection of relevant gene transcript alterations and/or may

187

reveal biomarkers to aid in diagnosis and treatment response.

188

peripheral blood contains cells of the immune system, such as lymphocytes that

189

could directly impact brain function (34). Alterations of the hypothalamic-pituitary-

190

adrenal (HPA) axis and glucocorticoid function, in particular, have been shown to

191

be important in the onset of PTSD. Therefore, it is possible that changes in the

192

blood transcriptome may reflect some changes in the brain and offer insight into

193

the underlying pathophysiology of PTSD.

SC

RI PT

In addition,

A number of studies have examined blood gene expression and

195

glucocorticoid activity to identify the molecular underpinnings of PTSD (Table 1

196

and Figure 2).

197

inflammatory control plays a role in PTSD(35; 36). Several of these studies

198

identified down regulation of interluekins-16 and -18 (IL-16 and !L-18) and

199

colony-stimulating factor expression (37; 38); three monocyte

200

(39), and down regulation of the major histocompatabiity complex II (40). These

201

same studies have also identified Fkbp5 and Stat5b gene expression as being

202

down regulated in PTSD patients(38; 40; 41).

203

known roles in regulating inflammatory and glucocorticoid responses (42; 43).

204

Fkbp5 protein in particular is a chaperone of the glucocorticoid receptor (GR)

205

(42) that prevents translocation of GR to the nucleus where it regulates many

206

genes that contain GR promoter elements(44; 45). Gene association studies

207

have identified four SNPs (rs9470080, rs360780, rs3800373, and rs9296158) in

208

the Fkbp5 gene that are predictors of adult PTSD onset(38; 46). The significance

209

of these studies on blood Fkbp5 gene expression are highlighted by a preliminary

210

transcriptomic study of post-mortem brain tissue of PTSD subjects, which also

211

demonstrates significantly decreased expression of Fkbp5 expression in the

212

subgenual PFC(47).

213

M AN U

194

specific genes

Fkbp5 and Stat5B have well

AC C

EP

TE D

The current literature suggests that disruption of immune-

Glucocorticoid dysfunction is thought to be a contributing factor to the

214

development of PTSD.

One study looked at whether components of GC

215

signaling could be used as a predictor of PTSD development (48).

6

They

ACCEPTED MANUSCRIPT

assessed GC receptor (GR) number, and mRNA expression of Fkbp5, and Gilz

217

(glucocorticoid-induced leucine zipper) in military personnel prior to deployment.

218

They found that high GR number, low Fkbp5 and high Gilz expression predicted

219

PTSD development. This same group further investigated whether GC sensitivity

220

of leukocytes in soldiers predicted the development of PTSD(49). After their

221

return, these soldiers were assessed for level of PTSD, depressive, and fatigue

222

symptoms.

223

development. One study found that GR sensitivity on leukocytes (regardless of

224

GR number, GR subtype, and GR target mRNA levels) and low levels of Fkbp5

225

mRNA were predictors of PTSD development(48). The same group reported that

226

combat soldiers with high sensitivity of GRs on leukocytes pre-deployment were

227

also more likely to develop PTSD(49). Conversely, it has been reported that

228

trauma exposed victims have lower GR sensitivity in peripheral blood monocytes

229

(PBMCs) and low gene expression levels of GR. A recent study performed RNA-

230

seq and microarray analysis on U.S. Marines, both pre- and post-deployment to

231

conflict zones (31). This group conducted higher order analysis of gene networks

232

rather than focus on individual genes, and identified changes in modules related

233

to wound healing and homeostasis. Interestingly, they identified dysregulation of

234

an innate immune response module in soldiers pre-deployment who later

235

developed PTSD (Figure 3).

RI PT

216

TE D

M AN U

SC

Higher sensitivity of GRs on leukocytes predicted PTSD

Alterations of transcription control mechanisms are also affected in PTSD.

237

One study reported decreased levels of blood Dicer transcript that correlated with

238

increased activation of the amygdala in response to fearful stimuli (50). Dicer is

239

an endonuclease that cleaves double-stranded RNA and is responsible for

240

processing pre-microRNA into smaller RNA fragments such as small interfering

241

RNAs and microRNAs. It is interesting to note that another study reported that

242

the Dicer pathway is also altered in a chronic social defeat stress model of

243

depression (51). miRNA dysfunction is an attractive hypothesis as it is possible

244

that a broad transcriptional regulator that lies upstream of many other regulated

245

genes could underlie PTSD.

AC C

EP

236

7

ACCEPTED MANUSCRIPT

These transcriptional studies of peripheral blood have yielded important

247

information, including evidence of impaired glucocorticoid signaling during PTSD,

248

transcriptional changes in immune and inflammatory response genes before and

249

after the onset of PTSD, and elevated proinflammatory factors associated with

250

the pathophysiology of PTSD. These findings could provide biomarkers of illness

251

and treatment response and clues to the pathophysiology of PTSD, but it is clear

252

that studies of blood cannot fully explain brain pathophysiology.

253 Transcript profiling of brain in rodent PTSD models

SC

254

RI PT

246

There are several established animal models for PTSD that are being

256

used to help elucidate the molecular and cellular basis of fear related disorders.

257

A powerful feature of animal models is the ability to manipulate all aspects of a

258

stressor including time interval, duration, and intensity with no preexisting

259

traumatic events and an essentially identical life history leading up to the test (52;

260

53).

261

conditioning, predator-scent-stress models, and social defeat (54).

M AN U

255

Typical animal models of PTSD include variations of classical fear

Studies of gene expression in animal models of PTSD have focused on

263

long-term recovery, with the assumption that sustained alterations could be

264

related to the long-lasting changes that underlie PTSD. One group found major

265

changes in the amygdala of rats subjected to stress-enhanced fear learning

266

(SEFL), in which exposure to stress increases the memory of fearful cues (55).

267

Three weeks after foot shock exposure, gene expression changes were

268

measured in the lateral and basolateral amygdala. Neuron- and glia-specific

269

modules were identified with considerable overlap suggesting a coordination of

270

transcription changes that could be mediated by cross talk between these two

271

cell types (55).

EP

AC C

272

TE D

262

One elegant study used predator-scent-stress to identify and correlate

273

genome-wide transcript changes in amygdala and hippocampus with gene

274

expression changes in peripheral blood (56). They identified transcription factor

275

signaling upstream of gene expression changes to describe convergent signaling

276

across brain, blood, and sex. Notably, they identified the GR signaling pathway

8

ACCEPTED MANUSCRIPT

277

as the only convergent pathway. Several other studies have also examined

278

correlations between brain and blood transcriptome changes in PTSD and other

279

stress models.

280

hippocampus after short and long-term recovery to chronic social defeat or

281

restraint stress (57). They identified regulation of genes and pathways associated

282

with glucocorticoid signaling and inflammation(57). Interestingly, both of these

283

studies (56; 57) found regulation of the gene encoding serum/glucocorticoid

284

kinase 1 (Sgk1) that was recently reported to be down regulated in postmortem

285

dorsolateral PFC of PTSD subjects (58).

SC

RI PT

One study mapped gene expression changes in blood and

Another recent report used a social-stress model where mice were

287

exposed to an aggressive intruder and were given follow-up reminders (exposure

288

without physical contact) of the trauma after rest periods (59). Whole-genome

289

arrays were used to map transcription changes across a wide variety of tissues

290

from both the aggressor, exposed and control mice and across several time

291

points after exposure. They identified alterations of several pathways associated

292

with PTSD, including HPA axis function, modulators of GR activity, neuronal

293

transmission, and fear memory and extinction.

TE D

294

M AN U

286

Chronic social defeat stress (CSDS) has emerged as a widely used psychosocial stressor in animals.

While generally considered a model of

296

depression(60), animals exposed to CSDS exhibit long-term signs of anxiety

297

(61) and submissive animals but not dominant animals display elevated plasma

298

levels of ACTH and glucocorticoids, suggesting psychological, not just physical

299

stress as the cause (62). Two recent papers have used RNA-seq on four brain

300

regions in animals that were either resistant or susceptible to CSDS (63). An

301

integrative network approach was used to identify transcriptional networks and

302

hub genes related to depressive-like symptoms. Transcript changes associated

303

with susceptibility of depressive-like phenotypes showed opposite regulation in

304

the hippocampus versus the PFC. Another study from the same group identified

305

changing transcription patterns for susceptibility and resilience during CSDS and

306

overlaid the changes caused by treatment with the conventional antidepressant

307

imipramine versus the rapid-acting antidepressant ketamine (64). This uniquely

AC C

EP

295

9

ACCEPTED MANUSCRIPT

308

large data set boasts numerous comparisons and provides a useful standard for

309

future transcriptomic studies of rodent PTSD models. Gene expression profiling in animal models of PTSD can provide

311

information on genes and pathways regulated in fear disorders. Because these

312

studies are able to directly examine brain tissue many of the regulated pathways

313

that were uncovered were different from those identified in peripheral blood.

314

Transcripts involved in neuromodulation and neurotransmitter regulation were

315

identified in several studies (55; 59; 65). It is interesting to note that the animal

316

gene expression studies also identified dysregulation in glucocorticoid and

317

inflammatory processes similar to those identified in whole blood (56; 59; 66).

318

However, it is important to point out the obvious caveats associated with animal

319

studies of complex disorders such as PTSD. Rodents are usually inbred and

320

lack genetic diversity, which is likely a major contributing factor to the onset of

321

PTSD pathology. Therefore while these results suggest that it is possible to

322

validate some molecular changes observed in blood and brain, it is necessary to

323

directly examine transcriptional changes occurring in humans with PTSD.

325

Transcript profiles of human post-mortem PTSD brain

TE D

324

M AN U

SC

RI PT

310

Human post-mortem genomic studies of brain tissue promise the best

327

source of information to unravel the complex brain molecular underpinnings of

328

PTSD. Unfortunately, there have been only a few such studies (see published

329

gene expression changes in Table 2, Figure 2). Here we will review the current

330

PTSD post-mortem studies and discuss the design and impact of future studies.

AC C

331

EP

326

The first PTSD post-mortem study examined a single transcript target in a

332

small cohort of subjects, identified up regulation of a multifunction protein p11,

333

that interacts with 5-HT receptors in dorsolateral PFC (67). This same PTSD

334

cohort was also subjected to the first multi-transcript post-mortem gene

335

expression profiling study (58).

336

significant up regulation of 42 transcripts and down regulation of 231 transcripts;

337

one of the most highly regulated genes was Sgk1, which was decreased by 80

338

percent compared to matched control subjects. Further studies in rodent models

Whole genome microarray analysis showed

10

ACCEPTED MANUSCRIPT

339

showed that viral overexpression of dominant negative Sgk1 (dnSGK1) in an

340

auditory fear conditioning model caused higher levels of freezing in the

341

contextual memory recall test three days after conditioning. This indicates that

342

SGK1 inhibition, and presumably decreased Sgk1 expression, leads to enhanced

343

memory of contextual cues associated with fear conditioning.

344

overexpression of dnSGK1 also decreased dendritic spine density in medial PFC

345

and decreased amplitude and frequency of spontaneous mini excitatory post

346

synaptic currents (mEPSCs), suggesting a role for Sgk1 in structural, as well as

347

functional changes in PTSD patients.

SC

RI PT

Interestingly,

One recent study combined live human imaging and post-mortem gene

349

expression studies (47). Photon emission topography (PET) was used to show

350

that mGluR5 binding in PFC is increased in patients with PTSD, suggesting

351

dysfunction in glutamate cycling and/or transmission. Quantitative PCR analysis

352

of post-mortem subgenual PFC did not reveal regulation of mGlur5 transcripts

353

but did show significant increases in SH3 and multiple ankyrin repeat domains 1

354

(Shank1) transcription. Shank1 protein is responsible for tethering mGluR5 to

355

the cell surface and could underlie the increased mGluR5 binding observed in

356

PTSD patients. This post-mortem study also identified down regulation of Fkbp5

357

in subgenual PFC, consistent with reports of decreased Fkbp5 in blood,

358

suggesting a role for glucocorticoid signaling in brain of PTSD subjects.

TE D

M AN U

348

A recent report has highlighted the need for integration of gene expression

360

analysis with genome wide association studies of PTSD (32). This study tested

361

for expression quantitative trait loci (eQTLs) with several positive as well as

362

candidate risk alleles. While the study was ultimately unable to find eQTL for

363

GWAS positive SNPs, it did identify potential candidate PTSD risk SNPs that

364

approached significance. They found that SNP rs363276 in intron 14 of the

365

Solute Carrier Family 18 Member A 2 (Slc18a2) is associated with transcript

366

levels of this gene as well as those of PDZ Domain Containing 8 (Pdzd8) in the

367

dorsolateral PFC and to physiological responses of the amygdala during

368

exposure to fearful and angry faces measured by BOLD fMRI..

369

preliminary study is promising it should be noted that no PTSD regulated genes

AC C

EP

359

11

While this

ACCEPTED MANUSCRIPT

370

identified to date contain risk alleles identified in the largest PTSD GWAS study

371

conducted recently (6). There is now a major effort supported by the VA and under the direction of

373

the National Center for PTSD to develop a brain bank that will provide

374

postmortem tissue from PTSD subjects and matched controls for large scale

375

deep sequencing, transcriptome studies (33). Future studies should focus on key

376

brain regions implicated in PTSD pathophysiology, including the PFC, amygdala,

377

hippocampus, and hypothalamus as well as sub-regions of these areas (Figure

378

1A).

379

increasingly important to develop analysis pipelines that identify the most

380

important alterations in gene networks, including key hub genes in these

381

networks.

382

association studies to transcript levels will likely yield important targets for further

383

study. In addition, integration of mRNA and protein data from mass spectrometry

384

could provide insights into dysregulated biological processes not detected by

385

either technology individually. A further extension will be to integrate DNA

386

methylation studies to provide information on epigenetic changes that contribute

387

to alterations of gene expression in PTSD (Figure 1C). For example, epigenetic

388

alterations could explain how early life trauma increases susceptibility in later life,

389

and could even explain transgenerational susceptibility to psychiatric illness

390

including fear, anxiety, and PTSD (68).

393

SC

M AN U

TE D

As highlighted recently(32), matching previous genome-wide

EP

392

RNA-seq studies yield a large amount of data and it will become

Development of Novel Therapeutic Approaches

AC C

391

RI PT

372

Understanding the transcriptome changes of patients with PTSD is a

394

critical first step in identifying all of the molecular signaling pathways involved in

395

the underlying pathophysiology. In addition, a potential complement to these

396

approaches is cellular reprogramming including induced pluripotent stem cells

397

(iPSCs) that can be generated from accessible somatic cells from patients

398

suffering from psychiatric disorders(69). The exogenous introduction of specific

399

transcription factors can generate region and neurotransmitter-specific neural cell

400

types that would allow for modeling of psychiatric disorders(70). The ability to

12

ACCEPTED MANUSCRIPT

401

examine the neurodevelopment of these induced neurons from PTSD patients

402

compared to controls could shed light on the molecular changes that underlie

403

PTSD. High through-put transcriptomic studies such as RNA-seq can also

405

provide an important approach for investigating drug induced expression

406

changes, and may help to accelerate the process of identifying novel drug

407

targets. In the case of PTSD, it would be particularly interesting to examine

408

changes occurring in synaptic or glucocorticoid related transcripts. For example,

409

it

410

dexamethasone dynamically regulates and normalizes Fkbp5 transcript levels in

411

brain tissue of animal models (71) and peripheral blood taken from depressed

412

human patients (72). These types of studies can be further refined by targeting

413

brain regions that are dysfunctional in PTSD (Figure 1A). For example, based on

414

evidence that extinction of traumatic/fearful events requires connectivity of

415

infralimbic PFC and amygdala (16), we proposed that the rapid acting

416

antidepressant ketamine, which increases synaptic connections in the infralimbic

417

PFC, would enhance fear extinction learning. Consistent with this hypothesis, we

418

have reported that pretreatment with ketamine enhances fear extinction learning,

419

a preclinical model with relevance for understanding the neurobiological

420

mechanisms required for extinction of traumatic events(73). This finding is also

421

consistent with clinical studies reporting that ketamine rapidly decreases

422

symptom severity in PTSD patients (74). This type of information coupled with

423

transcriptome and epigenetic analysis will provide critical information for

424

development of novel therapeutic interventions.

426 427

that

treatment

with

the

synthetic

SC

shown

glucocorticoid

EP

TE D

M AN U

been

AC C

425

has

RI PT

404

Conclusion and Summary Studies to characterize the gene transcription changes that are involved in

428

PTSD are still at an early stage, but a hypothesis is starting to emerge from

429

currently available results (Figure 3) that is consistent with recent models of

430

glucocorticoid signaling and Fkbp5

431

patients that are susceptible to or have PTSD. Low blood levels of Fkbp5 and

(1; 42; 75).

13

GC levels are low in both

ACCEPTED MANUSCRIPT

432

Gilz (an ant-inflammatory protein regulated by GC signaling) in conjunction with

433

altered transcription levels of key innate immune system components predicts

434

PTSD susceptibility. This model further shows that after trauma and onset of

435

PTSD GC and Fkbp5 levels remain low (levels of Sgk1 are currently being

436

examined).

437

dissociation of GR and FKBP5, and subsequent translocation of GC-GR complex

438

to regulate gene transcription.

439

signaling to the nucleus, which then increases Fkbp5 and Sgk1 transcription

440

through the GC response element (GRE). In PTSD, low levels of Fkbp5 and

441

Sgk1 have been reported in post-mortem PFC. This is expected based on the

442

decreased availability of GR after PTSD onset.

443

transcription of many genes that contain GREs, as well as other downstream

444

signaling pathways. It is possible that these aberrant transcriptional programs

445

are related to the observed hypofunction of the PFC, but the mechanistic link

446

between low Fkbp5/Sgk1 expression and PFC hypofunction remains unknown.

447

Importantly, PFC hypofunction results in hyperfunction of the amygdala and

448

contributes to PTSD symptoms, including increased anxiety and fear memory

449

and decreased fear extinction.

RI PT

Under basal conditions GC binds to it’s receptor(GR), causing

SC

This includes feedback that increases GR

TE D

M AN U

This could potentially effect

Gene abundance represents only part of the complexity of the

451

transcriptome. RNA splicing and RNA editing, as well as expression levels (76)

452

are all critically important for normal brain function and it is likely that dysfunction

453

in these processes contribute to the pathophysiology of PTSD. Studies in blood

454

have

455

dysregulation of GR sensitivity, innate immune, and inflammation pathways.

456

Interestingly, many of these findings appear to be confirmed by animal models of

457

PTSD.

458

demonstrate some agreement with whole blood of PTSD patients and animal

459

model studies. Further, we will need to determine how gene expression changes

460

observed are influenced by risk variants identified by GWAS. Along this same

461

line, it will be important to determine if the changes in transcript levels are a

462

cause or a consequence of PTSD . It is likely that we are at the very early stages

EP

450

important

preliminary

information

providing

evidence

for

AC C

yielded

While limited in number, post-mortem studies of PTSD brain tissue

14

ACCEPTED MANUSCRIPT

463

of investigation and more extensive deep sequencing of PTSD brain subregions

464

will identify complex gene networks that underlie the pathophysiology of PTSD,

465

which could provide novel targets for therapeutic intervention.

466

AC C

EP

TE D

M AN U

SC

RI PT

467

15

ACCEPTED MANUSCRIPT

468

Acknowledgements

469

This work is supported by NIMH R01MH93897 (RSD), the State of Connecticut,

470

Yale University, and the VA National Center for PTSD.

471 R.S. Duman has consulted and/or received research support from Naurex, Lilly,

473

Forest, Johnson & Johnson, Taisho, and Sunovion. M.J. Girgenti reports no

474

biomedical financial interests or potential conflicts of interest.

RI PT

472

475

SC

476 477

EP

TE D

REFERENCES 1. Banerjee SB, Morrison FG, Ressler KJ (2017): ARTICLE IN PRESS. Neurosci Lett. 1–8. 2. Schizophrenia Working Group of the Psychiatric Genomics Consortium (2014): Biological insights from 108 schizophrenia-associated genetic loci. Nature. 511: 421–427. 3. Major Depressive Disorder Working Group of the Psychiatric GWAS Consortium, Ripke S, Wray NR, Lewis CM, Hamilton SP, Weissman MM, et al. (2013): A mega-analysis of genome-wide association studies for major depressive disorder. Molecular Psychiatry. 18: 497–511. 4. Stein MB, Chen C-Y, Ursano RJ, Cai T, Gelernter J, Heeringa SG, et al. (2016): Genome-wide Association Studies of Posttraumatic Stress Disorder in 2 Cohorts of US Army Soldiers. JAMA Psychiatry. 73: 695–704. 5. Parsons RG, Ressler KJ (2013): Implications of memory modulation for posttraumatic stress and fear disorders. Nature Publishing Group. 16: 146–153. 6. Duncan LE, Ratanatharathorn A, Aiello AE, Almli LM, Amstadter AB, AshleyKoch AE, et al. (2017): Largest GWAS of PTSD (N=20070) yields genetic overlap with schizophrenia and sex differences in heritability. 1–8. 7. Pape H-C, Paré D (2010): Plastic synaptic networks of the amygdala for the acquisition, expression, and extinction of conditioned fear. Physiol Rev. 90: 419–463. 8. LeDoux JE, Iwata J, Cicchetti P, Reis DJ (1988): Different projections of the central amygdaloid nucleus mediate autonomic and behavioral correlates of conditioned fear. J Neurosci. 8: 2517–2529. 9. Wilensky AE, Schafe GE, Kristensen MP, LeDoux JE (2006): Rethinking the fear circuit: the central nucleus of the amygdala is required for the acquisition,

AC C

479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508

M AN U

478

16

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

consolidation, and expression of Pavlovian fear conditioning. Journal of Neuroscience. 26: 12387–12396. 10. Ciocchi S, Herry C, Grenier F, Wolff SBE, Letzkus JJ, Vlachos I, et al. (2010): Encoding of conditioned fear in central amygdala inhibitory circuits. Nature. 468: 277–282. 11. Haubensak W, Kunwar PS, Cai H, Ciocchi S, Wall NR, Ponnusamy R, et al. (2010): Genetic dissection of an amygdala microcircuit that gates conditioned fear. Nature. 468: 270–276. 12. Etkin A, Wager TD (2007): Functional neuroimaging of anxiety: a metaanalysis of emotional processing in PTSD, social anxiety disorder, and specific phobia. Am J Psychiatry. 164: 1476–1488. 13. Corcoran KA, Desmond TJ, Frey KA, Maren S (2005): Hippocampal inactivation disrupts the acquisition and contextual encoding of fear extinction. Journal of Neuroscience. 25: 8978–8987. 14. Heldt SA, Stanek L, Chhatwal JP, Ressler KJ (2007): Hippocampus-specific deletion of BDNF in adult mice impairs spatial memory and extinction of aversive memories. Molecular Psychiatry. 12: 656–670. 15. Quirk GJ, Russo GK, Barron JL, Lebron K (2000): The role of ventromedial prefrontal cortex in the recovery of extinguished fear. J Neurosci. 20: 6225– 6231. 16. Milad MR, Quirk GJ (2002): Neurons in medial prefrontal cortex signal memory for fear extinction. Nature. 420: 70–74. 17. Peters J, Dieppa-Perea LM, Melendez LM, Quirk GJ (2010): Induction of fear extinction with hippocampal-infralimbic BDNF. Science. 328: 1288–1290. 18. Phelps EA, Delgado MR, Nearing KI, LeDoux JE (2004): Extinction learning in humans: role of the amygdala and vmPFC. Neuron. 43: 897–905. 19. Milad MR, Wright CI, Orr SP, Pitman RK, Quirk GJ, Rauch SL (2007): Recall of fear extinction in humans activates the ventromedial prefrontal cortex and hippocampus in concert. Biol Psychiatry. 62: 446–454. 20. Bremner JD (2007): Neuroimaging in posttraumatic stress disorder and other stress-related disorders. Neuroimaging Clin N Am. 17: 523–38– ix. 21. Green RE, Krause J, Ptak SE, Briggs AW, Ronan MT, Simons JF, et al. (2006): Analysis of one million base pairs of Neanderthal DNA. Nature. 444: 330–336. 22. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B (2008): Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Meth. 5: 621– 628. 23. Wilhelm BT, Landry J-R (2009): RNA-Seq-quantitative measurement of expression through massively parallel RNA-sequencing. Methods. 48: 249– 257. 24. Johnson DS, Mortazavi A, Myers RM, Wold B (2007): Genome-wide mapping of in vivo protein-DNA interactions. Science. 316: 1497–1502. 25. Barski A, Cuddapah S, Cui K, Roh T-Y, Schones DE, Wang Z, et al. (2007): High-Resolution Profiling of Histone Methylations in the Human Genome. Cell. 129: 823–837. 26. Jaffe AE, Gao Y, Deep-Soboslay A, Tao R, Hyde TM, Weinberger DR,

AC C

509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554

17

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Kleinman JE (2016): Mapping DNA methylation across development, genotype and schizophrenia in the human frontal cortex. Nature Publishing Group. 19: 40–47. 27. Li M, Jaffe AE, Straub RE, Tao R, Shin JH, Wang Y, et al. (2016): A humanspecific AS3MT isoform and BORCS7 are molecular risk factors in the 10q24.32 schizophrenia-associated locus. Nat Med. 22: 649–656. 28. Pantazatos SP, Huang Y-Y, Rosoklija GB, Dwork AJ, Arango V, Mann JJ (2016): Whole-transcriptome brain expression and exon-usage profiling in major depression and suicide: evidence for altered glial, endothelial and ATPase activity. Molecular Psychiatry. doi: 10.1038/mp.2016.130. 29. Kohen R, Dobra A, Tracy JH, Haugen E (2014): Transcriptome profiling of human hippocampus dentate gyrus granule cells in mental illness. Translational Psychiatry. 4: e366. 30. Farris SP, Arasappan D, Hunicke-Smith S, Harris RA, Mayfield RD (2015): Transcriptome organization for chronic alcohol abuse in human brain. Molecular Psychiatry. 20: 1438–1447. 31. Breen MS, Maihofer AX, Glatt SJ, Tylee DS, Chandler SD, Tsuang MT, et al. (2015): Gene networks specific for innate immunity define post-traumatic stress disorder. Molecular Psychiatry. 20: 1538–1545. 32. Bharadwaj RA, Jaffe AE, Chen Q, Deep-Soboslay A, Goldman AL, Mighdoll MI, et al. (2016): Genetic risk mechanisms of posttraumatic stress disorder in the human brain. J Neurosci Res. doi: 10.1002/jnr.23957. 33. Friedman MJ, Huber BR, Brady CB, Ursano RJ, Benedek DM, Kowall NW, et al. (n.d.): VA's National PTSD Brain Bank: A national Resource for Research. Curr Psychiatry Reports. In Press. 34. Thomson CA, McColl A, Cavanagh J, Graham GJ (2014): Peripheral inflammation is associated with remote global gene expression changes in the brain. Journal of Neuroinflammation. 11:73: 1–16. 35. Michopoulos V, Powers A, Gillespie CF, Ressler KJ, Jovanovic T (2016): Inflammation in Fear- and Anxiety-Based Disorders: PTSD, GAD, and Beyond. Neuropsychopharmacology. 1–17. 36. Pace TWW, Wingenfeld K, Schmidt I, Meinlschmidt G, Hellhammer DH, Heim CM (2012): Increased peripheral NF-κB pathway activity in women with childhood abuse-related posttraumatic stress disorder. Brain Behav Immun. 26: 13–17. 37. Zieker J, Zieker D, Jatzko A, Dietzsch J, Nieselt K, Schmitt A, et al. (2007): Differential gene expression in peripheral blood of patients suffering from post-traumatic stress disorder. Molecular Psychiatry. 12: 116–118. 38. Mehta D, Gonik M, Klengel T, Rex-Haffner M, Menke A, Rubel J, et al. (2011): Using polymorphisms in FKBP5 to define biologically distinct subtypes of posttraumatic stress disorder: evidence from endocrine and gene expression studies. Arch Gen Psychiatry. 68: 901–910. 39. Neylan TC, Sun B, Rempel H, Ross J, Lenoci M, O'Donovan A, Pulliam L (2011): Suppressed monocyte gene expression profile in men versus women with PTSD. Brain Behav Immun. 25: 524–531. 40. Yehuda R, Cai G, Golier JA, Sarapas C, Galea S, Ising M, et al. (2009):

AC C

555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600

18

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Gene expression patterns associated with posttraumatic stress disorder following exposure to the World Trade Center attacks. Biol Psychiatry. 66: 708–711. 41. Sarapas C, Cai G, Bierer LM, Golier JA, Galea S, Ising M, et al. (2011): Genetic markers for PTSD risk and resilience among survivors of the World Trade Center attacks. Dis Markers. 30: 101–110. 42. Binder EB (2009): The role of FKBP5, a co-chaperone of the glucocorticoid receptor in the pathogenesis and therapy of affective and anxiety disorders. Psychoneuroendocrinology. 34 Suppl 1: S186–95. 43. Maddox SA, Schafe GE, Ressler KJ (2013): Exploring epigenetic regulation of fear memory and biomarkers associated with post-traumatic stress disorder. Frontiers in Psychiatry. 4:62: 1–15. 44. Pratt WB, Toft DO (1997): Steroid receptor interactions with heat shock protein and immunophilin chaperones. Endocr Rev. 18: 306–360. 45. Denny WB, Valentine DL, Reynolds PD, Smith DF, Scammell JG (2000): Squirrel monkey immunophilin FKBP51 is a potent inhibitor of glucocorticoid receptor binding. Endocrinology. 141: 4107–4113. 46. Zannas AS, Binder EB (2013): Gene-environment interactions at the FKBP5locus: sensitive periods, mechanisms and pleiotropism. Genes, Brain and Behavior. 13: 25–37. 47. Holmes SE, Girgenti MJ, Davies MT, Pietrzak RH, DellaGioia N, Nabulsi N, et al. (2017): Altered PFC mGluR5 in PTSD: evidence from in vivo and post mortem work. (Vol. 114), Proc Natl Acad Sci USA. 48. van Zuiden M, Geuze E, Willemen HLDM, Vermetten E, Maas M, Amarouchi K, et al. (2012): Glucocorticoid receptor pathway components predict posttraumatic stress disorder symptom development: a prospective study. Biol Psychiatry. 71: 309–316. 49. van Zuiden M, Heijnen CJ, Maas M, Amarouchi K, Vermetten E, Geuze E, Kavelaars A (2012): Glucocorticoid sensitivity of leukocytes predicts PTSD, depressive and fatigue symptoms after military deployment: A prospective study. Psychoneuroendocrinology. 37: 1822–1836. 50. Wingo AP, Almli LM, Stevens JS, Stevens JJ, Klengel T, Uddin M, et al. (2015): DICER1 and microRNA regulation in post-traumatic stress disorder with comorbid depression. Nature Communications. 6: 10106. 51. Dias C, Feng J, Sun H, Shao NY, Mazei-Robison MS, Damez-Werno D, et al. (2014): β-catenin mediates stress resilience through Dicer1/microRNA regulation. Nature. 516: 51–55. 52. Flandreau EI, Toth M (2017): Animal Models of PTSD: A Critical Review. In:. Current Topics in Behavioral Neurosciences, 5 ed., Current Topics in Behavioral Neurosciences. (Vol. 62), Berlin, Heidelberg: Springer Berlin Heidelberg, p 674. 53. Paré D (2013): Animal models of post-traumatic stress disorder: face validity. Frontiers in Neuroscience. 7:89: 1–14. 54. Zovkic IB, Sweatt JD (2013): Epigenetic mechanisms in learned fear: implications for PTSD. Neuropsychopharmacology. 38: 77–93. 55. Ponomarev I, Rau V, Eger EI, Harris RA, Fanselow MS (2010): Amygdala

AC C

601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646

19

ACCEPTED MANUSCRIPT

EP

TE D

M AN U

SC

RI PT

Transcriptome and Cellular Mechanisms Underlying Stress-Enhanced Fear Learning in a Rat Model of Posttraumatic Stress Disorder. Neuropsychopharmacology. 35: 1402–1411. 56. Daskalakis NP, Cohen H, Cai G, Buxbaum JD, Yehuda R (2014): Expression profiling associates blood and brain glucocorticoid receptor signaling with trauma-related individual differences in both sexes. Proc Natl Acad Sci USA. 111: 13529–13534. 57. Gray JD, Rubin TG, Hunter RG, McEwen BS (2014): Hippocampal gene expression changes underlying stress sensitization and recovery. Molecular Psychiatry. 19: 1171–1178. 58. Licznerski P, Duric V, Banasr M, Alavian KN, Ota KT, Kang HJ, et al. (2015): Decreased SGK1 Expression and Function Contributes to Behavioral Deficits Induced by Traumatic Stress. (K. Ressler, editor) PLoS Biol. 13: e1002282. 59. Muhie S (2015): Brain transcriptome profiles in mouse model simulating features of post-traumatic stress disorder. Molecular Brain. 8:14: 1–21. 60. Krishnan V, Berton O, Nestler E (2008): The use of animal models in psychiatric research and treatment. Am J Psychiatry. 165: 1109. 61. Huhman KL (2006): Social conflict models: can they inform us about human psychopathology? Horm Behav. 50: 640–646. 62. Huhman KL, Moore TO, Mougey EH, Meyerhoff JL (1992): Hormonal responses to fighting in hamsters: separation of physical and psychological causes. Physiol Behav. 51: 1083–1086. 63. Bagot RC, Cates HM, Purushothaman I, Lorsch ZS, Walker DM, Wang J, et al. (2016): Circuit-wide Transcriptional Profiling Reveals Brain RegionSpecific Gene Networks Regulating Depression Susceptibility. Neuron. 90: 969–983. 64. Bagot RC, Cates HM, Purushothaman I, Vialou V, Heller EA, Yieh L, et al. (2017): Priority Communication. Biol Psychiatry. 81: 285–295. 65. Andero R, Brothers SP, Jovanovic T, Chen YT, Salah-Uddin H, Cameron M, et al. (2013): Amygdala-dependent fear is regulated by Oprl1 in mice and humans with PTSD. Sci Transl Med. 5: 188ra73. 66. Le-Niculescu H, Balaraman Y, Patel SD, Ayalew M, Gupta J, Kuczenski R, et al. (2011): Convergent functional genomics of anxiety disorders: translational identification of genes, biomarkers, pathways and mechanisms. Translational Psychiatry. 1: e9. 67. Zhang L, Li H, Su TP, Barker JL, Maric D, Fullerton CS, et al. (2008): p11 is up-regulated in the forebrain of stressed rats by glucocorticoid acting via two specific glucocorticoid response elements in the p11 promoter. Neuroscience. 153: 1126–1134. 68. Nestler EJ (2016): Transgenerational Epigenetic Contributions to Stress Responses: Fact or Fiction? PLoS Biol. 14: e1002426. 69. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S (2007): Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 131: 861–872. 70. Brennand K, Savas JN, Kim Y, Tran N, Simone A, Hashimoto-Torii K, et al. (2015): Phenotypic differences in hiPSC NPCs derived from patients with

AC C

647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692

20

ACCEPTED MANUSCRIPT

723 724 725 726 727 728 729 730 731 732 733 734 735 736 737

RI PT

SC

M AN U

TE D EP

722

schizophrenia. Molecular Psychiatry. 20: 361–368. 71. Sawamura T, Klengel T, Armario A, Jovanovic T, Norrholm SD, Ressler KJ, Andero RUL (2015): Dexamethasone Treatment Leads to Enhanced Fear Extinction and Dynamic Fkbp5 Regulation in Amygdala. 41: 832–846. 72. Menke A, Arloth J, Pütz B, Weber P, Klengel T, Mehta D, et al. (2012): Dexamethasone stimulated gene expression in peripheral blood is a sensitive marker for glucocorticoid receptor resistance in depressed patients. Neuropsychopharmacology. 37: 1455–1464. 73. Girgenti MJ, Ghosal S, Lopresto D, Taylor JR, Duman RS (2016): Ketamine accelerates fear extinction via mTORC1 signaling. Neurobiol Dis. 100: 1–8. 74. Feder A, Parides MK, Murrough JW, Perez AM, Morgan JE, Saxena S, et al. (2014): Efficacy of Intravenous Ketamine for Treatment of Chronic Posttraumatic Stress Disorder. 71: 681. 75. Daskalakis NP, Cohen H, Nievergelt CM, Baker DG, Buxbaum JD, Russo SJ, Yehuda R (2016): New translational perspectives for blood-based biomarkers of PTSD: From glucocorticoid to immune mediators of stress susceptibility. Exp Neurol. 284: 133–140. 76. Dillman AA, Hauser DN, Gibbs JR, Nalls MA, McCoy MK, Rudenko IN, et al. (2013): mRNA expression, splicing and editing in the embryonic and adult mouse cerebral cortex. Nature Publishing Group. 16: 499–506. 77. Matić G, Milutinović DV, Nestorov J, Elaković I, Jovanović SM, Perišić T, et al. (2013): Lymphocyte glucocorticoid receptor expression level and hormone-binding properties differ between war trauma-exposed men with and without PTSD. Prog Neuropsychopharmacol Biol Psychiatry. 43: 238– 245.

Figure Legends Figure 1: RNA-seq of human post-mortem brain tissue to identify molecular changes in PTSD neuropathology. (A) Several brain regions have been implicated in PTSD based on imaging studies of PTSD patients, including the vmPFC, amygdala, and hippocampus. To identify the molecular and cellular mechanisms underlying the neurobiology of PTSD, next generation analysis of gene expression must be conducted. (B) We have begun using high through-put RNA-seq to identify the transcriptomic changes in three cortical regions (subgenual, dorsolateral, and orbitofrontal), amygdala and hippocampus. A illustration of the primary steps in the RNA-seq process. Briefly, mRNA is isolated from dissected region-specific brain tissue from PTSD and matched controls. cDNA synthesis and adapter ligation are performed on the RNA and loaded onto a flow cell containing a lawn of matching adapters, allowing the

AC C

693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721

21

ACCEPTED MANUSCRIPT

RI PT

individual cDNAs to bind. Cluster growth begins and fluorescently tagged nucleotides are added to the matching base pair of the cDNA. The nucleotides are imaged and the sequence of the colored nucleotides allows for base calling or sequencing. The reads generated are then matched to the current human genome (Hg19) and differential gene expression is measured. (C) Volcano matrix plots for all gene expression changes in PTSD, MDD, and matched controls in human subgenual PFC. Red dots indicate statistically significant genes. RNAseq informs the proteomics assayed using tandem mass spectrometry for protein expression and alternative splicing. Overlay of CpG island methylation on detected transcript changes allows for further identification of the epigenetic alterations that contribute to differential gene expression.

TE D

M AN U

SC

Figure 2: PTSD associated changes in gene expression in human brain and peripheral blood. Decreased levels of Fkbp5 and Sgk1 gene expression have been identified in subgenual PFC and dorsolateral PFC indicating dysregulation of glucocorticoid signaling in CNS. The mGlur5 anchoring protein Shank1 is increased in subgenual PFC. This correlates with observed increases in mGlur5 protein binding availability, believed to be associated with the avoidance symptoms of PTSD. Studies of peripheral blood have identified gene expression changes associated with susceptibility to developing PTSD (left), as well as transcript changes in patients that were diagnosed with PTSD at the time of analysis (right). In some cases the gene lists were too long to be included in the figure. The differentially expressed genes have been summarized as reported pathways. Collectively, these findings suggest that a predisposition to developing PTSD may be attributed to aberrant expression of innate immune response leading to glucocortidcoid dysfunction during the onset of the disease.

EP

Figure 3: Proposed Model of Aberrant Transcription in PTSD. Normal levels of GCs are necessary for regulating transcription throughout the body and the CNS. Under basal conditions, GC receptor (GR) is bound to FKBP5 in the cytoplasm and is inactive until sufficient levels of GCs result the bound GC-GR state, leading to dissociation from FKBP5. The activated GC-GR complex then undergoes translocation to the nucleus where GR binds to the GC response element (GRE) in the genome to regulate gene transcription. Low levels of GCs have been reported in patients with PTSD and a preexisting low level prior to trauma is thought to predispose or create a susceptible condition in some patients to develop PTSD. Low levels of GCs could then results in decreased transcriptional regulation of genes with GREs, including Fkbp5 and Sgk1 and their downstream effectors. This may contribute to the observed hypofunction of the vmPFC in PTSD patients, which in turn limits top-down control and hyperfunction of the amygdala. Increased or uncontrolled function of the amygdala is directly related to the symptoms of PTSD, including increased fear learning and decreased fear extinction. There is currently no direct tie between alteration in CNS transcription and the reported changes in blood of the immune and inflammatory response in PTSD, though it is likely that these changes affect one another (indicated by the bidirectional area between blood and PFC).

AC C

738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783

22

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

784

23

ACCEPTED MANUSCRIPT Perturbed Pathway(s) IGF and interleukin

RNA Source Individuals with PTSD

signaling FKBP5, STAT and MHC

Survivors of 9/11

RNA Analysis

Study

Customized

Zieker, et al.,

Microarray

2007 (37)

qRT-PCR

Yehuda, et al., 2009 (40)

Monocyte gene

Trauma victims

signaling

Microarray

Nuclear Factor and

Survivors of 9/11

pathway signaling with

income bracket

FKBP5 SNPs interacts Nuclear factor-ΚB

Female child abuse

signaling increased

victims with PTSD

High expression of GR,

Military personnel

low FKBP5 and high

TE D

GILZ predict PTSD development

Microarray

Sarapas, et al.

Trauma exposed

decreased in PBMCs

military personnel

Changes in Innate

Military personnel

AC C

EP

GR signaling was

Dicer1 expression

Traumatized

reduced in PTSD

individuals. Partial

Mehta, et al.,

2011 (38)

M AN U

Individuals in low

PTSD development

2011 (39)

2011 (41)

Interleukin and Stat

expression predicts

Microarray

Neylan et al.,

SC

Stat5b signaling

Immunity gene

Customized

RI PT

signaling

qRT-PCR

qRT-PCR

qRT-PCR

Pace, et al., 2012 (36) van Zuiden, et al., 2012 (48)

Matic, et al., 2013 (77)

Microarray

Breen, et al.,

and RNA-seq

2015 (31)

Microarray

Wingo, et al., 2015 (50)

cohort PTSD with comorbid depression

Table 1. Transcriptomic studies from human peripheral blood

ACCEPTED MANUSCRIPT

Perturbed Pathway

RNA Source

RNA Analysis

Study

Prefrontal Cortex Candidate transcript

Zhang, et al., 2008

signaling

(Area 46)

qRT-PCR

(67)

SGK1 signaling

Dorsolateral

Whole Genome

Licznerski, et al.,

PFC

Microarray

Glutamate and

Subgenual PFC

Candidate transcript

Glucocorticoid

(Area 25)

qRT-PCR

Significant eQTL

Healthy

RNA-sequencing

association with

Dorsolateral

PTSD risk SNPs

PFC

RI PT

Glucocorticoid

2015 (58)

Holmes, et al., 2017 (47)

SC

signaling

Bharadwaj, et al.,

M AN U

2016 (32)

AC C

EP

TE D

Table 2. Transcriptomic studies from post mortem PTSD brain tissue

ACCEPTED MANUSCRIPT

Figure 1.! A.!

Dorsolateral PFC!

Hippocampus:! CA1, CA3, dentate ! gyrus!

RI PT

Orbitofrontal PFC! Subgenual PFC! Orbitofrontal PFC!

AAAA!

2. cDNA Synthesis/ Adapter Ligation!

AAAA!

RNA-seq!

C.! Mass Spectrometry! Proteomics!

TE D

1. mRNA Isolation!

M AN U

B.!

SC

Amygdala:! BLA, CeA!

TTTT!

EP

3. Cluster Growth!

AC C

T! A! C! G! G! C! A! T!

4. Base Calling! AGCT!

Bisulfite Chip! CpG Methylation!

Figure 2.!

ACCEPTED MANUSCRIPT

RI PT

Brain-PTSD! SGK1 !P11! FKBP5 !PZDZ8! SHANK1 !SLC18a2!

SC

Blood-PTSD! FKBP5, IL18, IL16, CSF1, ! STAT5B, MHCII, NFIA, DICER, PF4, SDPR, HIST1H2AC !! Innate Immunity Pathway! Stat Pathway! IGF2! Glutamate Transport!

AC C

EP

TE D

M AN U

Blood- Susceptibility to PTSD! Innate Immunity GR ! !! FKBP5 Pathway! !!   GILZ  

Figure 3.!

ACCEPTED MANUSCRIPT

Basal Conditions!

GC! FKBP5!

GILZ!

?!SGK1!

GC! GR!

Innate Immunity!

FKBP5!

M AN U

PFC neuron!

GC! FKBP5! ?!SGK1! blood! vessel!

SC

GC!

GR+ FKBP5 !

periphery!

RI PT

GC !

endothelial! cells!

PTSD!

PTSD Susceptible!

GR!

GR ! FKBP5! SGK1!

? FKBP5! ? SGK1!

TE D

FKBP5! SGK1!

GRE!

GRE!

GRE!

Transcript Changes! ?!

PFC Hypofunction!

EP

PFC Top Down Control!

AC C

CNS! ?! Inflammatory! ! Factors!

GC!

?! Amygdala! Neuron!

Amygdala Function!

Amygdala Hyperfunction!

Normal control of ! fear, extinction, and emotion.!

?!

GRE!

PTSD Symptoms! Enhanced Fear Memory! Decreased Fear Extinction!