1
|
Reed EC, Silva VA, Giebel KR, Natour T, Lauten TH, Jojo CN, Schleiker AE, Case AJ. Hemoglobin alpha is a redox-sensitive mitochondrial-related protein in T-lymphocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613298. [PMID: 39345360 PMCID: PMC11429782 DOI: 10.1101/2024.09.16.613298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Hemoglobin subunits, which form the well-characterized, tetrameric, oxygen-carrying protein, have recently been described to be expressed in various non-canonical cell types. However, the exact function of hemoglobin subunits within these cells remains to be fully elucidated. Herein, we report for the first time, the expression of hemoglobin alpha-a1 (Hba-a1) in T-lymphocytes and describe its role as a mitochondrial-associated antioxidant. Within naïve T-lymphocytes, Hba-a1 mRNA and HBA protein are present and highly induced by redox perturbations, particularly those arising from the mitochondria. Additionally, preliminary data using a T-lymphocyte specific Hba-a1 knock-out mouse model indicated that the loss of Hba-a1 led to an exacerbated production of mitochondrial reactive oxygen species and inflammatory cytokines after a stress challenge, further supporting the role of HBA acting to buffer the mitochondrial redox environment. Interestingly, we observed Hba-a1 expression to be significantly upregulated or downregulated depending on T-lymphocyte polarization and metabolic state, which appeared to be controlled by both transcriptional regulation and chromatin remodeling. Altogether, these data suggest Hba-a1 may function as a crucial mitochondrial-associated antioxidant and appears to possess critical and complex functions related to T-lymphocyte activation and differentiation.
Collapse
Affiliation(s)
- Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Valeria A. Silva
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Kristen R. Giebel
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Tatlock H. Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Caroline N. Jojo
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Abigail E. Schleiker
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
2
|
Flores A, Nguyen NM, Devanaboyina M, Sanketh S, Athota P, Jagadesan S, Guda C, Yelamanchili SV, Pendyala G. Neurobehavioral Characterization of Perinatal Oxycodone-Exposed Offspring in Early Adolescence. J Neuroimmune Pharmacol 2024; 19:29. [PMID: 38874861 DOI: 10.1007/s11481-024-10129-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 05/25/2024] [Indexed: 06/15/2024]
Abstract
The opioid epidemic has received considerable attention, but the impact on perinatal opioid-exposed (POE) offspring remains underexplored. This study addresses the emerging public health challenge of understanding and treating POE children. We examined two scenarios using preclinical models: offspring exposed to oxycodone (OXY) in utero (IUO) and acute postnatal OXY (PNO). We hypothesized exposure to OXY during pregnancy primes offspring for neurodevelopmental deficits and severity of deficits is dependent on timing of exposure. Notable findings include reduced head size and brain weight in offspring. Molecular analyses revealed significantly lower levels of inflammasome-specific genes in the prefrontal cortex (PFC). Gene Set Enrichment Analysis (GSEA) and Ingenuity Pathway Analysis (IPA) highlighted the enrichment of genes associated with mitochondrial and synapse dysfunction in POE offspring. Western blot analysis validated IPA predictions of mitochondrial dysfunction in PFC-derived synaptosomes. Behavioral studies identified significant social deficits in POE offspring. This study presents the first comparative analysis of acute PNO- and IUO-offspring during early adolescence finding acute PNO-offspring have considerably greater deficits. The striking difference in deficit severity in acute PNO-offspring suggests that exposure to opioids in late pregnancy pose the greatest risk for offspring well-being.
Collapse
Affiliation(s)
- Adrian Flores
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
- Department of Cellular and Integrative Physiology, UNMC, Omaha, NE, 68198, USA
| | - Nghi M Nguyen
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, 68198, USA
- Child Health Research Institute, Omaha, NE, 68198, USA
| | - Murali Devanaboyina
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
| | - Samarth Sanketh
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
| | - Pranavi Athota
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
| | | | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, 68198, USA
| | - Sowmya V Yelamanchili
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, 68198, USA
- National Strategic Research Institute, UNMC, Omaha, NE, USA
| | - Gurudutt Pendyala
- Department of Anesthesiology, University of Nebraska Medical Center (UNMC), Omaha, NE, 68198, USA.
- Department of Genetics, Cell Biology and Anatomy, UNMC, Omaha, NE, 68198, USA.
- Child Health Research Institute, Omaha, NE, 68198, USA.
- National Strategic Research Institute, UNMC, Omaha, NE, USA.
| |
Collapse
|
3
|
Lauten TH, Elkhatib SK, Natour T, Reed EC, Jojo CN, Case AJ. Beta-adrenergic signaling and T-lymphocyte-produced catecholamines are necessary for interleukin 17A synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597633. [PMID: 38895227 PMCID: PMC11185643 DOI: 10.1101/2024.06.05.597633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background Post-traumatic stress disorder (PTSD) is a debilitating psychological disorder that also presents with neuroimmune irregularities. Patients display elevated sympathetic tone and are at an increased risk of developing secondary autoimmune diseases. Previously, using a preclinical model of PTSD, we demonstrated that elimination of sympathetic signaling to T-lymphocytes specifically limited their ability to produce pro-inflammatory interleukin 17A (IL-17A); a cytokine implicated in the development of many autoimmune disorders. However, the mechanism linking sympathetic signaling to T-lymphocyte IL-17A production remained unclear. Methods Using a modified version of repeated social defeat stress (RSDS) that allows for both males and females, we assessed the impact of adrenergic receptor blockade (genetically and pharmacologically) and catecholamine depletion on T-lymphocyte IL-17A generation. Additionally, we explored the impact of adrenergic signaling and T-lymphocyte-produced catecholamines on both CD4+ and CD8+ T-lymphocytes polarized to IL-17A-producing phenotypes ex vivo. Results Only pharmacological inhibition of the beta 1 and 2 adrenergic receptors (β1/2) significantly decreased circulating IL-17A levels after RSDS, but did not impact other pro-inflammatory cytokines (e.g., IL-6, TNF-α, and IL-10). This finding was confirmed using RSDS with both global β1/2 receptor knock-out mice, as well as by adoptively transferring β1/2 knock-out T-lymphocytes into immunodeficient hosts. Furthermore, ex vivo polarized T-lymphocytes produced significantly less IL-17A with the blockade of β1/2 signaling, even in the absence of exogenous sympathetic neurotransmitter supplementation, which suggested T-lymphocyte-produced catecholamines may be involved in IL-17A production. Indeed, pharmacological depletion of catecholamines both in vivo and ex vivo abrogated T-lymphocyte IL-17A production demonstrating the importance of immune-generated neurotransmission in pro-inflammatory cytokine generation. Conclusions Our data depict a novel role for β1/2 adrenergic receptors and autologous catecholamine signaling during T-lymphocyte IL-17A production. These findings provide a new target for pharmacological therapy in both psychiatric and autoimmune diseases associated with IL-17A-related pathology.
Collapse
Affiliation(s)
- Tatlock H. Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Safwan K. Elkhatib
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Caroline N. Jojo
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
4
|
Lauten TH, Natour T, Case AJ. Innate and adaptive immune system consequences of post-traumatic stress disorder. Auton Neurosci 2024; 252:103159. [PMID: 38428324 PMCID: PMC11494466 DOI: 10.1016/j.autneu.2024.103159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/06/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Abstract
In the field of psychiatry, biological markers are rarely, if ever, used in the diagnosis of mental health disorders. Clinicians rely primarily on patient histories and behavioral symptoms to identify specific psychopathologies, which makes diagnosis highly subjective. Moreover, therapies for mental health disorders are aimed specifically at attenuating behavioral manifestations, which overlooks the pathophysiological indices of the disease. This is highly evident in posttraumatic stress disorder (PTSD) where inflammation and immune system perturbations are becoming increasingly described. Further, patients with PTSD possess significantly elevated risks of developing comorbid inflammatory diseases such as autoimmune and cardiovascular diseases, which are likely linked (though not fully proven) to the apparent dysregulation of the immune system after psychological trauma. To date, there is little to no evidence that demonstrates current PTSD therapies are able to reverse the increased risk for psychological trauma-induced inflammatory diseases, which suggests the behavioral and somatic consequences of PTSD may not be tightly coupled. This observation provides an opportunity to explore unique mechanisms outside of the brain that contribute to the long-term pathology of PTSD. Herein, we provide an overview of neuroimmune mechanisms, describe what is known regarding innate and adaptive immunity in PTSD, and suggest new directions that are needed to advance the understanding, diagnosis, and treatment of PTSD moving forward.
Collapse
Affiliation(s)
- Tatlock H Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States; Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States; Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Adam J Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States; Department of Medical Physiology, Texas A&M University, Bryan, TX, United States.
| |
Collapse
|
5
|
Ahmed Z, Tahmin CI, Tahsin CT, Michopoulos V, Mohamed A, Wattero R, Albott S, Cullen KR, Lowe DA, Osborn J, Fonkoue IT. Higher arterial stiffness and blunted vagal control of the heart in young women with compared to without a clinical diagnosis of PTSD. Clin Auton Res 2024; 34:165-175. [PMID: 38324188 PMCID: PMC10947824 DOI: 10.1007/s10286-024-01014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024]
Abstract
PURPOSE Young women are typically thought to be protected from cardiovascular disease (CVD) before menopause. However, posttraumatic stress disorder (PTSD) increases CVD risk in women by up to threefold. Data in predominantly male cohorts point to physiological mechanisms such as vascular and autonomic derangements as contributing to increased CVD risk. The purpose of the study reported here was to determine whether young women diagnosed with PTSD, compared to those without, present with arterial stiffness and impaired autonomic control of the heart. METHODS A total of 73 healthy young women, ranging in age from 18 to 40 years, with a history of trauma exposure were included in this study, 32 with and 41 without a clinical PTSD diagnosis. We measured resting pulse wave velocity (PWV), central hemodynamics, augmentation pressure and augmentation index (AI) via pulse wave analysis using applanation tonometry. Heart rate variability was also assessed via peripheral arterial tone. RESULTS In comparison to controls, women with PTSD showed higher central arterial pressure (mean ± standard deviation: systolic blood pressure 104 ± 8 vs. 97 ± 8 mmHg, p < 0.001; diastolic blood pressure 72 ± 7 vs. 67 ± 7 mmHg, p = 0.003), PWV (6 ± 0.3 vs. 5 ± 0.6 m/s, p < 0.001) and AI (22 ± 13 vs. 15 ± 12%, p = 0.007) but lower standard deviation of normal-to-normal intervals (SDNN; 44 ± 17 vs. 54 ± 18 ms, p = 0.005) and root mean square of successive differences between normal heartbeats (RMSSD; 37 ± 17 vs. 51 ± 22 ms, p = 0.002). CONCLUSION PTSD in young women is associated with higher brachial and central pressures, increased arterial stiffness and blunted parasympathetic control of the heart. These findings illustrate potential mechanisms underlying high risk for CVD in young women with PTSD, suggesting possible treatment targets for this at-risk group.
Collapse
Affiliation(s)
- Zynab Ahmed
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, 420 Delaware St. SE (MMC 388), Minneapolis, MN, USA
- Division of Health Policy and Management, School of Public Health, Minneapolis, MN, USA
| | - Chowdhury Ibtida Tahmin
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, 420 Delaware St. SE (MMC 388), Minneapolis, MN, USA
| | - Chowdhury Tasnova Tahsin
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, 420 Delaware St. SE (MMC 388), Minneapolis, MN, USA
| | - Vasiliki Michopoulos
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Azhaar Mohamed
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, 420 Delaware St. SE (MMC 388), Minneapolis, MN, USA
| | - Redeat Wattero
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, 420 Delaware St. SE (MMC 388), Minneapolis, MN, USA
| | - Sophia Albott
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
- Mental Health Service Line, Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Kathryn R Cullen
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Dawn A Lowe
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, 420 Delaware St. SE (MMC 388), Minneapolis, MN, USA
| | - John Osborn
- Department of Surgery, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Ida T Fonkoue
- Divisions of Physical Therapy and Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota Medical School, 420 Delaware St. SE (MMC 388), Minneapolis, MN, USA.
| |
Collapse
|
6
|
Shawky HA, Abdel Hafez SMN, Hasan NAK, Elbassuoni E, Abdelbaky FAF, AboBakr AHS. Changes in Rat Adrenal Cortex and Pineal Gland in Inverted Light-Dark Cycle: A Biochemical, Histological, and Immunohistochemical Study. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2023; 29:2037-2052. [PMID: 37738357 DOI: 10.1093/micmic/ozad101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 08/13/2023] [Accepted: 08/25/2023] [Indexed: 09/24/2023]
Abstract
Poor sleep standards are common in everyday life; it is frequently linked to a rise in stress levels. The adrenal gland interacts physiologically with the pineal gland in the stress response. Pineal gland is a small endocrine organ that modulates sleep patterns. This work aimed to evaluate the inverted light-dark cycle rhythm on the histological changes within the adrenal cortex and pineal gland in adult male albino rats. Twenty adult male albino rats were equally divided into two groups: For the first control group, animals were kept on daylight-darkness for 12-12 h. The second group was kept under an inverted 12- to 12-h light-darkness cycle for 4 weeks. Adrenal sections were subjected to biochemical, histological, and immunohistochemical study. Inverted light-dark cycle group recorded a significant elevation of plasma corticosterone, tissue malondialdehyde, tumor necrosis factor-α, and interleukin-1β (IL-1β) associated with a significant reduction of catalase and superoxide dismutase. Adrenal cortex showed biochemical and histological changes. Pineal glands also showed loss of lobular architecture. A significant upregulation in activated inducible nitric oxide synthase (iNOS) and B-cell lymphoma-associated X (Bax) immunohistochemical expression was recorded in adrenal cortex associating with downregulation in B-cell lymphoma 2 (Bcl-2). It could be concluded that subchronic inverted light-dark cycle exerted direct effects on adrenal cortex and the pineal glands.
Collapse
Affiliation(s)
- Heba A Shawky
- Anatomy and Embryology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Sara Mohamed Naguib Abdel Hafez
- Histology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Nabil Abdel Kader Hasan
- Anatomy and Embryology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Eman Elbassuoni
- Physiology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Fatma Alzhraa Fouad Abdelbaky
- Anatomy and Embryology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| | - Abdel Hamid Sayed AboBakr
- Anatomy and Embryology Department, Faculty of Medicine, Minia University, Minia Governorate, Minia City, Cairo-Aswan Agricultural Road, El-Minia 61511, Egypt
| |
Collapse
|
7
|
Bekhbat M, Drake J, Reed EC, Lauten TH, Natour T, Vladimirov VI, Case AJ. Repeated social defeat stress leads to immunometabolic shifts in innate immune cells of the spleen. Brain Behav Immun Health 2023; 34:100690. [PMID: 37791319 PMCID: PMC10543777 DOI: 10.1016/j.bbih.2023.100690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/05/2023] Open
Abstract
Psychosocial stress has been shown to prime peripheral innate immune cells, which take on hyper-inflammatory phenotypes and are implicated in depressive-like behavior in mouse models. However, the impact of stress on cellular metabolic states that are thought to fuel inflammatory phenotypes in immune cells are unknown. Using single cell RNA-sequencing, we investigated mRNA enrichment of immunometabolic pathways in innate immune cells of the spleen in mice subjected to repeated social defeat stress (RSDS) or no stress (NS). RSDS mice displayed a significant increase in the number of splenic macrophages and granulocytes (p < 0.05) compared to NS littermates. RSDS-upregulated genes in macrophages, monocytes, and granulocytes significantly enriched immunometabolic pathways thought to play a role in myeloid-driven inflammation (glycolysis, HIF-1 signaling, MTORC1 signaling) as well as pathways related to oxidative phosphorylation (OXPHOS) and oxidative stress (p < 0.05 and FDR<0.1). These results suggest that the metabolic enhancement reflected by upregulation of glycolytic and OXPHOS pathways may be important for cellular proliferation of splenic macrophages and granulocytes following repeated stress exposure. A better understanding of these intracellular metabolic mechanisms may ultimately help develop novel strategies to reverse the impact of stress and associated peripheral immune changes on the brain and behavior.
Collapse
Affiliation(s)
- Mandakh Bekhbat
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, 30322, USA
| | - John Drake
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
| | - Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Medical Physiology, Texas A&M University, Bryan, TX, 77807, USA
| | - Tatlock H. Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Medical Physiology, Texas A&M University, Bryan, TX, 77807, USA
| | - Tamara Natour
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Medical Physiology, Texas A&M University, Bryan, TX, 77807, USA
| | - Vladimir I. Vladimirov
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Psychiatry, University of Arizona, Phoenix, AZ, 85004, USA
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, 77807, USA
- Department of Medical Physiology, Texas A&M University, Bryan, TX, 77807, USA
| |
Collapse
|
8
|
Moshfegh CM, Elkhatib SK, Watson GF, Drake J, Taylor ZN, Reed EC, Lauten TH, Clopp AJ, Vladimirov VI, Case AJ. S100a9 Protects Against the Effects of Repeated Social Defeat Stress. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:919-929. [PMID: 37881565 PMCID: PMC10593888 DOI: 10.1016/j.bpsgos.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/07/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Background Posttraumatic stress disorder, a consequence of psychological trauma, is associated with increased inflammation and an elevated risk of developing comorbid inflammatory diseases. However, the mechanistic link between this mental health disorder and inflammation remains elusive. We previously found that S100a8 and S100a9 messenger RNA, genes that encode the protein calprotectin, were significantly upregulated in T lymphocytes and positively correlated with inflammatory gene expression and the mitochondrial redox environment in these cells. Therefore, we hypothesized that genetic deletion of calprotectin would attenuate the inflammatory and redox phenotype displayed after psychological trauma. Methods We used a preclinical mouse model of posttraumatic stress disorder known as repeated social defeat stress (RSDS) combined with pharmacological and genetic manipulation of S100a9 (which functionally eliminates calprotectin). A total of 186 animals (93 control, 93 RSDS) were used in these studies. Results Unexpectedly, we observed worsening of behavioral pathology, inflammation, and the mitochondrial redox environment in mice after RSDS compared with wild-type animals. Furthermore, loss of calprotectin significantly enhanced the metabolic demand on T lymphocytes, suggesting that this protein may play an undescribed role in mitochondrial regulation. This was further supported by single-cell RNA sequencing analysis demonstrating that RSDS and loss of S100a9 primarily altered genes associated with mitochondrial function and oxidative phosphorylation. Conclusions These data demonstrate that the loss of calprotectin potentiates the RSDS-induced phenotype, which suggests that its observed upregulation after psychological trauma may provide previously unexplored protective functions.
Collapse
Affiliation(s)
- Cassandra M. Moshfegh
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Safwan K. Elkhatib
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Gabrielle F. Watson
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - John Drake
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
| | - Zachary N. Taylor
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
| | - Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Medical Physiology, Texas A&M University, Bryan, Texas
| | - Tatlock H. Lauten
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Medical Physiology, Texas A&M University, Bryan, Texas
| | - Amelia J. Clopp
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Medical Physiology, Texas A&M University, Bryan, Texas
| | - Vladimir I. Vladimirov
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Psychiatry, University of Arizona, Phoenix, Arizona
- Lieber Institute for Brain Development, Johns Hopkins University, Baltimore, Maryland
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, Texas
- Department of Medical Physiology, Texas A&M University, Bryan, Texas
| |
Collapse
|
9
|
Levesque P, Desmeules C, Béchard L, Huot-Lavoie M, Demers MF, Roy MA, Deslauriers J. Sex-specific immune mechanisms in PTSD symptomatology and risk: A translational overview and perspectives. Brain Res Bull 2023; 195:120-129. [PMID: 36822271 DOI: 10.1016/j.brainresbull.2023.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 02/23/2023]
Abstract
Altered immune function in patients with posttraumatic stress disorder (PTSD) may play a role in the disorder pathophysiology and onset. Women are more likely to develop PTSD, suggesting potential sex-specific inflammatory mechanisms underlying the dichotomous prevalence and risk of PTSD in men and women. In this review we examine the available literature to better assess the state of knowledge in the field. In humans, increased systemic inflammation is found in both men and women with PTSD, but seems to be at a greater extend in women. Despite the existence of few clinical studies taking account of sex as a factor in the observed immune changes in PTSD, challenges in the study of sex-specific immune function in humans include: controlling for confounding variates such as the type of trauma and the ethnicity; and limited methodologies available to study central nervous system (CNS)-relevant changes. Thus, preclinical studies are a valuable tool to provide us with key insights on sex-specific peripheral and CNS immune mechanisms underlying PTSD. Available preclinical studies reported increased systemic and CNS inflammation, as well as elevated trafficking of monocytes from the periphery to the brain in both male and female rodents. To date, psychological trauma-induced inflammation is more robust in female vs male rodents. Limitations of preclinical studies include animal models hardly applicable to female rodents, and hormonal changes across estrus phases that may affect immune function. The present review: (1) highlights the key findings from both human and animal studies, (2) provides guidance to address limitations; and (3) discusses the gap of knowledge on the complex intertwined interaction between the brain, neurovascular, and systemic units.
Collapse
Affiliation(s)
- Pascal Levesque
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada
| | - Charles Desmeules
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC G1V 4G2, Canada; Centre de recherche CERVO, Québec, QC G1E 1T2, Canada; Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Laurent Béchard
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada; Centre de recherche CERVO, Québec, QC G1E 1T2, Canada; Institut universitaire en santé mentale de Québec, CIUSSS-CN, Québec, QC G1J 2G3, Canada
| | - Maxime Huot-Lavoie
- Centre de recherche CERVO, Québec, QC G1E 1T2, Canada; Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada; Institut universitaire en santé mentale de Québec, CIUSSS-CN, Québec, QC G1J 2G3, Canada
| | - Marie-France Demers
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada; Centre de recherche CERVO, Québec, QC G1E 1T2, Canada; Institut universitaire en santé mentale de Québec, CIUSSS-CN, Québec, QC G1J 2G3, Canada
| | - Marc-André Roy
- Centre de recherche CERVO, Québec, QC G1E 1T2, Canada; Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada; Institut universitaire en santé mentale de Québec, CIUSSS-CN, Québec, QC G1J 2G3, Canada
| | - Jessica Deslauriers
- Centre de Recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Québec, QC G1V 4G2, Canada; Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
10
|
Reed EC, Case AJ. Defining the nuanced nature of redox biology in post-traumatic stress disorder. Front Physiol 2023; 14:1130861. [PMID: 37007993 PMCID: PMC10060537 DOI: 10.3389/fphys.2023.1130861] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a mental health disorder that arises after experiencing or witnessing a traumatic event. Despite affecting around 7% of the population, there are currently no definitive biological signatures or biomarkers used in the diagnosis of PTSD. Thus, the search for clinically relevant and reproducible biomarkers has been a major focus of the field. With significant advances of large-scale multi-omic studies that include genomic, proteomic, and metabolomic data, promising findings have been made, but the field still has fallen short. Amongst the possible biomarkers examined, one area is often overlooked, understudied, or inappropriately investigated: the field of redox biology. Redox molecules are free radical and/or reactive species that are generated as a consequence of the necessity of electron movement for life. These reactive molecules, too, are essential for life, but in excess are denoted as "oxidative stress" and often associated with many diseases. The few studies that have examined redox biology parameters have often utilized outdated and nonspecific methods, as well as have reported confounding results, which has made it difficult to conclude the role for redox in PTSD. Herein, we provide a foundation of how redox biology may underlie diseases like PTSD, critically examine redox studies of PTSD, and provide future directions the field can implement to enhance standardization, reproducibility, and accuracy of redox assessments for the use of diagnosis, prognosis, and therapy of this debilitating mental health disorder.
Collapse
Affiliation(s)
- Emily C. Reed
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| | - Adam J. Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M University, Bryan, TX, United States
- Department of Medical Physiology, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
11
|
McMurray KMJ, Sah R. Neuroimmune mechanisms in fear and panic pathophysiology. Front Psychiatry 2022; 13:1015349. [PMID: 36523875 PMCID: PMC9745203 DOI: 10.3389/fpsyt.2022.1015349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/02/2022] [Indexed: 12/02/2022] Open
Abstract
Panic disorder (PD) is unique among anxiety disorders in that the emotional symptoms (e.g., fear and anxiety) associated with panic are strongly linked to body sensations indicative of threats to physiological homeostasis. For example, panic attacks often present with feelings of suffocation that evoke hyperventilation, breathlessness, or air hunger. Due to the somatic underpinnings of PD, a major focus has been placed on interoceptive signaling and it is recognized that dysfunctional body-to-brain communication pathways promote the initiation and maintenance of PD symptomatology. While body-to-brain signaling can occur via several pathways, immune and humoral pathways play an important role in communicating bodily physiological state to the brain. Accumulating evidence suggests that neuroimmune mediators play a role in fear and panic-associated disorders, although this has not been systematically investigated. Currently, our understanding of the role of immune mechanisms in the etiology and maintenance of PD remains limited. In the current review, we attempt to summarize findings that support a role of immune dysregulation in PD symptomology. We compile evidence from human studies and panic-relevant rodent paradigms that indicate a role of systemic and brain immune signaling in the regulation of fear and panic-relevant behavior and physiology. Specifically, we discuss how immune signaling can contribute to maladaptive body-to-brain communication and conditioned fear that are relevant to spontaneous and conditioned symptoms of PD and identify putative avenues warranting future investigation.
Collapse
Affiliation(s)
- Katherine M. J. McMurray
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- Veterans Affairs Medical Center, Cincinnati, OH, United States
| | - Renu Sah
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
- Veterans Affairs Medical Center, Cincinnati, OH, United States
| |
Collapse
|
12
|
Katrinli S, Oliveira NCS, Felger JC, Michopoulos V, Smith AK. The role of the immune system in posttraumatic stress disorder. Transl Psychiatry 2022; 12:313. [PMID: 35927237 PMCID: PMC9352784 DOI: 10.1038/s41398-022-02094-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/14/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) develops in a subset of individuals upon exposure to traumatic stress. In addition to well-defined psychological and behavioral symptoms, some individuals with PTSD also exhibit elevated concentrations of inflammatory markers, including C-reactive protein, interleukin-6, and tumor necrosis factor-α. Moreover, PTSD is often co-morbid with immune-related conditions, such as cardiometabolic and autoimmune disorders. Numerous factors, including lifetime trauma burden, biological sex, genetic background, metabolic conditions, and gut microbiota, may contribute to inflammation in PTSD. Importantly, inflammation can influence neural circuits and neurotransmitter signaling in regions of the brain relevant to fear, anxiety, and emotion regulation. Given the link between PTSD and the immune system, current studies are underway to evaluate the efficacy of anti-inflammatory treatments in those with PTSD. Understanding the complex interactions between PTSD and the immune system is essential for future discovery of diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Seyma Katrinli
- Department of Gynecology and Obstetrics, Emory University, Atlanta, GA, USA.
| | - Nayara C. S. Oliveira
- grid.189967.80000 0001 0941 6502Department of Gynecology and Obstetrics, Emory University, Atlanta, GA USA ,National Institute of Woman, Child, and Adolescence Health Fernandes Figueira, Rio de Janeiro, RJ Brazil ,grid.418068.30000 0001 0723 0931Department of Violence and Health Studies Jorge Careli, National School of Public Health, Fiocruz, Rio de Janeiro, RJ Brazil
| | - Jennifer C. Felger
- grid.189967.80000 0001 0941 6502Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502The Winship Cancer Institute, Emory University, Atlanta, GA USA
| | - Vasiliki Michopoulos
- grid.189967.80000 0001 0941 6502Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA
| | - Alicia K. Smith
- grid.189967.80000 0001 0941 6502Department of Gynecology and Obstetrics, Emory University, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA
| |
Collapse
|
13
|
Elkhatib SK, Moshfegh CM, Watson GF, Case AJ. T-lymphocyte tyrosine hydroxylase regulates T H17 T-lymphocytes during repeated social defeat stress. Brain Behav Immun 2022; 104:18-28. [PMID: 35580792 PMCID: PMC9659669 DOI: 10.1016/j.bbi.2022.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is a debilitating psychiatric disorder which results in deleterious changes to psychological and physical health. Patients with PTSD are especially susceptible to life-threatening co-morbid inflammation-driven pathologies, such as autoimmunity, while also demonstrating increased T-helper 17 (TH17) lymphocyte-driven inflammation. While the exact mechanism of this increased inflammation is unknown, overactivity of the sympathetic nervous system is a hallmark of PTSD. Neurotransmitters of the sympathetic nervous system (i.e., catecholamines) can alter T-lymphocyte function, which we have previously demonstrated to be partially mitochondrial redox-mediated. Furthermore, we have previously elucidated that T-lymphocytes generate their own catecholamines, and strong associations exist between tyrosine hydroxylase (TH; the rate-limiting enzyme in the synthesis of catecholamines) and pro-inflammatory interleukin 17A (IL-17A) expression within purified T-lymphocytes in a rodent model of psychological trauma. Therefore, we hypothesized that T-lymphocyte-generated catecholamines drive TH17 T-lymphocyte polarization through a mitochondrial superoxide-dependent mechanism during psychological trauma. To test this, T-lymphocyte-specific TH knockout mice (THT-KO) were subjected to psychological trauma utilizing repeated social defeat stress (RSDS). RSDS characteristically increased tumor necrosis factor-α (TNFα), IL-6, IL-17A, and IL-22, however, IL-17A and IL-22 (TH17 produced cytokines) were selectively attenuated in circulation and in T-lymphocytes of THT-KO animals. When activated ex vivo, secretion of IL-17A and IL-22 by THT-KO T-lymphocytes was also found to be reduced, but could be partially rescued with supplementation of norepinephrine specifically. Interestingly, THT-KO T-lymphocytes were still able to polarize to TH17 under exogenous polarizing conditions. Last, contrary to our hypothesis, we found RSDS-exposed THT-KO T-lymphocytes still displayed elevated mitochondrial superoxide, suggesting increased mitochondrial superoxide is upstream of T-lymphocyte TH induction, activity, and TH17 regulation. Overall, these data demonstrate TH in T-lymphocytes plays a critical role in RSDS-induced TH17 T-lymphocytes and offer a previously undescribed regulator of inflammation in RSDS.
Collapse
Affiliation(s)
- Safwan K Elkhatib
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Cassandra M Moshfegh
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Gabrielle F Watson
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Adam J Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M Health Science Center, College Station, TX, United States; Department of Medical Physiology, Texas A&M Health Science Center, College Station, TX, United States.
| |
Collapse
|
14
|
Grotle AK, Darling AM, Saunders EF, Fadel PJ, Trott DW, Greaney JL. Augmented T-cell mitochondrial reactive oxygen species in adults with major depressive disorder. Am J Physiol Heart Circ Physiol 2022; 322:H568-H574. [PMID: 35179977 PMCID: PMC8917910 DOI: 10.1152/ajpheart.00019.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 11/22/2022]
Abstract
The prevalence of major depressive disorder (MDD) is highest in young adulthood, an effect that has been magnified by the COVID-19 pandemic. Importantly, individuals with MDD are at a greater risk of developing cardiovascular disease (CVD). Accumulating evidence supports immune system dysregulation as a major contributor to the elevated CVD risk in older adults with MDD; however, whether this is present in young adults with MDD without comorbid disease remains unclear. Interestingly, recent data suggest augmented T-cell mitochondrial reactive oxygen species (T-cell mitoROS) as a potent driver of immune dysregulation in animal models of psychiatric disease. With this background in mind, we tested the hypothesis that young adults with MDD would have augmented T-cell mitoROS and circulating proinflammatory cytokines compared with healthy young adults without MDD (HA). Whole blood was drawn from 14 young adults with MDD (age: 23 ± 2 yr) and 11 HA (age: 22 ± 1 yr). T-cell mitoROS (MitoSOX red; total: CD3+, T-helper: CD4+, T cytotoxic: CD8+) and serum cytokines were assessed by flow cytometry. Total T-cell mitoROS was significantly greater in adults with MDD compared with HA [median: 14,089 arbitrary units (AU); median: 1,362 AU, P = 0.01]. Likewise, both T-helper and T-cytotoxic cell mitoROS were significantly greater in adults with MDD compared with HA (both: P < 0.05). There were no differences in circulating cytokines between groups (all cytokines: P > 0.05). Collectively, these findings suggest that elevated T-cell mitoROS may represent an early marker of immune system dysregulation in young, otherwise healthy, adults with MDD.NEW & NOTEWORTHY To our knowledge, we provide the first evidence of augmented T-cell mitochondrial reactive oxygen species (T-cell mitoROS) in young, otherwise healthy adults with MDD. Although the elevated T-cell mitoROS did not correspond to a proinflammatory profile, these findings suggest that elevated T-cell mitoROS may be an early marker of immune system dysregulation in young adults with MDD.
Collapse
Affiliation(s)
- Ann-Katrin Grotle
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - Ashley M Darling
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - Erika F Saunders
- Department of Psychiatry and Behavioral Health, Penn State College of Medicine, and Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Paul J Fadel
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - Daniel W Trott
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - Jody L Greaney
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
15
|
Interleukine-17 Modulates Neurogenesis and Behavior Following Exposure to Trauma in Mice. Cells 2022; 11:cells11030343. [PMID: 35159158 PMCID: PMC8834196 DOI: 10.3390/cells11030343] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric disorder accompanied by deficits in cognitive and social skills. Adult hippocampal neurogenesis is a lifelong phenomenon, with new neurons being formed in the granular cell layer of the dentate gyrus. Impaired neurogenesis is associated with multiple behavioral disorders including Alzheimer's disease and schizophrenia. PTSD patients often present hippocampal atrophy and animal models clearly present impaired neurogenesis. Previous studies on PTSD patients demonstrated elevated levels of Th17 cells and plasma levels of the pro-inflammatory cytokine interleukin-17A (IL-17A). Since IL-17A can impair neurogenesis in mice, we thus hypothesized that decreasing the serum levels of IL-17A will increase hippocampal neurogenesis and alleviate symptoms in a murine model of PTSD. Surprisingly, our results showed that attempting to neutralize IL-17A with an antibody resulted in increased serum levels of IL-17A, while targeting IL-23, the upstream regulator of IL-17, did lower the levels of IL-17A in trauma-exposed mice. As expected, increased levels of serum IL-17A (in anti-IL-17A treated mice) resulted in impaired neurogenesis, reflected by reduced number of proliferating Ki67+ neural progenitors and newly formed DCX+ neurons, which was correlated with increased expression of Hes1. Nevertheless, increased maturation was noted by the expression of Slit2 and Ache. In contrast, treatment with anti-IL-23 indeed resulted in increased neurogenesis. Behaviorally, both treatments did not affect trauma-related freezing behavior but did affect trauma-related social deficits. Unexpectedly, increased levels of serum IL-17A (in anti-IL-17A treated mice) prevented social deficits in trauma-exposed mice while anti-IL-23 exacerbated these deficits. We thus conclude that IL-17 is involved in regulating neurogenesis following exposure to stress but may be important in maintaining social behavior.
Collapse
|
16
|
Johnson E, J M, I L, R S. Asthma and posttraumatic stress disorder (PTSD): Emerging links, potential models and mechanisms. Brain Behav Immun 2021; 97:275-285. [PMID: 34107349 PMCID: PMC8453093 DOI: 10.1016/j.bbi.2021.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/16/2021] [Accepted: 06/04/2021] [Indexed: 12/31/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is a highly prevalent, debilitating mental health condition. A better understanding of contributory neurobiological mechanisms will lead to effective treatments, improving quality of life for patients. Given that not all trauma-exposed individuals develop PTSD, identification of pre-trauma susceptibility factors that can modulate posttraumatic outcomes is important. Recent clinical evidence supports a strong link between inflammatory conditions and PTSD. A particularly strong association has been reported between asthma and PTSD prevalence and severity. Unlike many other PTSD-comorbid inflammatory conditions, asthma often develops in children, sensitizing them to subsequent posttraumatic pathology throughout their lifetime. Currently, there is a significant need to understand the neurobiology, shared mechanisms, and inflammatory mediators that may contribute to comorbid asthma and PTSD. Here, we provide a translational perspective of asthma and PTSD risk and comorbidity, focusing on clinical associations, relevant rodent paradigms and potential mechanisms that may translate asthma-associated inflammation to PTSD development.
Collapse
Affiliation(s)
- Emily Johnson
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati OH, 45220,Neuroscience Graduate Program, University of Cincinnati, Cincinnati OH, 45220
| | - McAlees J
- Division of Immunobiology, Children’s Hospital Medical Center, Cincinnati OH, 45220
| | - Lewkowich I
- Division of Immunobiology, Children’s Hospital Medical Center, Cincinnati OH, 45220,Department of Pediatrics, University of Cincinnati, Cincinnati OH, 45220
| | - Sah R
- Dept. of Pharmacology & Systems Physiology, University of Cincinnati, Cincinnati OH, 45220,Neuroscience Graduate Program, University of Cincinnati, Cincinnati OH, 45220,VA Medical Center, Cincinnati, OH, 45220
| |
Collapse
|
17
|
Sakamoto S, Zhu X, Hasegawa Y, Karma S, Obayashi M, Alway E, Kamiya A. Inflamed brain: Targeting immune changes and inflammation for treatment of depression. Psychiatry Clin Neurosci 2021; 75:304-311. [PMID: 34227186 PMCID: PMC8683253 DOI: 10.1111/pcn.13286] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/22/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022]
Abstract
Although there are a number of clinically effective treatments for depression, many patients exhibit treatment resistance. Recent clinical and preclinical studies reveal that peripheral and brain immune changes and inflammation are involved in the pathophysiology of depression. This 'Inflamed Brain' research provides critical clues for understanding of disease pathophysiology and many candidate molecules that are potentially useful for identifying novel drug targets for the treatment of depression. In this review, we will present clinical evidence on the role of inflammation in the pathophysiology of depression. We will also summarize current clinical trials which test drugs targeting inflammation for the treatment of patients with depression. Furthermore, we will briefly provide preclinical evidence demonstrating altered immune system function and inflammation in stress-induced animal models and will discuss the future potential of inflammation-related drug targets. Collectively, inflammatory signatures identified in clinical and preclinical studies may allow us to stratify depressive patients based on biotypes, contributing to the development of novel mechanism-based interventions that target specific patient populations.
Collapse
Affiliation(s)
- Shinji Sakamoto
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaolei Zhu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yuto Hasegawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sadik Karma
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mizuho Obayashi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emily Alway
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atsushi Kamiya
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
18
|
Kim E, Zhao Z, Rzasa JR, Glassman M, Bentley WE, Chen S, Kelly DL, Payne GF. Association of acute psychosocial stress with oxidative stress: Evidence from serum analysis. Redox Biol 2021; 47:102138. [PMID: 34555595 PMCID: PMC8458980 DOI: 10.1016/j.redox.2021.102138] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 02/08/2023] Open
Abstract
Growing evidence implicates an association between psychosocial stress and oxidative stress (OxSt) although there are not yet reliable biomarkers to study this association. We used a Trier Social Stress Test (TSST) and compared the response of a healthy control group (HC; N=10) against the response of a schizophrenia group (SCZ; N=10) that is expected to have higher levels of OxSt. Because our previous study showed inconsistent changes in conventional molecular markers for stress responses in the neuroendocrine and immune systems, we analyzed the same serum samples using a separate reducing capacity assay that provides a more global measurement of OxSt. This assay uses the moderately strong oxidizing agent iridium (Ir) to probe a sample's reducing capacity. Specifically, we characterized OxSt by this Ir-reducing capacity assay (Ir-RCA) using two measurement modalities (optical and electrochemical) and we tuned this assay by imposing an input voltage sequence that generates multiple output metrics for data-driven analysis. We defined five OxSt metrics (one optical and four electrochemical metrics) and showed: (i) internal consistency among each metric in the measurements of all 40 samples (baseline and post TSST for N=20); (ii) all five metrics were consistent with expectations of higher levels of OxSt for the SCZ group (three individual metrics showed statistically significant differences); and (iii) all five metrics showed higher levels of OxSt Post-TSST (one metric showed statistically significant difference). Using multivariant analysis, we showed that combinations of OxSt metrics could discern statistically significant increases in OxSt for both the SCZ and HC groups 90 min after the imposed acute psychosocial stress. Ir-reducing capacity assay (Ir-RCA) provides a robust global measure of oxidative stress in serum. The multiple oxidative stress (OxSt) output metrics of this Ir-RCA are useful for data-driven analysis. The combination of OxSt metrics can discern significant increases in OxStwithin 90 mins of an imposed psychosocial stress.
Collapse
Affiliation(s)
- Eunkyoung Kim
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD, 20742, USA; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA
| | - Zhiling Zhao
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD, 20742, USA; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA
| | - John Robertson Rzasa
- Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA
| | - Matthew Glassman
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, 21228, USA
| | - William E Bentley
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD, 20742, USA; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA
| | - Shuo Chen
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, 21228, USA
| | - Deanna L Kelly
- Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, MD, 21228, USA
| | - Gregory F Payne
- Institute for Bioscience & Biotechnology Research, University of Maryland, College Park, MD, 20742, USA; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
19
|
Elkhatib SK, Moshfegh CM, Watson GF, Schwab AD, Katsurada K, Patel KP, Case AJ. Splenic denervation attenuates repeated social defeat stress-induced T-lymphocyte inflammation. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 1:190-200. [PMID: 35330608 PMCID: PMC8941638 DOI: 10.1016/j.bpsgos.2021.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/12/2021] [Accepted: 05/06/2021] [Indexed: 11/28/2022] Open
Abstract
Background Post-traumatic stress disorder (PTSD) is a devastating psychological disorder. Patients with PTSD canonically demonstrate an increased risk for inflammatory diseases, as well as increased sympathetic tone and norepinephrine (NE) outflow. Yet, the exact etiology and causal nature of these physiologic changes remain unclear. Previously, we demonstrated that exogenous NE alters mitochondrial superoxide in T-lymphocytes to produce a pro-inflammatory T-helper 17 (TH17) phenotype, and observed similar TH17 polarization in a preclinical model of PTSD. Therefore, we hypothesized sympathetic-driven neuroimmune interactions could mediate psychological trauma-induced T-lymphocyte inflammation. Methods Repeated social defeat stress (RSDS) is a preclinical murine model that recapitulates the behavioral, autonomic, and inflammatory aspects of PTSD. Targeted splenic denervation (Dnx) was performed to deduce the contribution of splenic sympathetic nerves to RSDS-induced inflammation. Eighty-five C57BL/6J mice underwent Dnx or sham-operation, followed by RSDS or control paradigms. Animals were assessed for behavioral, autonomic, inflammatory, and redox profiles. Results Dnx did not alter the antisocial or anxiety-like behavior induced by RSDS. In circulation, RSDS Dnx animals exhibited diminished levels of T-lymphocyte-specific cytokines (IL-2, IL-17A, and IL-22) compared to intact animals, whereas other non-specific inflammatory cytokines (e.g., IL-6, TNF-α, and IL-10) were unaffected by Dnx. Importantly, Dnx specifically ameliorated the increases in RSDS-induced T-lymphocyte mitochondrial superoxide, TH17 polarization, and pro-inflammatory gene expression with minimal impact to non-T-lymphocyte immune populations. Conclusions Overall, our data suggest that sympathetic nerves regulate RSDS-induced splenic T-lymphocyte inflammation, but play less of a role in the behavioral and non-T-lymphocyte inflammatory phenotypes induced by this psychological trauma paradigm.
Collapse
Affiliation(s)
- Safwan K. Elkhatib
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Cassandra M. Moshfegh
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Gabrielle F. Watson
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Aaron D. Schwab
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kenichi Katsurada
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- School of Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Adam J. Case
- Department of Cellular and Integrative Physiology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
- Department of Psychiatry and Behavioral Sciences, College of Medicine, Texas A&M Health Science Center, Bryan, Texas
| |
Collapse
|
20
|
Weggen JB, Darling AM, Autler AS, Hogwood AC, Decker KP, Imthurn B, Tuzzolo GM, Garten RS. Impact of acute antioxidant supplementation on vascular function and autonomic nervous system modulation in young adults with PTSD. Am J Physiol Regul Integr Comp Physiol 2021; 321:R49-R61. [PMID: 34075811 DOI: 10.1152/ajpregu.00054.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Posttraumatic stress disorder (PTSD) has been associated with an increase in risk of cardiovascular disease (CVD). The goal of this study was to determine if peripheral vascular dysfunction, a precursor to CVD, was present in young adults with PTSD, and if an acute antioxidant (AO) supplementation could modify this potential PTSD-induced vascular dysfunction. Thirteen individuals with PTSD were recruited for this investigation and were compared with 35 age- and sex-matched controls (CTRL). The PTSD group participated in two visits, consuming either a placebo (PTSD-PL) or antioxidants (PTSD-AO; vitamins C and E; α-lipoic acid) before their visits, whereas the CTRL subjects only participated in one visit. Upper and lower limb vascular functions were assessed via flow-mediated dilation and passive leg movement technique. Heart rate variability was utilized to assess autonomic nervous system modulation. The PTSD-PL condition, when compared with the CTRL group, reported lower arm and leg microvascular function as well as sympathetic nervous system (SNS) predominance. After acute AO supplementation, arm, but not leg, microvascular function was improved and SNS predominance was lowered to which the prior difference between PTSD group and CTRL was no longer significant. Young individuals with PTSD demonstrated lower arm and leg microvascular function as well as greater SNS predominance when compared with age- and sex-matched controls. Furthermore, this lower vascular/autonomic function was augmented by an acute AO supplementation to the level of the healthy controls, potentially implicating oxidative stress as a contributor to this blunted vascular/autonomic function.
Collapse
Affiliation(s)
- Jennifer B Weggen
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Ashley M Darling
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - Aaron S Autler
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Austin C Hogwood
- Department of Kinesiology, University of Virginia, Charlottesville, Virginia
| | - Kevin P Decker
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Brandon Imthurn
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Gina M Tuzzolo
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Ryan S Garten
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
21
|
Moshfegh CM, Case AJ. The Redox-Metabolic Couple of T Lymphocytes: Potential Consequences for Hypertension. Antioxid Redox Signal 2021; 34:915-935. [PMID: 32237890 PMCID: PMC8035925 DOI: 10.1089/ars.2020.8042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/17/2020] [Accepted: 03/18/2020] [Indexed: 12/25/2022]
Abstract
Significance: T lymphocytes, as part of the adaptive immune system, possess the ability to activate and function in extreme cellular microenvironments, which requires these cells to remain highly malleable. One mechanism in which T lymphocytes achieve this adaptability is by responding to cues from both reactive oxygen and nitrogen species, as well as metabolic flux, which together fine-tune the functional fate of these adaptive immune cells. Recent Advances: To date, examinations of the redox and metabolic effects on T lymphocytes have primarily investigated these biological processes as separate entities. Given that the redox and metabolic environments possess significant overlaps of pathways and molecular species, it is inevitable that perturbations in one environment affect the other. Recent consideration of this redox-metabolic couple has demonstrated the strong link and regulatory consequences of these two systems in T lymphocytes. Critical Issues: The redox and metabolic control of T lymphocytes is essential to prevent dysregulated inflammation, which has been observed in cardiovascular diseases such as hypertension. The role of the adaptive immune system in hypertension has been extensively investigated, but the understanding of how the redox and metabolic environments control T lymphocytes in this disease remains unclear. Future Directions: Herein, we provide a discussion of the redox and metabolic control of T lymphocytes as separate entities, as well as coupled to one another, to regulate adaptive immunity. While investigations examining this pair together in T lymphocytes are sparse, we speculate that T lymphocyte destiny is shaped by the redox-metabolic couple. In contrast, disrupting this duo may have inflammatory consequences such as hypertension.
Collapse
Affiliation(s)
- Cassandra M. Moshfegh
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Adam J. Case
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
22
|
Elkhatib SK, Moshfegh CM, Watson GF, Case AJ. Peripheral inflammation is strongly linked to elevated zero maze behavior in repeated social defeat stress. Brain Behav Immun 2020; 90:279-285. [PMID: 32890698 PMCID: PMC7568442 DOI: 10.1016/j.bbi.2020.08.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 12/25/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a psychiatric illness that results in an increased risk for a variety of inflammatory diseases. The exact etiology of this increased risk is unknown, and thus several animal models have been developed to investigate the neuroimmune interactions of PTSD. Repeated social defeat stress (RSDS) is an established preclinical model of psychological trauma that recapitulates certain behavioral and inflammatory aspects of human PTSD. Furthermore, RSDS has been utilized to subgroup animals into susceptible and resilient populations based on one specific behavioral phenotype (i.e., social interaction). Herein, we conducted an extensive investigation of circulating inflammatory proteins after RSDS and found significant elevations in various cytokines and chemokines after exposure to RSDS. When categorizing animals into either susceptible or resilient populations based on social interaction, we found no inflammatory or other behavioral differences between these subgroups. Furthermore, correlative analyses found no significant correlation between social interaction parameters and inflammation. In contrast, parameters from the elevated zero maze (EZM) demonstrated significant associations and clustering to five circulating cytokines. When animals were subdivided into susceptible and resilient populations solely based upon combined EZM performance, significant inflammatory differences were evident between these groups. Strikingly, these circulating inflammatory proteins displayed a stronger predictive ability of EZM performance compared to social interaction test performance. These findings provide new insights into inflammatory markers associated with RSDS, and the utility of EZM to effectively group RSDS-exposed mice into populations with differential levels of peripheral inflammation.
Collapse
Affiliation(s)
- Safwan K Elkhatib
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Cassandra M Moshfegh
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Gabrielle F Watson
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Adam J Case
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States.
| |
Collapse
|
23
|
Verbitsky A, Dopfel D, Zhang N. Rodent models of post-traumatic stress disorder: behavioral assessment. Transl Psychiatry 2020; 10:132. [PMID: 32376819 PMCID: PMC7203017 DOI: 10.1038/s41398-020-0806-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 03/17/2020] [Accepted: 04/08/2020] [Indexed: 12/29/2022] Open
Abstract
Although the etiology and expression of psychiatric disorders are complex, mammals show biologically preserved behavioral and neurobiological responses to valent stimuli which underlie the use of rodent models of post-traumatic stress disorder (PTSD). PTSD is a complex phenotype that is difficult to model in rodents because it is diagnosed by patient interview and influenced by both environmental and genetic factors. However, given that PTSD results from traumatic experiences, rodent models can simulate stress induction and disorder development. By manipulating stress type, intensity, duration, and frequency, preclinical models reflect core PTSD phenotypes, measured through various behavioral assays. Paradigms precipitate the disorder by applying physical, social, and psychological stressors individually or in combination. This review discusses the methods used to trigger and evaluate PTSD-like phenotypes. It highlights studies employing each stress model and evaluates their translational efficacies against DSM-5, validity criteria, and criteria proposed by Yehuda and Antelman's commentary in 1993. This is intended to aid in paradigm selection by informing readers about rodent models, their benefits to the clinical community, challenges associated with the translational models, and opportunities for future work. To inform PTSD model validity and relevance to human psychopathology, we propose that models incorporate behavioral test batteries, individual differences, sex differences, strain and stock differences, early life stress effects, biomarkers, stringent success criteria for drug development, Research Domain Criteria, technological advances, and cross-species comparisons. We conclude that, despite the challenges, animal studies will be pivotal to advances in understanding PTSD and the neurobiology of stress.
Collapse
Affiliation(s)
- Alexander Verbitsky
- Department of Engineering Science and Mechanics, The Pennsylvania State University, University Park, PA, 16802, USA
| | - David Dopfel
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Nanyin Zhang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA, 16802, USA.
- The Huck Institutes of Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA.
| |
Collapse
|