1
|
Naseri Alavi SA, Pourasghary S, Rezakhah A, Habibi MA, Kazempour A, Mahdkhah A, Kobets A. Assessment of the Sex Hormone Profile and Its Predictive Role in Consciousness Recovery Following Severe Traumatic Brain Injury. Life (Basel) 2025; 15:359. [PMID: 40141704 PMCID: PMC11943621 DOI: 10.3390/life15030359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/15/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
INTRODUCTION Traumatic brain injuries (TBIs) are conditions affecting brain function caused by blunt or penetrating forces to the head. Symptoms may include confusion, impaired consciousness, coma, seizures, and focal or sensory neurological motor injuries. OBJECTIVE This study evaluated sex hormone profiles and their predictive role in returning consciousness after severe traumatic brain injury. MATERIALS AND METHODS We included 120 patients with TBIs and collected comprehensive information about each patient, including the cause of the trauma, age, gender, Glasgow Coma Scale (GCS) score, Injury Severity Score (ISS), and neuroradiological imaging data. The ISS was used to assess the severity of the trauma. At the same time, the lowest GCS score was recorded either before sedation and intubation in the emergency room or by emergency medical services personnel. For female participants, samples were collected during the luteal phase of the menstrual cycle (days 18 to 23). RESULTS The mean age of male patients was 33.40 years, ranging from 23 to 45 years, while female patients had an average age of 34.25 years, ranging from 25 to 48 years. The primary cause of injury for both genders was motor vehicle accidents. In male patients, testosterone levels were significantly higher in those classified as responsive (RC) compared to those non-responsive (NRC), with levels of 2.56 ± 0.47 ng/mL versus 0.81 ± 0.41 ng/mL (p = 0.003). A cut-off point of 1.885 ng/mL for testosterone levels in males was established, achieving a sensitivity and specificity of 86.7% and 86.7%, respectively. In female patients, progesterone levels were elevated in those who regained consciousness, measuring 1.80 ± 0.31 ng/mL compared to 0.62 ± 0.31 ng/mL (p = 0.012). A cut-off point of 1.335 ng/mL for progesterone levels in females was determined, with a sensitivity and specificity of 93.3% and 86.7%, respectively. CONCLUSIONS We can conclude that sex hormone levels in the acute phase of TBIs can vary between males and females. Notably, serum testosterone levels in males and progesterone levels in females with TBIs are significant prognostic factors for assessing the likelihood of regaining consciousness after such injuries. These findings underscore the importance of considering sex hormone profiles in TBI recovery prognosis.
Collapse
Affiliation(s)
| | - Sajjad Pourasghary
- Faculty of Medicine, Urmia University of Medical Sciences, Urmia 5714783734, Iran (A.R.); (A.K.); (A.M.)
| | - Amir Rezakhah
- Faculty of Medicine, Urmia University of Medical Sciences, Urmia 5714783734, Iran (A.R.); (A.K.); (A.M.)
| | - Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran 1474833163, Iran;
| | - Aydin Kazempour
- Faculty of Medicine, Urmia University of Medical Sciences, Urmia 5714783734, Iran (A.R.); (A.K.); (A.M.)
| | - Ata Mahdkhah
- Faculty of Medicine, Urmia University of Medical Sciences, Urmia 5714783734, Iran (A.R.); (A.K.); (A.M.)
| | - Andrew Kobets
- Department of Neurological Surgery, Montefiore Medical, Bronx, NY 10467, USA;
| |
Collapse
|
2
|
Barker RM, Chambers A, Kehoe PG, Rowe E, Perks CM. Untangling the role of tau in sex hormone responsive cancers: lessons learnt from Alzheimer's disease. Clin Sci (Lond) 2024; 138:1357-1369. [PMID: 39469929 PMCID: PMC11522895 DOI: 10.1042/cs20230317] [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: 05/22/2024] [Revised: 09/20/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Tubulin associated unit has been extensively studied in neurodegenerative diseases including Alzheimer's disease (AD), whereby its hyperphosphorylation and accumulation contributes to disease pathogenesis. Tau is abundantly expressed in the central nervous system but is also present in non-neuronal tissues and in tumours including sex hormone responsive cancers such as breast and prostate. Curiously, hormonal effects on tau also exist in an AD context from numerous studies on menopause, hormone replacement therapy, and androgen deprivation therapy. Despite sharing some risk factors, most importantly advancing age, there are numerous reports from population studies of, currently poorly explained inverse associations between cancer and Alzheimer's disease. We previously reviewed important components of the phosphoinositide-3-kinase/protein kinase B (PI3K/Akt) signalling pathway and their differential modulation in relation to the two diseases. Similarly, receptor tyrosine kinases, estrogen receptor and androgen receptor have all been implicated in the pathogenesis of both cancer and AD. In this review, we focus on tau and its effects in hormone responsive cancer in terms of development, progression, and treatment and in relation to sex hormones and PI3K/Akt signalling molecules including IRS-1, PTEN, Pin1, and p53.
Collapse
Affiliation(s)
- Rachel M. Barker
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Alfie Chambers
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Patrick G. Kehoe
- Department of Urology, Bristol Urological Institute, Southmead Hospital, Bristol BS10 5NB, UK
| | - Edward Rowe
- Dementia Research Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| | - Claire M. Perks
- Cancer Endocrinology Group, Learning & Research Building, Southmead Hospital, Translational Health Sciences, Bristol Medical School, Bristol BS10 5NB, UK
| |
Collapse
|
3
|
Nasirzadeh S, Hamidi GA, Banafshe HR, Tehrani MN, Shabani M, Abed A. The mutual effect of progesterone and vitamin D in an animal model of peripheral nerve injury. Res Pharm Sci 2024; 19:415-424. [PMID: 39399728 PMCID: PMC11468167 DOI: 10.4103/rps.rps_18_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/17/2023] [Accepted: 12/16/2023] [Indexed: 10/15/2024] Open
Abstract
Background and purpose Experimental and clinical studies have shown the potential role of progesterone in relieving neural injury. In addition, emerging data on vitamin D, a steroid hormone, have shown its neuroprotective properties. This study was designed to evaluate the mutual effect of vitamin D and progesterone on neuropathic pain (NP) in male rats. Experimental approach Chronic constriction injury (CCI) was induced by inserting four ligatures around the sciatic nerve. Hyperalgesia and allodynia (cold and mechanical) were considered positive behavioral scores of NP. After surgery, Sprague Dawley male rats (weighing 200-250 g) were assigned into 7 groups. Vitamin D (250 and 500 units/kg/day, i.p.) and progesterone (4 and 6 mg/kg/day, i.p.) were injected from the 1st day after CCI which continued for 21 days. Moreover, one group received the co-administration of vitamin D (500 units/kg/day, i.p.) and progesterone (6 mg/kg/day, i.p.) from the 1st day until the 21st post-CCI day. Behavioral tests were performed on the 7th, 14th, and 21st days. Findings/Results Daily supplementation with vitamin D (250 and 500 units/kg) did not alter nociception. Progesterone (4 and 6 mg/kg/day) was ineffective on thermal hyperalgesia. In the allodynia test, progesterone significantly decreased pain-related behaviors. The co-administration of vitamin D (500 units/kg/day) with progesterone (6 mg/kg/day) significantly relieved thermal hyperalgesia. Finally, the combination significantly decreased cold and mechanical allodynia. Conclusion and implications This study showed the mutual effect of progesterone and vitamin D on NP for the first time. Hyperalgesia and allodynia were significantly relieved following co-administration of vitamin D and progesterone.
Collapse
Affiliation(s)
- Sedighe Nasirzadeh
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Gholam Ali Hamidi
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Hamid Reza Banafshe
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Monireh Naderi Tehrani
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Mohammad Shabani
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Alireza Abed
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| |
Collapse
|
4
|
Liang C, Dobson AJ, Chung HF, van der Schouw YT, Sandin S, Weiderpass E, Mishra GD. Association of infertility and recurrent pregnancy loss with the risk of dementia. Eur J Epidemiol 2024; 39:785-793. [PMID: 38888679 PMCID: PMC11343804 DOI: 10.1007/s10654-024-01135-3] [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: 02/23/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
Emerging evidence has shown the association between female reproductive histories (e.g., menarche age, parity, premature and early menopause) and the risk of dementia. However, little attention has been given to infertility and pregnancy loss. To examine the associations of infertility, recurrent miscarriages, and stillbirth with the risk of dementia, this study used data from four cohorts in the International Collaboration for a Life Course Approach to Reproductive Health and Chronic Disease Events. Women with data on at least one of the reproductive exposures of interest, dementia, and all covariates were included. Histories of infertility, miscarriage, and stillbirth were self-reported. Dementia (including Alzheimer's disease) was identified through surveys, aged care, pharmaceutical, hospital, and death registry data. Cause-specific Cox regression models were used to estimate the hazard ratios of dementia, accounting for well-established risk factors of dementia, study variability, and within-study correlation. Overall, 291,055 women were included at a median (interquartile range) age of 55.0 (47.0-62.0) at baseline. During the median (interquartile range) follow-up period of 13.0 (12.0-14.0) years, 3334 (1.2%) women developed dementia. Compared to women without stillbirth, a history of recurrent stillbirths (≥ 2) was associated with 64% higher risk of dementia (adjusted hazard ratio = 1.64, 95% confidence interval: 1.46-1.85). Compared to women without miscarriage, women with recurrent miscarriages (≥ 3) were at 22% higher risk of dementia (adjusted hazard ratio = 1.22, 95% confidence interval: 1.19-1.25). These findings suggest that recurrent stillbirths is a risk factor for dementia and may need to be considered in risk assessment of dementia in women.
Collapse
Affiliation(s)
- Chen Liang
- School of Public Health, University of Queensland, Public Health Building, 288 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| | - Annette J Dobson
- School of Public Health, University of Queensland, Public Health Building, 288 Herston Road, Herston, Brisbane, QLD, 4006, Australia.
| | - Hsin-Fang Chung
- School of Public Health, University of Queensland, Public Health Building, 288 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Sven Sandin
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Gita D Mishra
- School of Public Health, University of Queensland, Public Health Building, 288 Herston Road, Herston, Brisbane, QLD, 4006, Australia
| |
Collapse
|
5
|
Almohaimeed HM, Almars AI, Alsulaimani F, Basri AM, Althobaiti NA, Albalaw AE, Alsharif I, Al Abdulmonem W, Hershan AA, Soliman MH. Investigating the potential neuroprotective benefits of taurine and Dihydrotestosterone and Hydroxyprogesterone levels in SH-SY5Y cells. Front Aging Neurosci 2024; 16:1379431. [PMID: 38867846 PMCID: PMC11168113 DOI: 10.3389/fnagi.2024.1379431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/05/2024] [Indexed: 06/14/2024] Open
Abstract
Background Taurine, an amino acid abundantly found in the brain and other tissues, has potential neuroprotective properties. Alzheimer's disease (AD) is a commonly occurring type of dementia, which becomes more prevalent as people age. This experiment aimed to assess the neuroprotective effects of taurine on SH-SY5Y cells by examining its impact on Dihydrotestosterone (DHT), Dihydroprogesterone (DHP), as well as the expression of miRNA-21 and miRNA-181. Methods The effects of various taurine concentrations (0.25, and 0.75 mg/mL), and LPS (0.1, and 12 mg/mL) on the SH-SY5Y cell line were assessed using the MTT assay. The levels of DHT and DHP were quantified using an ELISA kit. Additionally, the expression levels of miRNA-181 and miRNA-21 genes were examined through Real-Time PCR analysis. Results The results of the MTT assay showed that treatment with taurine at concentrations of 0.25, and 0.75 mg/mL reduces the toxicity of LPS in SH-SY5Y cells. ELISA results indicated that taurine at a concentration of 0.25, and 0.75 mg/mL significantly elevated DHT and DHP hormones in the SH-SY5Y cell line compared to the untreated group (p < 0.01). The expression levels of IL-1β and IL-6 were decreased under the influence of LPS in SH-SY5Y cells after taurine treatment (p < 0.01). Gene expression analysis revealed that increasing taurine concentration resulted in heightened expression of miRNA-181 and miRNA-21, with the most significant increase observed at a concentration of 0.75 mg/mL (p < 0.001). Conclusion Our study findings revealed that the expression of miRNA-181 and miRNA-21 can be enhanced by taurine. Consequently, exploring the targeting of taurine, miRNA-181, and miRNA-21 or considering hormone therapy may offer potential therapeutic approaches for treating AD or alleviating severe symptoms. Nonetheless, in order to fully comprehend the precise mechanisms involved, additional research is required.
Collapse
Affiliation(s)
- Hailah M. Almohaimeed
- Department of Basic Science, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Amany I. Almars
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fayez Alsulaimani
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed M. Basri
- Department of Medial Laboratory Sciences, Faculty of Applied Medical Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Norah A. Althobaiti
- Biology Department, College of Science and Humanities Al Quwaiiyah, Shaqra University, Al Quwaiiyah, Saudi Arabia
| | - Aishah E. Albalaw
- Department of Biology, Faculty of Science, University of Tabuk, Tabuk, Saudi Arabia
| | - Ifat Alsharif
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| | - Almonther Abdullah Hershan
- Department of Medical Microbiology and Parasitology, College of Medicine, The University of Jeddah, Jeddah, Saudi Arabia
| | - Mona H. Soliman
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza, Egypt
- Biology Department, Faculty of Science, Taibah University, Yanbu, Saudi Arabia
| |
Collapse
|
6
|
Olivares-González L, Velasco S, Campillo I, Millán JM, Rodrigo R. Redox Status in Retinitis Pigmentosa. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:443-448. [PMID: 37440070 DOI: 10.1007/978-3-031-27681-1_65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Retinitis pigmentosa (RP) is the most common form of inherited retinal dystrophy characterized by the progressive loss of vision. It is a rare disease. Despite being a genetic disease, its progression is influenced by oxidative damage and chemokines and cytokines released by the activated immune cells (e.g., macrophages or microglia). The role of oxidative stress is very important in the retina. Rods are the main consumers of oxygen (O2), so they are constantly exposed to oxidative stress and lipid peroxidation. According to the oxidative hypothesis, after rod death in the early stages of the disease, O2 would accumulate in large quantities in the retina, producing hyperoxia and favoring the accumulation of reactive oxygen species and reactive nitrogen species that would cause oxidative damage to lipids, proteins, and DNA, exacerbating the process of retinal degeneration. Evidence shows alterations in the antioxidant-oxidant state in patients and in animal models of RP. In recent years, therapeutic approaches aimed at reducing oxidative stress have emerged as useful therapies to slow down the progression of RP. We focus this review on oxidative stress and its relationship with the progression of RP.
Collapse
Affiliation(s)
- L Olivares-González
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center (CIPF), Valencia, Spain
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
| | - S Velasco
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center (CIPF), Valencia, Spain
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
| | - I Campillo
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center (CIPF), Valencia, Spain
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
| | - J M Millán
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain
- Rare Diseases Networking Biomedical Research Centre (CIBERER), Madrid, Spain
- Molecular, Cellular and Genomic Biomedicine, Health Research Institute La Fe, Valencia, Spain
| | - R Rodrigo
- Pathophysiology and Therapies for Vision Disorders, Principe Felipe Research Center (CIPF), Valencia, Spain.
- Joint Unit on Rare Diseases CIPF-La Fe, Valencia, Spain.
- Rare Diseases Networking Biomedical Research Centre (CIBERER), Madrid, Spain.
| |
Collapse
|
7
|
Eickhoff A, Tjaden J, Stahlke S, Vorgerd M, Theis V, Matschke V, Theiss C. Effects of progesterone on T-type-Ca 2+-channel expression in Purkinje cells. Neural Regen Res 2022; 17:2465-2471. [PMID: 35535898 PMCID: PMC9120685 DOI: 10.4103/1673-5374.339008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Plasticity of cerebellar Purkinje cells (PC) is influenced by progesterone via the classical progesterone receptors PR-A and PR-B by stimulating dendritogenesis, spinogenesis, and synaptogenesis in these cells. Dissociated PC cultures were used to analyze progesterone effects at a molecular level on the voltage-gated T-type-Ca2+-channels Cav3.1, Cav3.2, and Cav3.3 as they helped determine neuronal plasticity by regulating Ca2+-influx in neuronal cells. The results showed direct effects of progesterone on the mRNA expression of T-type-Ca2+-channels, as well as on the protein kinases A and C being involved in downstream signaling pathways that play an important role in neuronal plasticity. For the mRNA expression studies of T-type-Ca2+-channels and protein kinases of the signaling cascade, laser microdissection and purified PC cultures of different maturation stages were used. Immunohistochemical staining was also performed to characterize the localization of T-type-Ca2+-channels in PC. Experimental progesterone treatment was performed on the purified PC culture for 24 and 48 hours. Our results show that progesterone increases the expression of Cav3.1 and Cav3.3 and associated protein kinases A and C in PC at the mRNA level within 48 hours after treatment at latest. These effects extend the current knowledge of the function of progesterone in the central nervous system and provide an explanatory approach for its influence on neuronal plasticity.
Collapse
Affiliation(s)
- Annika Eickhoff
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Jonas Tjaden
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Sarah Stahlke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Neuromuscular Center Ruhrgebiet, University Hospital Bergmannsheil, Ruhr-Universität Bochum, Bochum, Germany
| | - Verena Theis
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Veronika Matschke
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| | - Carsten Theiss
- Department of Cytology, Institute of Anatomy, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
8
|
Sex Steroid Receptors in Polycystic Ovary Syndrome and Endometriosis: Insights from Laboratory Studies to Clinical Trials. Biomedicines 2022; 10:biomedicines10071705. [PMID: 35885010 PMCID: PMC9312843 DOI: 10.3390/biomedicines10071705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/13/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) and endometriosis are reproductive disorders that may cause infertility. The pathology of both diseases has been suggested to be associated with sex steroid hormone receptors, including oestrogen receptors (ER), progesterone receptors (PRs) and androgen receptors (ARs). Therefore, with this review, we aim to provide an update on the available knowledge of these receptors and how their interactions contribute to the pathogenesis of PCOS and endometriosis. One of the main PCOS-related medical conditions is abnormal folliculogenesis, which is associated with the downregulation of ER and AR expression in the ovaries. In addition, metabolic disorders in PCOS are caused by dysregulation of sex steroid hormone receptor expression. Furthermore, endometriosis is related to the upregulation of ER and the downregulation of PR expression. These receptors may serve as therapeutic targets for the treatment of PCOS-related disorders and endometriosis, considering their pathophysiological roles. Receptor agonists may be applied to increase the expression of a specific receptor and treat endometriosis or metabolic disorders. In contrast, receptor antagonist functions to reduce receptor expression and can be used to treat endometriosis and induce ovulation. Understanding PCOS and the pathological roles of endometriosis sex steroid receptors is crucial for developing potential therapeutic strategies to treat infertility in both conditions. Therefore, research should be continued to fill the knowledge gap regarding the subject.
Collapse
|
9
|
Sulliman NC, Ghaddar B, Gence L, Patche J, Rastegar S, Meilhac O, Diotel N. HDL biodistribution and brain receptors in zebrafish, using HDLs as vectors for targeting endothelial cells and neural progenitors. Sci Rep 2021; 11:6439. [PMID: 33742021 PMCID: PMC7979862 DOI: 10.1038/s41598-021-85183-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 02/24/2021] [Indexed: 12/25/2022] Open
Abstract
High density lipoproteins (HDLs) display pleiotropic functions such as anti-inflammatory, antioxidant, anti-protease, and anti-apoptotic properties. These effects are mediated by four main receptors: SCARB1 (SR-BI), ABCA1, ABCG1, and CD36. Recently, HDLs have emerged for their potential involvement in brain functions, considering their epidemiological links with cognition, depression, and brain plasticity. However, their role in the brain is not well understood. Given that the zebrafish is a well-recognized model for studying brain plasticity, metabolic disorders, and apolipoproteins, it could represent a good model for investigating the role of HDLs in brain homeostasis. By analyzing RNA sequencing data sets and performing in situ hybridization, we demonstrated the wide expression of scarb1, abca1a, abca1b, abcg1, and cd36 in the brain of adult zebrafish. Scarb1 gene expression was detected in neural stem cells (NSCs), suggesting a possible role of HDLs in NSC activity. Accordingly, intracerebroventricular injection of HDLs leads to their uptake by NSCs without modulating their proliferation. Next, we studied the biodistribution of HDLs in the zebrafish body. In homeostatic conditions, intraperitoneal injection of HDLs led to their accumulation in the liver, kidneys, and cerebral endothelial cells in zebrafish, similar to that observed in mice. After telencephalic injury, HDLs were diffused within the damaged parenchyma and were taken up by ventricular cells, including NSCs. However, they failed to modulate the recruitment of microglia cells at the injury site and the injury-induced proliferation of NSCs. In conclusion, our results clearly show a functional HDL uptake process involving several receptors that may impact brain homeostasis and suggest the use of HDLs as delivery vectors to target NSCs for drug delivery to boost their neurogenic activity.
Collapse
Affiliation(s)
- Nora Cassam Sulliman
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Batoul Ghaddar
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Laura Gence
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Jessica Patche
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Postfach 3640, 76021, Karlsruhe, Germany
| | - Olivier Meilhac
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France
- CHU de La Réunion, Saint-Denis de La Réunion, France
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188, Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Saint-Denis de La Réunion, France.
| |
Collapse
|
10
|
Kumar RG, DiSanto D, Awan N, Vaughan LE, Levochkina MS, Weppner JL, Wright DW, Berga SL, Conley YP, Brooks MM, Wagner AK. Temporal Acute Serum Estradiol and Tumor Necrosis Factor-α Associations and Risk of Death after Severe Traumatic Brain Injury. J Neurotrauma 2020; 37:2198-2210. [PMID: 32375598 DOI: 10.1089/neu.2019.6577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Severe traumatic brain injury (TBI) activates a robust systemic response that involves inflammatory and other factors, including estradiol (E2), associated with increased deaths. Tumor necrosis factor-alpha (TNFα) is a significant mediator of systemic shock, and it is an extra-gonadal transcription factor for E2 production. The study objectives were to test the hypotheses: (1) a positive feedback relationship exists between acute serum TNFα and E2; and (2) acute concentrations of E2 and TNFα are prognostic indicators of death after severe TBI. This prospective cohort study included N = 157 adults with severe TBI. Serum samples were collected for the first five days post-injury. The TNFα and E2 levels were averaged into two time epochs: first 72 h (T1) and second 72 h post-injury (T2). A cross-lag panel analysis conducted between T1 and T2 TNFα and E2 levels showed significant cross-lag effects: T1 TNFα and T1 E2 were related to T2 E2 and T2 TNFα, respectively. Cox proportional hazards multi variable regression models determined that increases in T1 E2 (hazard ratio [HR] = 1.79, 95% confidence interval [CI]: 1.15, 2.81), but not T2 E2 (HR = 0.91, 95% CI: 0.56, 1.47), were associated with increased risk of death. Increased T2 TNFα (HR = 2.47, 95% CI: 1.35, 4.53), and T1 TNFα (HR = 1.47, 95% CI: 0.99, 2.19), to a lesser degree, were associated with increased risk of death. Relationships of death with T2 TNFα and T1 E2 were mediated partially by cardiovascular, hepatic, and renal dysfunction. Both E2 and TNFα are systemic, reciprocally related biomarkers that may be indicative of systemic compromise and increased risk of death after severe TBI.
Collapse
Affiliation(s)
- Raj G Kumar
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dominic DiSanto
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Nabil Awan
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Leah E Vaughan
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marina S Levochkina
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Justin L Weppner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David W Wright
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| | - Sarah L Berga
- Department of Reproductive Endocrinology, University of Utah, Salt Lake City, Utah
| | - Yvette P Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Maria M Brooks
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Amy K Wagner
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania.,Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania.,Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
11
|
Maloney KA, Schmidt AT, Hanten GR, Levin HS. Executive dysfunction in children and adolescents with behavior disorders and traumatic brain injury. Child Neuropsychol 2020; 26:69-82. [PMID: 31311419 DOI: 10.1080/09297049.2019.1640868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 06/30/2019] [Indexed: 10/26/2022]
Abstract
Traumatic brain injury (TBI) is known to contribute to deficits in executive functioning (EF). Executive functioning abilities are disrupted in adolescents with either conduct disorder or oppositional defiant disorder, collectively known as disruptive behavior disorders (DBDs). There is little research on the relationship between executive dysfunction and DBDs in a group with a confirmed history of TBI. The current study endeavored to examine EF abilities, as measured by parent report on the Behavior Rating Inventory of Executive Function (BRIEF), in four groups: (1) adolescents with a TBI history and co-occurring DBDs history, (2) adolescents with a TBI history and no DBDs history, (3) adolescents with an orthopedic injury (OI) history and co-occurring DBDs history, and (4) adolescents with an OI history and no DBDs history. Groups were matched on the basis of age at injury and estimated socioeconomic status. Participants were evaluated at five time-points throughout the study, within 1 month of injury (initial assessment), 3, 12, 18, and 24 months post-injury. Results indicated the TBI and DBDs group was not significantly different from the OI and DBDs group, and both DBDs groups suffered higher levels of executive dysfunction than the TBI only and OI only groups, which were not significantly different from each other. Results also showed across the four groups, EF deficits were significantly lower at 1 month and 24 months post-injury, suggesting a positive trajectory in EF skill development. Results are discussed in terms of the prognostic importance of EF deficits in children with DBDs.
Collapse
Affiliation(s)
- Kelsey A Maloney
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Adam T Schmidt
- Department of Psychological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Gerri R Hanten
- Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| | - Harvey S Levin
- Physical Medicine and Rehabilitation, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
12
|
Duncan KA. Estrogen Formation and Inactivation Following TBI: What we Know and Where we Could go. Front Endocrinol (Lausanne) 2020; 11:345. [PMID: 32547495 PMCID: PMC7272601 DOI: 10.3389/fendo.2020.00345] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/04/2020] [Indexed: 01/27/2023] Open
Abstract
Traumatic brain injury (TBI) is responsible for various neuronal and cognitive deficits as well as psychosocial dysfunction. Characterized by damage inducing neuroinflammation, this response can cause an acute secondary injury that leads to widespread neurodegeneration and loss of neurological function. Estrogens decrease injury induced neuroinflammation and increase cell survival and neuroprotection and thus are a potential target for use following TBI. While much is known about the role of estrogens as a neuroprotective agent following TBI, less is known regarding their formation and inactivation following damage to the brain. Specifically, very little is known surrounding the majority of enzymes responsible for the production of estrogens. These estrogen metabolizing enzymes (EME) include aromatase, steroid sulfatase (STS), estrogen sulfotransferase (EST/SULT1E1), and some forms of 17β-hydroxysteroid dehydrogenase (HSD17B) and are involved in both the initial conversion and interconversion of estrogens from precursors. This article will review and offer new prospective and ideas on the expression of EMEs following TBI.
Collapse
|
13
|
Abstract
In aerobes, oxygen is essential for maintenance of life. However, incomplete reduction of oxygen leads to generation of reactive oxygen species. These oxidants oxidise biological macromolecules present in their vicinity and thereby impair cellular functions causing oxidative stress (OS). Aerobes have evolved both enzymatic and nonenzymatic antioxidant defences to protect themselves from OS. Although hormones as means of biological coordination involve in regulation of physiological activities of tissues by regulating metabolism, any change in their normal titre leads to pathophysiological states. While, hormones such as melatonin, insulin, oestrogen, progesterone display antioxidant features, thyroid hormone, corticosteroids and catecholamines elicit free radical generation and OS, and the role of testosterone in inducing OS is debateable. This review is an attempt to understand the impact of free radical generation and cross talk between the hormones modulating antioxidant defence system under various pathophysiological conditions.
Collapse
Affiliation(s)
- Gagan B N Chainy
- Department of Biotechnology, Utkal University, Bhubaneswar, India
| | | |
Collapse
|
14
|
Biological Sex/Gender and Biopsychosocial Determinants of Traumatic Brain Injury Recovery Trajectories. CURRENT PHYSICAL MEDICINE AND REHABILITATION REPORTS 2019. [DOI: 10.1007/s40141-019-00238-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
15
|
Benlloch-Navarro S, Trachsel-Moncho L, Fernández-Carbonell Á, Olivar T, Soria JM, Almansa I, Miranda M. Progesterone anti-inflammatory properties in hereditary retinal degeneration. J Steroid Biochem Mol Biol 2019; 189:291-301. [PMID: 30654106 DOI: 10.1016/j.jsbmb.2019.01.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/07/2019] [Accepted: 01/13/2019] [Indexed: 01/20/2023]
Abstract
The interactions between steroid gonadal hormones and the retina (a part of the visual system and the central nervous system (CNS)) have received limited attention and beneficial effects of these hormones in retinal diseases is controversial. Retinitis pigmentosa (RP) is the most common cause of retinal hereditary blindness and to date no treatment is available. However, results regarding the effects of progesterone on the progression of RP are promising. With the idea of demonstrating if the progesterone retinal protection in RP is related to its possible anti-inflammatory properties, we have administered orally progesterone to rd10 mice, an animal model of RP. We observed that progesterone decreased photoreceptors cell death, reactive gliosis and the increase in microglial cells caused by RP. We also examined the expression of neuronal and inducible nitric oxide synthase (nNOS and iNOS), the enzyme responsible for NO production. The results demonstrated a decrease in nNOS expression only in control mice treated with progesterone. Inflammation has been related with an increase in lipid peroxidation. Noticeably progesterone administration was able to diminish retinal malondialdehyde (MDA, a lipid peroxidation product) concentrations in rd10 mice. Altogether, we can conclude that progesterone could be a good therapeutic option not only in RP but also for other retinal diseases that have been associated with inflammation and lipid peroxidation.
Collapse
Affiliation(s)
- Soledad Benlloch-Navarro
- Departamento Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain
| | - Laura Trachsel-Moncho
- Departamento Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain
| | | | - Teresa Olivar
- Departamento Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain
| | - José Miguel Soria
- Departamento Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain; Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain
| | - Inmaculada Almansa
- Departamento Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain; Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain.
| | - María Miranda
- Departamento Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain; Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU Universities, Valencia, Spain.
| |
Collapse
|
16
|
Progesterone, Lipoic Acid, and Sulforaphane as Promising Antioxidants for Retinal Diseases: A Review. Antioxidants (Basel) 2019; 8:antiox8030053. [PMID: 30832304 PMCID: PMC6466531 DOI: 10.3390/antiox8030053] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress has been documented to be a key factor in the cause and progression of different retinal diseases. Oxidative cellular unbalance triggers a sequence of reactions which prompt cell degeneration and retinal dysfunction, both hallmarks of several retinal pathologies. There is no effective treatment, yet, for many retinal diseases. Antioxidant treatment have been pointed out to be an encouraging palliative treatment; the beneficial effects documented involve slowing the progression of the disease, a reduction of cell degeneration, and improvement of retinal functions. There is a vast information corpus on antioxidant candidates. In this review, we expose three of the main antioxidant treatments, selected for their promising results that has been reported to date. Recently, the sulforaphane, an isothiocyanate molecule, has been unveiled as a neuroprotective candidate, by its antioxidant properties. Progesterone, a neurosteroid has been proposed to be a solid and effective neuroprotective agent. Finally, the lipoic acid, an organosulfur compound, is a well-recognized antioxidant. All of them, have been tested and studied on different retinal disease models. In this review, we summarized the published results of these works, to offer a general view of the current antioxidant treatment advances, including the main effects and mechanisms described.
Collapse
|
17
|
A Repeated Measures Pilot Comparison of Trajectories of Fluctuating Endogenous Hormones in Young Women with Traumatic Brain Injury, Healthy Controls. Behav Neurol 2019; 2019:7694503. [PMID: 30891100 PMCID: PMC6390250 DOI: 10.1155/2019/7694503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 11/20/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022] Open
Abstract
Objective To compare baseline and 72-hour hormone levels in women with traumatic brain injury (TBI) and controls. Setting Hospital emergency department. Participants 21 women ages 18-35 with TBI and 21 controls. Design Repeated measures. Main Measures Serum samples at baseline and 72 hours; immunoassays for estradiol (E2), progesterone (PRO), luteinizing hormone (LH), follicle-stimulating hormone (FSH), and cortisol (CORT); and health history. Results Women with TBI had lower E2 (p = 0.042) and higher CORT (p = 0.028) levels over time. Lower Glasgow Coma Scale (GSC) and OCs were associated with lower FSH (GCS p = 0.021; OCs p = 0.016) and higher CORT (GCS p = 0.001; OCs p = 0.008). Conclusion Acute TBI may suppress E2 and increase CORT in young women. OCs appeared to independently affect CORT and FSH responses. Future work is needed with a larger sample to characterize TBI effects on women's endogenous hormone response to injury and OC use's effects on post-TBI stress response and gonadal function, as well as secondary injury.
Collapse
|
18
|
Atwood CS, Ekstein SF. Human versus non-human sex steroid use in hormone replacement therapies part 1: Preclinical data. Mol Cell Endocrinol 2019; 480:12-35. [PMID: 30308266 DOI: 10.1016/j.mce.2018.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 03/12/2018] [Accepted: 10/04/2018] [Indexed: 11/24/2022]
Abstract
Prior to 2002, hormone replacement therapy (HRT) was considered to be an important component of postmenopausal healthcare. This was based on a plethora of basic, epidemiological and clinical studies demonstrating the health benefits of supplementation with human sex steroids. However, adverse findings from the Women's Health Initiative (WHI) studies that examined the 2 major forms of HRT in use in the US at that time - Premarin (conjugated equine estrogens; CEE) and Prempro (CEE + medroxyprogesterone acetate; MPA), cast a shadow over the use of any form of HRT. Here we review the biochemical and physiological differences between the non-human WHI study hormones - CEE and MPA, and their respective human counterparts 17β-estradiol (E2) and progesterone (P4). Preclinical data from the last 30 years demonstrate clear differences between human and non-human sex steroids on numerous molecular, physiological and functional parameters in brain, heart and reproductive tissue. In contrast to CEE supplementation, which is not always detrimental although certainly not as optimal as E2 supplementation, MPA is clearly not equivalent to P4, having detrimental effects on cognitive, cardiac and reproductive function. Moreover, unlike P4, MPA is clearly antagonistic of the positive effects of E2 and CEE on tissue function. These data indicate that minor chemical changes to human sex steroids result in physiologically distinct actions that are not optimal for tissue health and functioning.
Collapse
Affiliation(s)
- Craig S Atwood
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, USA; Geriatric Research, Education and Clinical Center, Veterans Administration Hospital, Madison, WI, 53705, USA; School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, 6027, WA, Australia.
| | - Samuel F Ekstein
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, USA
| |
Collapse
|
19
|
Robison LS, Gannon OJ, Salinero AE, Zuloaga KL. Contributions of sex to cerebrovascular function and pathology. Brain Res 2018; 1710:43-60. [PMID: 30580011 DOI: 10.1016/j.brainres.2018.12.030] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/13/2022]
Abstract
Sex differences exist in how cerebral blood vessels function under both physiological and pathological conditions, contributing to observed sex differences in risk and outcomes of cerebrovascular diseases (CBVDs), such as vascular contributions to cognitive impairment and dementia (VCID) and stroke. Throughout most of the lifespan, women are protected from CBVDs; however, risk increases following menopause, suggesting sex hormones may play a significant role in this protection. The cerebrovasculature is a target for sex hormones, including estrogens, progestins, and androgens, where they can influence numerous vascular functions and pathologies. While there is a plethora of information on estrogen, the effects of progestins and androgens on the cerebrovasculature are less well-defined. Estrogen decreases cerebral tone and increases cerebral blood flow, while androgens increase tone. Both estrogens and androgens enhance angiogenesis/cerebrovascular remodeling. While both estrogens and androgens attenuate cerebrovascular inflammation, pro-inflammatory effects of androgens under physiological conditions have also been demonstrated. Sex hormones exert additional neuroprotective effects by attenuating oxidative stress and maintaining integrity and function of the blood brain barrier. Most animal studies utilize young, healthy, gonadectomized animals, which do not mimic the clinical conditions of aging individuals likely to get CBVDs. This is also concerning, as sex hormones appear to mediate cerebrovascular function differently based on age and disease state (e.g. metabolic syndrome). Through this review, we hope to inspire others to consider sex as a key biological variable in cerebrovascular research, as greater understanding of sex differences in cerebrovascular function will assist in developing personalized approaches to prevent and treat CBVDs.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Olivia J Gannon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Abigail E Salinero
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| | - Kristen L Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave, Albany, NY 12208, United States.
| |
Collapse
|
20
|
Mirshekar MA, Sarkaki A, Farbood Y, Gharib Naseri MK, Badavi M, Mansouri MT, Haghparast A. Neuroprotective effects of gallic acid in a rat model of traumatic brain injury: behavioral, electrophysiological, and molecular studies. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2018; 21:1056-1063. [PMID: 30524680 PMCID: PMC6281072 DOI: 10.22038/ijbms.2018.29639.7165] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Objective(s): Traumatic brain injury (TBI) is one of the main causes of intellectual and cognitive disabilities. Clinically, it is essential to limit the development of cognitive impairment after TBI. In the present study, the neuroprotective effects of gallic acid (GA) on neurological score, memory, long-term potentiation (LTP) from hippocampal dentate gyrus (hDG), brain lipid peroxidation and cytokines after TBI were evaluated. Materials and Methods: Seventy-two adult male Wistar rats divided randomly into three groups with 24 in each: Veh + Sham, Veh + TBI and GA + TBI (GA; 100 mg/kg, PO for 7 days before TBI induction). Brain injury was made by Marmarou’s method. Briefly, a 200 g weight was fallen down from a 2 m height through a free-falling tube onto the head of anesthetized animal. Results: Veterinary coma scores (VCS), memory and recorded hDG -LTP significantly reduced in Veh + TBI group at 1 and 24 hr after TBI when compared to Veh + Sham (P<0.001), respectively, while brain tissue content of interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α) and malondialdehyde (MDA) were increased significantly (P<0.001). Pretreatment of TBI rats with GA improved clinical signs, memory and hDG-LTP significantly (P<0.001) compared to Veh + TBI group, while brain tissue content of IL-1β, IL-6, TNF-α and MDA were decreased significantly (P<0.001). Conclusion: Our results propose that GA has neuroprotective effect on memory and LTP impairment due to TBI through decrement of brain lipid peroxidation and cerebral pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Mohammad Ali Mirshekar
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Physiology, School of Medicine and Clinical Immunology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Alireza Sarkaki
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoub Farbood
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Mohammad Badavi
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Taghi Mansouri
- Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Department of Pharmacology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Muñoz-Mayorga D, Guerra-Araiza C, Torner L, Morales T. Tau Phosphorylation in Female Neurodegeneration: Role of Estrogens, Progesterone, and Prolactin. Front Endocrinol (Lausanne) 2018; 9:133. [PMID: 29643836 PMCID: PMC5882780 DOI: 10.3389/fendo.2018.00133] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/14/2018] [Indexed: 01/01/2023] Open
Abstract
Sex differences are important to consider when studying different psychiatric, neurodevelopmental, and neurodegenerative disorders, including Alzheimer's disease (AD). These disorders can be affected by dimorphic changes in the central nervous system and be influenced by sex-specific hormones and neuroactive steroids. In fact, AD is more prevalent in women than in men. One of the main characteristics of AD is the formation of neurofibrillary tangles, composed of the phosphoprotein Tau, and neuronal loss in specific brain regions. The scope of this work is to review the existing evidence on how a set of hormones (estrogen, progesterone, and prolactin) affect tau phosphorylation in the brain of females under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Daniel Muñoz-Mayorga
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - Teresa Morales
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
- *Correspondence: Teresa Morales,
| |
Collapse
|
22
|
Oh H, Park J, Seo W. A 2-year prospective follow-up study of temporal changes associated with post-stroke cognitive impairment. Int J Nurs Pract 2018; 24:e12618. [PMID: 29291599 DOI: 10.1111/ijn.12618] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 11/03/2017] [Accepted: 11/11/2017] [Indexed: 12/15/2022]
Abstract
AIMS To explore temporal patterns of change in cognitive impairments during the 2 years following stroke and to identify factors that affect these temporal changes. BACKGROUND Despite the prognostic importance, temporal changes in post-stroke cognitive impairment have not been systematically investigated. DESIGN A non-experimental, prospective, longitudinal descriptive study design. METHODS Fifty-two stroke patients were enrolled. Data were collected from April 2015 to September 2017. Cognitive function was evaluated at 5 different times (immediately, and at 3, 6, 12, and 24 months post-stroke). RESULTS Significant changes in cognitive function following stroke exhibited an "s-shaped" curve, and the most rapid changes were observed between 3 and 6 months after stroke. The incidence of post-stroke cognitive impairment ranged from 23.1% to 42.3% and was highest at 3 months and lowest at 6 months. Gender, educational level, pre-stroke cognitive and functional abilities, haematoma, and brain surgery were associated with incidence of post-stroke cognitive impairment. CONCLUSIONS The ongoing changes exhibited by patterns of cognitive impairment provide evidence that consistent efforts are required to achieve positive changes in post-stroke cognitive function. Our findings may be helpful to develop nursing care strategies aimed at improving cognitive ability and consequently the quality of life of stroke patients.
Collapse
Affiliation(s)
- HyunSoo Oh
- Department of Nursing, Inha University, Incheon, Republic of Korea
| | - JongSuk Park
- Department of Nursing, Inha University; and Nurse, Inha University Hospital, Incheon, Republic of Korea
| | - WhaSook Seo
- Department of Nursing, Inha University, Incheon, Republic of Korea
| |
Collapse
|
23
|
Oh H, Lee K, Shin S, Seo W. Temporal Patterns and Influential Factors of Blood Glucose Levels During the First 10-Day Critical Period After Brain Injury. Clin Nurs Res 2017; 28:744-761. [PMID: 29254374 DOI: 10.1177/1054773817749725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This study was conducted to document temporal patterns of blood glucose level changes during the first 10-day critical period and to identify factors that influence stress-induced hyperglycemia development in brain injury patients. The medical records of 190 brain injury patients were retrospectively reviewed. Blood glucose levels in the poor recovery group were significantly higher than in the good recovery group, particularly during the first 72 hr (158-172 mg/dl). The poor recovery group showed persistent, fluctuating hyperglycemia, whereas the good recovery group exhibited hyperglycemic peaks during the first 3 days that subsequently reduced linearly to normal. Gender, preexisting hypertension, disease severity at admission, total calorie intake, and steroid use were found to influence stress-induced hyperglycemia development significantly. In conclusion, close monitoring and adjustment are required to maintain safe blood glucose levels and the development of protocols for safe glycemic management is essential to improve critical care in brain injury patients.
Collapse
Affiliation(s)
- HyunSoo Oh
- 1 Inha University, Incheon, Republic of Korea
| | - KangIm Lee
- 1 Inha University, Incheon, Republic of Korea
| | | | - WhaSook Seo
- 1 Inha University, Incheon, Republic of Korea
| |
Collapse
|
24
|
Effects of Female Sex Steroids Administration on Pathophysiologic Mechanisms in Traumatic Brain Injury. Transl Stroke Res 2017; 9:393-416. [PMID: 29151229 DOI: 10.1007/s12975-017-0588-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/16/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
Secondary brain damage following initial brain damage in traumatic brain injury (TBI) is a major cause of adverse outcomes. There are many gaps in TBI research and a lack of therapy to limit debilitating outcomes in TBI or enhance the neurogenesis, despite pre-clinical and clinical research performed in TBI. Females show harmful outcomes against brain damage including TBI less than males, independent of different TBI occurrence. A significant reduction in secondary brain damage and improvement in neurologic outcome post-TBI has been reported following the use of progesterone and estrogen in many experimental studies. Although useful features of sex steroids including progesterone have been identified in TBI clinical trials I and II, clinical trials III have been unsuccessful. This review article focuses on evidence of secondary injury mechanisms and neuroprotective effects of estrogen and progesterone in TBI. Understanding these mechanisms may enable researchers to achieve greater success in TBI clinical studies. It seems that the design of clinical studies should be revised due to translation loss of animal studies to clinical studies. The heterogeneous and complex nature of TBI, the endogenous levels of sex hormones at the time of taking these hormones, the therapeutic window of the drug, the dosage of the drug, the selection of appropriate targets in evaluation, the determination of responsive population, gender and age based on animal studies should be considered in the design of TBI human studies in future.
Collapse
|
25
|
Mirshekar MA, Fanaei H, Keikhaei F, Javan FS. Diosmin improved cognitive deficit and amplified brain electrical activity in the rat model of traumatic brain injury. Biomed Pharmacother 2017; 93:1220-1229. [PMID: 28738538 DOI: 10.1016/j.biopha.2017.07.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Traumatic brain injury (TBI) is one of the main causes of intellectual and cognitive disabilities in humans. Clinically, it is essential to limit the progress of cognitive impairment after TBI. It is reported that diosmin has a neuroprotective effect that can limit the progress of the impairment. The aim of this study was to evaluate the effects of diosmin on neurological score, memory, tumor necrosis factor-α (TNF-α) level and long-term potentiation in hippocampal dentate gyrus after the injury. METHODS A total of ninety six adult male Wistar rats were used as test subjects in this study. The animals were randomly assigned into one of the following three groups (n=32/group): Sham, TBI and diosmin (100mg/kg, p.o for seven consecutive days before TBI induction). TBI was induced into the animals by Marmarou's method. Briefly, a 200g weight was dropped from a 1m height through a free-falling tube onto the head of the anesthetized rats. RESULTS The veterinary coma scale scores, memory and long-term potentiation in TBI group showed significant decrease at different times after the onset of TBI when compared with Sham (p<0.001). The TNF-α level in the hippocampus of the TBI group of animals was significantly higher than that found in the test subjects from the Sham group (p<0.001). The pre-treatment of the TBI group with diosmin significantly improved their neurological scores, memory and long-term potentiation (p<0.001) when compared with the TBI group. The TNF-α level in hippocampus of the diosmin group was significantly lower than the TBI group (p<0.001). CONCLUSION Based on the results of the present study, pre-treatment with diosmin has protective effects against TBI-induced memory and long-term potentiation impairment. The effects of diosmin may be mediated through a decrement in the TNF-α concentration of hippocampus as a pro-inflammatory cytokine.
Collapse
Affiliation(s)
- Mohammad Ali Mirshekar
- Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Hamed Fanaei
- Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran; Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Fereshteh Keikhaei
- Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fatemeh Sargolzaee Javan
- Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
26
|
Maggio DM, Singh A, Iorgulescu JB, Bleicher DH, Ghosh M, Lopez MM, Tuesta LM, Flora G, Dietrich WD, Pearse DD. Identifying the Long-Term Role of Inducible Nitric Oxide Synthase after Contusive Spinal Cord Injury Using a Transgenic Mouse Model. Int J Mol Sci 2017; 18:ijms18020245. [PMID: 28125047 PMCID: PMC5343782 DOI: 10.3390/ijms18020245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 01/05/2017] [Accepted: 01/15/2017] [Indexed: 02/07/2023] Open
Abstract
Inducible nitric oxide synthase (iNOS) is a potent mediator of oxidative stress during neuroinflammation triggered by neurotrauma or neurodegeneration. We previously demonstrated that acute iNOS inhibition attenuated iNOS levels and promoted neuroprotection and functional recovery after spinal cord injury (SCI). The present study investigated the effects of chronic iNOS ablation after SCI using inos-null mice. iNOS-/- knockout and wild-type (WT) control mice underwent a moderate thoracic (T8) contusive SCI. Locomotor function was assessed weekly, using the Basso Mouse Scale (BMS), and at the endpoint (six weeks), by footprint analysis. At the endpoint, the volume of preserved white and gray matter, as well as the number of dorsal column axons and perilesional blood vessels rostral to the injury, were quantified. At weeks two and three after SCI, iNOS-/- mice exhibited a significant locomotor improvement compared to WT controls, although a sustained improvement was not observed during later weeks. At the endpoint, iNOS-/- mice showed significantly less preserved white and gray matter, as well as fewer dorsal column axons and perilesional blood vessels, compared to WT controls. While short-term antagonism of iNOS provides histological and functional benefits, its long-term ablation after SCI may be deleterious, blocking protective or reparative processes important for angiogenesis and tissue preservation.
Collapse
Affiliation(s)
- Dominic M Maggio
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, VA 22908, USA.
- Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institute of Heath, Bethesda, MD 20824, USA.
| | - Amanpreet Singh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - J Bryan Iorgulescu
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Drew H Bleicher
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Michael M Lopez
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Luis M Tuesta
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
| | - Govinder Flora
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Cell Biology and Anatomy, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
- Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA.
| |
Collapse
|
27
|
Brotfain E, Gruenbaum SE, Boyko M, Kutz R, Zlotnik A, Klein M. Neuroprotection by Estrogen and Progesterone in Traumatic Brain Injury and Spinal Cord Injury. Curr Neuropharmacol 2017; 14:641-53. [PMID: 26955967 PMCID: PMC4981744 DOI: 10.2174/1570159x14666160309123554] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 12/31/2015] [Accepted: 02/25/2016] [Indexed: 12/25/2022] Open
Abstract
In recent years there has been a growing body of clinical and laboratory evidence demonstrating the neuroprotective effects of estrogen and progesterone after traumatic brain injury (TBI) and spinal cord injury (SCI). In humans, women have been shown to have a lower incidence of morbidity and mortality after TBI compared with age-matched men. Similarly, numerous laboratory studies have demonstrated that estrogen and progesterone administration is associated with a mortality reduction, improvement in neurological outcomes, and a reduction in neuronal apoptosis after TBI and SCI. Here, we review the evidence that supports hormone-related neuroprotection and discuss possible underlying mechanisms. Estrogen and progesterone-mediated neuroprotection are thought to be related to their effects on hormone receptors, signaling systems, direct antioxidant effects, effects on astrocytes and microglia, modulation of the inflammatory response, effects on cerebral blood flow and metabolism, and effects on mediating glutamate excitotoxicity. Future laboratory research is needed to better determine the mechanisms underlying the hormones' neuroprotective effects, which will allow for more clinical studies. Furthermore, large randomized clinical control trials are needed to better assess their role in human neurodegenerative conditions.
Collapse
Affiliation(s)
- Evgeni Brotfain
- Department of Anesthesiology and Critical Care, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | | | | | | | | | |
Collapse
|
28
|
Hirst JJ, Cumberland AL, Shaw JC, Bennett GA, Kelleher MA, Walker DW, Palliser HK. Loss of neurosteroid-mediated protection following stress during fetal life. J Steroid Biochem Mol Biol 2016; 160:181-8. [PMID: 26365557 DOI: 10.1016/j.jsbmb.2015.09.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/01/2015] [Accepted: 09/08/2015] [Indexed: 11/22/2022]
Abstract
Elevated levels of neurosteroids during late gestation protect the fetal brain from hypoxia/ischaemia and promote neurodevelopment. Suppression of allopregnanolone production during pregnancy leads to the onset of seizure-like activity and potentiates hypoxia-induced brain injury. Markers of myelination are reduced and astrocyte activation is increased. The placenta has a key role in maintaining allopregnanolone concentrations in the fetal circulation and brain during gestation and levels decline markedly after both normal and preterm birth. This leads to the preterm neonate developing in a neurosteroid deficient environment between delivery and term equivalence. The expression of 5α-reductases is also lower in the fetus prior to term. These deficiencies in neurosteroid exposure may contribute to the increase in incidence of the adverse patterns of behaviour seen in children that are born preterm. Repeated exposure to glucocorticoid stimulation suppresses 5α-reductase expression and allopregnanolone levels in the fetus and results in reduced myelination. Both fetal growth restriction and prenatal maternal stress lead to increased cortisol concentrations in the maternal and fetal circulation. Prenatal stress results in reduced expression of key GABAA receptor subunits that normally heighten neurosteroid sensitivity. These stressors also result in altered placental allopregnanolone metabolism pathways. These findings suggest that reduced neurosteroid production and action in the perinatal period may contribute to some of the adverse neurodevelopmental and behavioural outcomes that result from these pregnancy compromises. Studies examining perinatal steroid supplementation therapy with non-metabolisable neurosteroid analogues to improve these outcomes are warranted.
Collapse
Affiliation(s)
- Jonathan J Hirst
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia.
| | - Angela L Cumberland
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Julia C Shaw
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | - Greer A Bennett
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| | | | - David W Walker
- Ritchie Centre for Baby Health Research, Department of Obstetrics and Gynaecology, Monash University, VIC 3800, Australia
| | - Hannah K Palliser
- School of Biomedical Sciences, University of Newcastle, Callaghan, NSW 2308, Australia
| |
Collapse
|
29
|
Goldstein FC, Caveney AF, Hertzberg VS, Silbergleit R, Yeatts SD, Palesch YY, Levin HS, Wright DW. Very Early Administration of Progesterone Does Not Improve Neuropsychological Outcomes in Subjects with Moderate to Severe Traumatic Brain Injury. J Neurotrauma 2016; 34:115-120. [PMID: 26973025 DOI: 10.1089/neu.2015.4313] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A Phase III, double-blind, placebo-controlled trial (ProTECT III) found that administration of progesterone did not reduce mortality or improve functional outcome as measured by the Glasgow Outcome Scale Extended (GOSE) in subjects with moderate to severe traumatic brain injury. We conducted a secondary analysis of neuropsychological outcomes to evaluate whether progesterone is associated with improved recovery of cognitive and motor functioning. ProTECT III was conducted at 49 level I trauma centers in the United States. Adults with moderate to severe TBI were randomized to receive intravenous progesterone or placebo within 4 h of injury for a total of 4 days. At 6 months, subjects underwent evaluation of memory, attention, executive functioning, language, and fine motor coordination/dexterity. Chi-square analysis revealed no significant difference in the proportion of subjects (263/280 progesterone, 283/295 placebo) with Galveston Orientation and Amnesia Test scores ≥75. Analyses of covariance did not reveal significant treatment effects for memory (Buschke immediate recall, p = 0.53; delayed recall, p = 0.94), attention (Trails A speed, p = 0.81 and errors, p = 0.22; Digit Span Forward length, p = 0.66), executive functioning (Trails B speed, p = 0.97 and errors, p = 0.93; Digit Span Backward length, p = 0.60), language (timed phonemic fluency, p = 0.05), and fine motor coordination/dexterity (Grooved Pegboard dominant hand time, p = 0.75 and peg drops, p = 0.59; nondominant hand time, p = 0.74 and peg drops, p = 0.61). Pearson Product Moment Correlations demonstrated significant (p < 0.001) associations between better neuropsychological performance and higher GOSE scores. Similar to the ProTECT III trial's results of the primary outcome, the secondary outcomes do not provide evidence of a neuroprotective effect of progesterone.
Collapse
Affiliation(s)
- Felicia C Goldstein
- 1 Department of Neurology, Emory University School of Medicine , Atlanta, Georgia
| | - Angela F Caveney
- 2 Department of Psychiatry, University of Michigan , Ann Arbor, Michigan
| | - Vicki S Hertzberg
- 3 School of Nursing, Emory University School of Medicine , Atlanta, Georgia
| | - Robert Silbergleit
- 4 Department of Emergency Medicine, University of Michigan Medical School , Ann Arbor, Michigan
| | - Sharon D Yeatts
- 5 Department of Public Health Sciences, Medical University of South Carolina , Charleston, South Carolina
| | - Yuko Y Palesch
- 5 Department of Public Health Sciences, Medical University of South Carolina , Charleston, South Carolina
| | - Harvey S Levin
- 6 Department of Physical Medicine & Rehabilitation, Baylor College of Medicine , Houston, Texas
| | - David W Wright
- 7 Department of Emergency Medicine, Emory University School of Medicine , Atlanta, Georgia
| |
Collapse
|
30
|
Coronel MF, Raggio MC, Adler NS, De Nicola AF, Labombarda F, González SL. Progesterone modulates pro-inflammatory cytokine expression profile after spinal cord injury: Implications for neuropathic pain. J Neuroimmunol 2016; 292:85-92. [DOI: 10.1016/j.jneuroim.2016.01.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/11/2016] [Accepted: 01/14/2016] [Indexed: 12/30/2022]
|
31
|
Hsieh JT, Lei B, Sheng H, Venkatraman T, Lascola CD, Warner DS, James ML. Sex-Specific Effects of Progesterone on Early Outcome of Intracerebral Hemorrhage. Neuroendocrinology 2016; 103:518-30. [PMID: 26356626 DOI: 10.1159/000440883] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 09/07/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Preclinical evidence suggests that progesterone improves recovery after intracerebral hemorrhage (ICH); however, gonadal hormones have sex-specific effects. Therefore, an experimental model of ICH was used to assess recovery after progesterone administration in male and female rats. METHODS ICH was induced in male and female Wistar rats via stereotactic intrastriatal injection of clostridial collagenase (0.5 U). Animals were randomized to receive vehicle or 8 mg/kg progesterone intraperitoneally at 2 h, then subcutaneously at 5, 24, 48, and 72 h after injury. Outcomes included relevant physiology during the first 3 h, hemorrhage and edema evolution over the first 24 h, proinflammatory transcription factor and cytokine regulation at 24 h, rotarod latency and neuroseverity score over the first 7 days, and microglial activation/macrophage recruitment at 7 days after injury. RESULTS Rotarod latency (p = 0.001) and neuroseverity score (p = 0.01) were improved in progesterone-treated males, but worsened in progesterone-treated females (p = 0.028 and p = 0.008, respectively). Progesterone decreased cerebral edema (p = 0.04), microglial activation/macrophage recruitment (p < 0.001), and proinflammatory transcription factor phosphorylated nuclear factor-x03BA;B p65 expression (p = 0.0038) in males but not females, independent of tumor necrosis factor-α, interleukin-6, and toll-like receptor-4 expression. Cerebral perfusion was increased in progesterone-treated males at 4 h (p = 0.043) but not 24 h after injury. Hemorrhage volume, arterial blood gases, glucose, and systolic blood pressure were not affected. CONCLUSIONS Progesterone administration improved early neurobehavioral recovery and decreased secondary neuroinflammation after ICH in male rats. Paradoxically, progesterone worsened neurobehavioral recovery and did not modify neuroinflammation in female rats. Future work should isolate mechanisms of sex-specific progesterone effects after ICH.
Collapse
|
32
|
Efficacy of progesterone for moderate to severe traumatic brain injury: a meta-analysis of randomized clinical trials. Sci Rep 2015; 5:13442. [PMID: 26304556 PMCID: PMC4548259 DOI: 10.1038/srep13442] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/31/2015] [Indexed: 11/08/2022] Open
Abstract
Progesterone has been shown to have neuroprotective effects in multiple animal models of brain injury, whereas the efficacy and safety in patients with traumatic brain injury (TBI) remains contentious. Here, a total of seven randomized controlled trials (RCTs) with 2492 participants were included to perform this meta-analysis. Compared with placebo, there was no significant decrease to be found in the rate of death or vegetative state for patients with acute TBI (RR = 0.88, 95%CI = 0.70, 1.09, p = 0.24). Furthermore, progesterone was not associated with good recovery in comparison with placebo (RR = 1.00, 95%CI = 0.88, 1.14, p = 0.95). Together, our study suggested that progesterone did not improve outcomes over placebo in the treatment of acute TBI.
Collapse
|
33
|
Connecting prosocial behavior to improved physical health: Contributions from the neurobiology of parenting. Neurosci Biobehav Rev 2015; 55:1-17. [DOI: 10.1016/j.neubiorev.2015.04.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 03/19/2015] [Accepted: 04/10/2015] [Indexed: 12/13/2022]
|
34
|
Sarkaki A, Farbood Y, Gharib-Naseri MK, Badavi M, Mansouri MT, Haghparast A, Mirshekar MA. Gallic acid improved behavior, brain electrophysiology, and inflammation in a rat model of traumatic brain injury. Can J Physiol Pharmacol 2015. [DOI: 10.1139/cjpp-2014-0546] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Traumatic brain injury (TBI) is one of the main causes of intellectual and cognitive disabilities. In the clinic it is essential to limit the development of cognitive impairment after TBI. In this study, the effects of gallic acid (GA; 100 mg/kg, per oral, from 7 days before to 2 days after TBI induction) on neurological score, passive avoidance memory, long-term potentiation (LTP) deficits, and levels of proinflammatory cytokines including interleukin-1 beta (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor-α (TNF-α) in the brain have been evaluated. Brain injury was induced following Marmarou’s method. Data were analyzed by one-way and repeated measures ANOVA followed by Tukey’s post-hoc test. The results indicated that memory was significantly impaired (p < 0.001) in the group treated with TBI + vehicle, together with deterioration of the hippocampal LTP and increased brain tissue levels of IL-1β, IL-6, and TNF-α. GA treatment significantly improved memory and LTP in the TBI rats. The brain tissue levels of IL-1β, IL-6, and TNF-α were significantly reduced (p < 0.001) in the group treated with GA. The results suggest that GA has neuroprotective properties against TBI-induced behavioral, electrophysiological, and inflammatory disorders, probably via the decrease of cerebral proinflammatory cytokines.
Collapse
Affiliation(s)
- Alireza Sarkaki
- Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Yaghoub Farbood
- Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Mohammad Badavi
- Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Taghi Mansouri
- Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Pharmacology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Mirshekar
- Ahvaz Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
35
|
Datto JP, Bastidas JC, Miller NL, Shah AK, Arheart KL, Marcillo AE, Dietrich WD, Pearse DD. Female Rats Demonstrate Improved Locomotor Recovery and Greater Preservation of White and Gray Matter after Traumatic Spinal Cord Injury Compared to Males. J Neurotrauma 2015; 32:1146-57. [PMID: 25715192 DOI: 10.1089/neu.2014.3702] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The possibility of a gender-related difference in recovery after spinal cord injury (SCI) remains a controversial subject. Current empirical animal research lacks sizable test groups to definitively determine whether significant differences exist. Evaluating locomotor recovery variances between sexes following a precise, clinically relevant spinal cord contusion model can provide valuable insight into a possible gender-related advantage in outcome post-SCI. In the current study, we hypothesized that by employing larger sample sizes in a reproducible contusive SCI paradigm, subtle distinctions in locomotor recovery between sexes, if they exist, would be elucidated through a broad range of behavioral tests. During 13 weeks of functional assessment after a thoracic (T8) contusive SCI in rat, significant differences owing to gender existed for the Basso, Beattie, and Bresnahan score and CatWalk hindlimb swing, support four, and single stance analyses. Significant differences in locomotor performance were noticeable as early as 4 weeks post-SCI. Stereological tissue-volume analysis determined that females, more so than males, also exhibited greater volumes of preserved gray and white matter within the injured cord segment as well as more spared ventral white matter area at the center of the lesion. The stereological tissue analysis differences favoring females directly correlated with the female rats' greater functional improvement observed at endpoint.
Collapse
Affiliation(s)
- Jeffrey P Datto
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - Johana C Bastidas
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - Nicole L Miller
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - Anna K Shah
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - Kristopher L Arheart
- 2 The Departments of Public Health Sciences, University of Miami Miller School of Medicine , Miami, Florida
| | - Alexander E Marcillo
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida
| | - W Dalton Dietrich
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida.,3 The Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida.,4 The Department of Cell Biology, University of Miami Miller School of Medicine , Miami, Florida.,5 The Department of Neurology, University of Miami Miller School of Medicine , Miami, Florida.,6 The Neuroscience Program, University of Miami Miller School of Medicine , Miami, Florida
| | - Damien D Pearse
- 1 The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine , Miami, Florida.,3 The Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida.,6 The Neuroscience Program, University of Miami Miller School of Medicine , Miami, Florida.,7 The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
36
|
Guennoun R, Labombarda F, Gonzalez Deniselle MC, Liere P, De Nicola AF, Schumacher M. Progesterone and allopregnanolone in the central nervous system: response to injury and implication for neuroprotection. J Steroid Biochem Mol Biol 2015; 146:48-61. [PMID: 25196185 DOI: 10.1016/j.jsbmb.2014.09.001] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 08/29/2014] [Accepted: 09/02/2014] [Indexed: 01/26/2023]
Abstract
Progesterone is a well-known steroid hormone, synthesized by ovaries and placenta in females, and by adrenal glands in both males and females. Several tissues are targets of progesterone and the nervous system is a major one. Progesterone is also locally synthesized by the nervous system and qualifies, therefore, as a neurosteroid. In addition, the nervous system has the capacity to bio-convert progesterone into its active metabolite allopregnanolone. The enzymes required for progesterone and allopregnanolone synthesis are widely distributed in brain and spinal cord. Increased local biosynthesis of pregnenolone, progesterone and 5α-dihydroprogesterone may be a part of an endogenous neuroprotective mechanism in response to nervous system injuries. Progesterone and allopregnanolone neuroprotective effects have been widely recognized. Multiple receptors or associated proteins may contribute to the progesterone effects: classical nuclear receptors (PR), membrane progesterone receptor component 1 (PGRMC1), membrane progesterone receptors (mPR), and γ-aminobutyric acid type A (GABAA) receptors after conversion to allopregnanolone. In this review, we will succinctly describe progesterone and allopregnanolone biosynthetic pathways and enzyme distribution in brain and spinal cord. Then, we will summarize our work on progesterone receptor distribution and cellular expression in brain and spinal cord; neurosteroid stimulation after nervous system injuries (spinal cord injury, traumatic brain injury, and stroke); and on progesterone and allopregnanolone neuroprotective effects in different experimental models including stroke and spinal cord injury. We will discuss in detail the neuroprotective effects of progesterone on the nervous system via PR, and of allopregnanolone via its modulation of GABAA receptors.
Collapse
Affiliation(s)
- R Guennoun
- UMR 788, Inserm and University Paris-Sud, 80 rue du Général Leclerc, 94276 Bicêtre, Kremlin-Bicêtre, France.
| | - F Labombarda
- Instituto de Biologia y Medicina Experimental and University of Buenos Aires, Argentina
| | | | - P Liere
- UMR 788, Inserm and University Paris-Sud, 80 rue du Général Leclerc, 94276 Bicêtre, Kremlin-Bicêtre, France
| | - A F De Nicola
- Instituto de Biologia y Medicina Experimental and University of Buenos Aires, Argentina
| | - M Schumacher
- UMR 788, Inserm and University Paris-Sud, 80 rue du Général Leclerc, 94276 Bicêtre, Kremlin-Bicêtre, France
| |
Collapse
|
37
|
Lazarus RC, Buonora JE, Jacobowitz DM, Mueller GP. Protein carbonylation after traumatic brain injury: cell specificity, regional susceptibility, and gender differences. Free Radic Biol Med 2015; 78:89-100. [PMID: 25462645 DOI: 10.1016/j.freeradbiomed.2014.10.507] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/07/2014] [Accepted: 10/10/2014] [Indexed: 12/15/2022]
Abstract
Protein carbonylation is a well-documented and quantifiable consequence of oxidative stress in several neuropathologies, including multiple sclerosis, Alzheimer׳s disease, and Parkinson׳s disease. Although oxidative stress is a hallmark of traumatic brain injury (TBI), little work has explored the specific neural regions and cell types in which protein carbonylation occurs. Furthermore, the effect of gender on protein carbonylation after TBI has not been studied. The present investigation was designed to determine the regional and cell specificity of TBI-induced protein carbonylation and how this response to injury is affected by gender. Immunohistochemistry was used to visualize protein carbonylation in the brains of adult male and female Sprague-Dawley rats subjected to controlled cortical impact (CCI) as an injury model of TBI. Cell-specific markers were used to colocalize the presence of carbonylated proteins in specific cell types, including astrocytes, neurons, microglia, and oligodendrocytes. Results also indicated that the injury lesion site, ventral portion of the dorsal third ventricle, and ventricular lining above the median eminence showed dramatic increases in protein carbonylation after injury. Specifically, astrocytes and limited regions of ependymal cells adjacent to the dorsal third ventricle and the median eminence were most susceptible to postinjury protein carbonylation. However, these patterns of differential susceptibility to protein carbonylation were gender dependent, with males showing significantly greater protein carbonylation at sites distant from the lesion. Proteomic analyses were also conducted and determined that the proteins most affected by carbonylation in response to TBI include glial fibrillary acidic protein, dihydropyrimidase-related protein 2, fructose-bisphosphate aldolase C, and fructose-bisphosphate aldolase A. Many other proteins, however, were not carbonylated by CCI. These findings indicate that there is both regional and protein specificity in protein carbonylation after TBI. The marked increase in carbonylation seen in ependymal layers distant from the lesion suggests a mechanism involving the transmission of a cerebral spinal fluid-borne factor to these sites. Furthermore, this process is affected by gender, suggesting that hormonal mechanisms may serve a protective role against oxidative stress.
Collapse
Affiliation(s)
- Rachel C Lazarus
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - John E Buonora
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - David M Jacobowitz
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Gregory P Mueller
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Department of Anatomy, Physiology, and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| |
Collapse
|
38
|
Skolnick BE, Maas AI, Narayan RK, van der Hoop RG, MacAllister T, Ward JD, Nelson NR, Stocchetti N. A clinical trial of progesterone for severe traumatic brain injury. N Engl J Med 2014; 371:2467-76. [PMID: 25493978 DOI: 10.1056/nejmoa1411090] [Citation(s) in RCA: 338] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Progesterone has been associated with robust positive effects in animal models of traumatic brain injury (TBI) and with clinical benefits in two phase 2 randomized, controlled trials. We investigated the efficacy and safety of progesterone in a large, prospective, phase 3 randomized clinical trial. METHODS We conducted a multinational placebo-controlled trial, in which 1195 patients, 16 to 70 years of age, with severe TBI (Glasgow Coma Scale score, ≤8 [on a scale of 3 to 15, with lower scores indicating a reduced level of consciousness] and at least one reactive pupil) were randomly assigned to receive progesterone or placebo. Dosing began within 8 hours after injury and continued for 120 hours. The primary efficacy end point was the Glasgow Outcome Scale score at 6 months after the injury. RESULTS Proportional-odds analysis with covariate adjustment showed no treatment effect of progesterone as compared with placebo (odds ratio, 0.96; confidence interval, 0.77 to 1.18). The proportion of patients with a favorable outcome on the Glasgow Outcome Scale (good recovery or moderate disability) was 50.4% with progesterone, as compared with 50.5% with placebo. Mortality was similar in the two groups. No relevant safety differences were noted between progesterone and placebo. CONCLUSIONS Primary and secondary efficacy analyses showed no clinical benefit of progesterone in patients with severe TBI. These data stand in contrast to the robust preclinical data and results of early single-center trials that provided the impetus to initiate phase 3 trials. (Funded by BHR Pharma; SYNAPSE ClinicalTrials.gov number, NCT01143064.).
Collapse
Affiliation(s)
- Brett E Skolnick
- From the Department of Neurosurgery, Cushing Neuroscience Institute, Hofstra North Shore-LIJ School of Medicine, Manhasset, NY (B.E.S., R.K.N.); the Department of Neurosurgery, University Hospital Antwerp and University of Antwerp, Edegem, Belgium (A.I.M.); BHR Pharma, Herndon (R.G.H., T.M., N.R.N.), and the Department of Neurosurgery, Virginia Commonwealth University, Richmond (J.D.W.) - both in Virginia; and the Department of Physiopathology and Transplantation, Milan University and Neuro Intensive Care Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Cà Granda Ospedale Maggiore Policlinico, Milan (N.S.)
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Spratt NJ, Tomkins AJ, Pepperall D, McLeod DD, Calford MB. Allopregnanolone and its precursor progesterone do not reduce injury after experimental stroke in hypertensive rats - role of postoperative temperature regulation? PLoS One 2014; 9:e107752. [PMID: 25248155 PMCID: PMC4172598 DOI: 10.1371/journal.pone.0107752] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 08/23/2014] [Indexed: 11/21/2022] Open
Abstract
Allopregnanolone is a neurosteroid synthesized from progesterone in brain. It increases inhibition through modulation of the gamma-aminobutyric acid type A (GABA-A) receptor. Both agents are putative neuroprotectants after ischemic stroke. We sought to confirm their effectiveness in a hypertensive rat stroke model, with intra- and post-operative temperature regulation. The primary study compared allopregnanolone, progesterone or vehicle control treatments, administered 105 minutes after induction of temporary middle cerebral artery occlusion in spontaneously hypertensive rats. Temperature was controlled intraoperatively and a heat mat used in the 6 hours postoperatively to permit animal temperature self-regulation. The primary outcome was infarct volume and secondary outcomes were tests of sensory and motor function. There was no significant effect of treatment on any outcome measure. Given prior reports of GABA-A receptor agonists causing hypothermia, follow-up experiments were conducted to examine postoperative temperature regulation. These did not reveal a difference in postoperative temperature in neurosteroid-treated animals compared to control. However, in all rats maintained postoperatively in ambient temperature, moderate hypothermia was observed. This was in contrast to rats maintained over a heat mat. The lowest mean postoperative temperature was between 34.4-34.9°C in all 3 groups. These data do not support a neuroprotective effect of allopregnanolone or progesterone in ischemic stroke in hypertensives in the setting of normothermia. Given previous evidence of synergy between neuroprotective agents and hypothermia, demonstration of neuroprotective effect of these agents in the absence of postoperative hypothermia would be prudent before consideration of these agents for further clinical investigation.
Collapse
Affiliation(s)
- Neil J. Spratt
- School of Biomedical Sciences, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
- Hunter New England Local Health District, Newcastle, Australia
| | - Amelia J. Tomkins
- School of Biomedical Sciences, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
| | - Debbie Pepperall
- School of Biomedical Sciences, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
| | - Damian D. McLeod
- School of Biomedical Sciences, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
| | - Mike B. Calford
- School of Biomedical Sciences, University of Newcastle, and Hunter Medical Research Institute, Newcastle, Australia
- School of Medicine, The University of Tasmania, Hobart, Australia
| |
Collapse
|
40
|
Xu FF, Sun S, Ho ASW, Lee D, Kiang KMY, Zhang XQ, Wang AM, Wu EX, Lui WM, Liu BY, Leung GKK. Effects of progesterone vs. dexamethasone on brain oedema and inflammatory responses following experimental brain resection. Brain Inj 2014; 28:1594-601. [PMID: 25093611 DOI: 10.3109/02699052.2014.943289] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Fei-Fan Xu
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University
BeijingPR China
| | - Stella Sun
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
| | - Amy S. W. Ho
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
| | - Derek Lee
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
| | - Karrie M. Y. Kiang
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
| | - Xiao-Qin Zhang
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
| | - Anna M. Wang
- Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong
Hong KongPR China
- Department of Electrical and Electronic Engineering, The University of Hong Kong
Hong KongPR China
| | - Ed X. Wu
- Laboratory of Biomedical Imaging and Signal Processing, The University of Hong Kong
Hong KongPR China
- Department of Electrical and Electronic Engineering, The University of Hong Kong
Hong KongPR China
| | - Wai-Man Lui
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
| | - Bai-Yun Liu
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
- Beijing Neurosurgical Institute
BeijingPR China
- Department of Neurotrauma, General Hospital of Chinese People’s Armed Police Force
BeijingPR China
| | - Gilberto K. K. Leung
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital
Hong KongPR China
| |
Collapse
|
41
|
Niemeier JP, Perrin PB, Holcomb MG, Rolston CD, Artman LK, Lu J, Nersessova KS. Gender differences in awareness and outcomes during acute traumatic brain injury recovery. J Womens Health (Larchmt) 2014; 23:573-80. [PMID: 24932911 DOI: 10.1089/jwh.2013.4535] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Recent literature on traumatic brain injury (TBI), though mixed when reporting outcomes, seems collectively to suggest possible gender advantage for women in postinjury recovery, especially in executive functions. Hormonal neuroprotection, through female reproductive hormones, is often proposed as an underlying factor in these results. We explored potential gender differences in an aspect of executive functions, self-awareness (SA), which is often impaired after TBI, limits patient effort in critical rehabilitation, and increases caregiver burden. METHODS Within a prospective survey, repeated-measures design, 121 patients with moderate or severe TBI undergoing acute rehabilitation in a Level 1 trauma center, a family member or caregiver informant, and a treating clinician were asked to complete the Patient Competency Rating Scale (PCRS) and the Frontal Systems Behavior Scale (FrSBe) at admission and discharge. RESULTS Although overall, women and men with TBI showed generally similar levels of SA, women had significantly better awareness of their injury-related deficits at acute rehabilitation discharge, even when controlling for age, education, and injury severity. CONCLUSIONS Mixed findings in this study mirror the pattern of results that dominate the published literature on gender and TBI. Gender differences in executive dysfunction may not be as large or robust as some researchers argue. In addition, complex interplays of socialization, gender-role expectations, naturally occurring male and female ability differences, and differences in access to postinjury rehabilitation are understudied potential moderators.
Collapse
Affiliation(s)
- Janet P Niemeier
- 1 Department of Physical Medicine and Rehabilitation, Carolinas Rehabilitation , Charlotte, North Carolina
| | | | | | | | | | | | | |
Collapse
|
42
|
Hohl A, Ronsoni MF, Debona R, Ben J, Schwarzbold ML, Diaz AP, Thais MERDO, Linhares MN, Latini A, Prediger RD, Pizzol FD, Walz R. Role of hormonal levels on hospital mortality for male patients with severe traumatic brain injury. Brain Inj 2014; 28:1262-9. [PMID: 24841415 DOI: 10.3109/02699052.2014.915986] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
INTRODUCTION Changes in hormone blood levels during the acute phase of traumatic brain injury (TBI) have been described in the literature. The objective was to investigate the association among several hormones plasma levels in the acute phase of severe TBI and the hospital mortality rate of male patients. METHODS The independent association among plasma levels of TSH, LH, FSH, GH, free T4, cortisol, IGF-1 and total testosterone was measured 10 hours and 30 hours after severe TBI and the hospital mortality of 60 consecutive male patients was evaluated. RESULTS At least one hormonal level abnormality was demonstrated in 3.6-73.1% of patients. The multiple logistic regressions showed a trend for an independent association among hospital mortality and normal or elevated LH levels measured at 10 hours (OR = 3.7, 95% CI = 0.8-16.3, p = 0.08) and 30 hours (OR = 3.9, 95% CI = 0.9-16.7, p = 0.06). Admission with abnormal pupils and a lower Glasgow Coma Score also were independently associated with hospital mortality. CONCLUSION The hormonal changes are frequent in the acute phase of severe TBI. The hormones plasma levels, excepting the LH, are not highly consistent with the hospital mortality of male patients.
Collapse
|
43
|
Wali B, Ishrat T, Won S, Stein DG, Sayeed I. Progesterone in experimental permanent stroke: a dose-response and therapeutic time-window study. Brain 2014; 137:486-502. [PMID: 24374329 PMCID: PMC3914469 DOI: 10.1093/brain/awt319] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/10/2013] [Accepted: 09/23/2013] [Indexed: 11/14/2022] Open
Abstract
Currently, the only approved treatment for ischaemic stroke is tissue plasminogen activator, a clot-buster. This treatment can have dangerous consequences if not given within the first 4 h after stroke. Our group and others have shown progesterone to be beneficial in preclinical studies of stroke, but a progesterone dose-response and time-window study is lacking. We tested male Sprague-Dawley rats (12 months old) with permanent middle cerebral artery occlusion or sham operations on multiple measures of sensory, motor and cognitive performance. For the dose-response study, animals received intraperitoneal injections of progesterone (8, 16 or 32 mg/kg) at 1 h post-occlusion, and subcutaneous injections at 6 h and then once every 24 h for 7 days. For the time-window study, the optimal dose of progesterone was given starting at 3, 6 or 24 h post-stroke. Behavioural recovery was evaluated at repeated intervals. Rats were killed at 22 days post-stroke and brains extracted for evaluation of infarct volume. Both 8 and 16 mg/kg doses of progesterone produced attenuation of infarct volume compared with the placebo, and improved functional outcomes up to 3 weeks after stroke on locomotor activity, grip strength, sensory neglect, gait impairment, motor coordination and spatial navigation tests. In the time-window study, the progesterone group exhibited substantial neuroprotection as late as 6 h after stroke onset. Compared with placebo, progesterone showed a significant reduction in infarct size with 3- and 6-h delays. Moderate doses (8 and 16 mg/kg) of progesterone reduced infarct size and improved functional deficits in our clinically relevant model of stroke. The 8 mg/kg dose was optimal in improving motor, sensory and memory function, and this effect was observed over a large therapeutic time window. Progesterone shows promise as a potential therapeutic agent and should be examined for safety and efficacy in a clinical trial for ischaemic stroke.
Collapse
Affiliation(s)
- Bushra Wali
- Department of Emergency Medicine, Brain Research Laboratory, Emory University, Atlanta, GA, USA
| | | | | | | | | |
Collapse
|
44
|
Progesterone attenuates hemorrhagic transformation after delayed tPA treatment in an experimental model of stroke in rats: involvement of the VEGF-MMP pathway. J Cereb Blood Flow Metab 2014; 34:72-80. [PMID: 24045404 PMCID: PMC3887344 DOI: 10.1038/jcbfm.2013.163] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/19/2013] [Accepted: 08/23/2013] [Indexed: 12/30/2022]
Abstract
Tissue plasminogen activator (tPA) is the only FDA-approved treatment for acute stroke, but its use remains limited. Progesterone (PROG) has shown neuroprotection in ischemia, but before clinical testing, we must determine how it affects hemorrhagic transformation in tPA-treated ischemic rats. Male Sprague-Dawley rats underwent middle cerebral artery occlusion with reperfusion at 4.5 hours and tPA treatment at 4.5 hours, or PROG treatment intraperitoneally at 2 hours followed by subcutaneous injection at 6 hours post occlusion. Rats were killed at 24 hours and brains evaluated for cerebral hemorrhage, swelling, blood-brain barrier (BBB) permeability, and levels of matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor level (VEGF), and tight junction (TJ) proteins. We also evaluated PROG's efficacy in preventing tPA-induced impairment of transendothelial electrical resistance (TEER) and TJ proteins under hypoxia/reoxygenation in the endothelial cells. Delayed tPA treatment induced significant hemorrhagic conversion and brain swelling. Treatment with PROG plus tPA ameliorated hemorrhage, hemispheric swelling, BBB permeability, MMP-9 induction, and VEGF levels compared with controls. Progesterone treatment significantly prevented tPA-induced decrease in TEER and expression of occludin and claudin-5, and attenuated VEGF levels in culture media subjected to hypoxia. The study concluded that PROG may extend the time window for tPA administration in ischemic stroke and reduce hemorrhagic conversion.
Collapse
|
45
|
Son I, Shin HJ, Ryu JH, Kim HK, Do SH, Zuo Z. Progesterone increases the activity of glutamate transporter type 3 expressed in Xenopus oocytes. Eur J Pharmacol 2013; 715:414-9. [PMID: 23602688 DOI: 10.1016/j.ejphar.2013.03.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Revised: 03/25/2013] [Accepted: 03/28/2013] [Indexed: 11/15/2022]
Abstract
Progesterone is an important sex hormone for pregnancy and also has neuroprotective and anticonvulsant effects. It is well-known that full-term parturients become more susceptible to volatile anesthetics. Glutamate transporters are important for preventing neurotoxicity and anesthetic action in the central nervous system. We investigated the effects of progesterone on the activity of glutamate transporter type 3 (EAAT3), the major neuronal EAAT. EAAT3 was expressed in Xenopus laevis oocytes by injecting its mRNA. Oocytes were incubated with diluted progesterone for 72 h. Two-electrode voltage clamping was used to measure membrane currents before, during, and after applying 30 μML-glutamate. Progesterone (1-100 nM) significantly increased EAAT3 activity in a dose-dependent manner. Our kinetic study showed that the Vmax was increased in the progesterone group compared with that in the control group (2.7 ± 0.2 vs. 3.6 ± 0.2μC for control group vs. progesterone group; n=18-23; P<0.05), however, Km was unaltered (46.7 ± 10.2μM vs. 55.9 ± 10.5μM for control group vs. progesterone group; n=18-23; P>0.05). Phorbol-12-myristate-13-acetate, a protein kinase C (PKC) activator, did not change progesterone-enhanced EAAT3 activity. Inhibitors of PKC or phosphatidylinositol 3-kinase (PI3K) abolished the progesterone-induced increases in EAAT3 activity. Our results suggest that progesterone enhances EAAT3 activity and that PKC and PI3K are involved in mediating these effects. These effects of progesterone may contribute to its anticonvulsant and anesthesia-related properties.
Collapse
Affiliation(s)
- Ilsoon Son
- Department of Anesthesiology & Pain Medicine, Konkuk University School of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
46
|
Marshall SA, Kalanuria A, Markandaya M, Nyquist PA. Management of intracerebral pressure in the neurosciences critical care unit. Neurosurg Clin N Am 2013; 24:361-73. [PMID: 23809031 DOI: 10.1016/j.nec.2013.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Management of intracranial pressure in neurocritical care remains a potentially valuable target for improvements in therapy and patient outcomes. Surrogate markers of increased intracranial pressure, invasive monitors, and standard therapy, as well as promising new approaches to improve cerebral compliance are discussed, and a current review of the literature addressing this metric in neuroscience critical care is provided.
Collapse
Affiliation(s)
- Scott A Marshall
- Neurology and Critical Care, Department of Medicine, San Antonio Military Medical Center, 3551 Roger Brooke Drive, Fort Sam Houston, Texas, TX 78234, USA.
| | | | | | | |
Collapse
|
47
|
Hralová M, Angerová Y, Gueye T, Bortelová J, Svestková O, Zima T, Lippertová-Grünerová M. Long-term results of enriched environment and erythropoietin after hypobaric hypoxia in rats. Physiol Res 2013; 62:463-70. [PMID: 23590602 DOI: 10.33549/physiolres.932354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
After global cerebral hypoxia, many patients are severely disabled even after intensive neurorehabilitation. Secondary mechanisms of brain injury as a result of biochemical and physiological events occur within a period of hours to months, and provide a window of opportunity for therapeutic intervention. Erythropoietin (EPO) has been shown to be neuroprotective in the brain subjected to a variety of injuries. Fifty-nine 3-month-old male Wistar rats were randomly distributed to experimental groups with respect to the housing (enriched environment - EE, standard housing - SH), to hypoxia exposure, and to EPO treatment. An acute mountain sickness model was used as a hypobaric hypoxia simulating an altitude of 8000 m. One half of the animals received erythropoietin injections, while the others were injected saline. Spatial memory was tested in a Morris water maze (MWM). The escape latency and the path length were measured. Better spatial learning in MWM was only seen in the group that received erythropoietin together with enriched environment. EPO administration itself had no influence on spatial memory. The results were very similar for both latencies and path lengths. These results support the idea that after brain injuries, the recovery can be potentiated by EPO administration combined with neurorehabilitation.
Collapse
Affiliation(s)
- M Hralová
- Department of Rehabilitation Medicine, General Teaching Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
48
|
Gautam P, Cherbuin N, Sachdev PS, Wen W, Anstey KJ. Sex differences in cortical thickness in middle aged and early old-aged adults: Personality and Total Health Through Life study. Neuroradiology 2013; 55:697-707. [DOI: 10.1007/s00234-013-1144-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 01/21/2013] [Indexed: 11/24/2022]
|
49
|
Niemeier JP, Marwitz JH, Walker WC, Davis LC, Bushnik T, Ripley DL, Ketchum JM. Are there cognitive and neurobehavioural correlates of hormonal neuroprotection for women after TBI? Neuropsychol Rehabil 2013; 23:363-82. [PMID: 23362827 DOI: 10.1080/09602011.2012.761944] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
This study examined possible cognitive correlates of hormonal neuroprotection following traumatic brain injury (TBI) using archival neuropsychological findings for 1563 individuals undergoing acute TBI rehabilitation between 1989 and 2002. Presumed age of menopause was based on the STRAW (Stages of Reproductive Aging) staging system (Soules, 2005; Soules et al., 2001) and general linear model (GLM) analysis of performance on neuropsychological testing by participants across gender and age groups (25-34, 35-44, 45-54, and 55-64) was performed. Hypotheses were (1) women with TBI in the oldest age group would have lower scores on neuropsychological tests and functional outcome measures than women in the younger groups, and (2) men in the oldest age group would have higher scores than women of the same age group. Analyses revealed that oldest females had significantly worse Trails B and SDMT written and oral scores than the youngest females. In addition, oldest females had significantly better Trails B, Rey AVLT and SDMT written scores than the oldest males. Possible cohort exposure to hormone replacement therapy, unknown hormonal status at time of testing, and sample-specific injury characteristics may have contributed to these findings.
Collapse
Affiliation(s)
- Janet P Niemeier
- Carolinas Rehabilitation, Carolinas Healthcare System, Charlotte, NC 28203, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Ye JN, Chen XS, Su L, Liu YL, Cai QY, Zhan XL, Xu Y, Zhao SF, Yao ZX. Progesterone alleviates neural behavioral deficits and demyelination with reduced degeneration of oligodendroglial cells in cuprizone-induced mice. PLoS One 2013; 8:e54590. [PMID: 23359803 PMCID: PMC3554738 DOI: 10.1371/journal.pone.0054590] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 12/14/2012] [Indexed: 12/30/2022] Open
Abstract
Demyelination occurs widely in neurodegenerative diseases. Progesterone has neuroprotective effects, is known to reduce the clinical scores and the inflammatory response. Progesterone also promotes remyelination in experimental autoimmune encephalomyelitis and cuprizone-induced demyelinating brain. However, it still remains unclear whether progesterone can alleviate neural behavioral deficits and demyelination with degeneration of oligodendroglial cells in cuprizone-induced mice. In this study, mice were fed with 0.2% cuprizone to induce demyelination, and treated with progesterone to test its potential protective effect on neural behavioral deficits, demyelination and degeneration of oligodendroglial cells. Our results showed noticeable alleviation of neural behavioral deficits following progesterone treatment as assessed by changes in average body weight, and activity during the open field and Rota-rod tests when compared with the vehicle treated cuprizone group. Progesterone treatment alleviated demyelination as shown by Luxol fast blue staining, MBP immunohistochemical staining, and electron microscopy. There was an obvious decrease in TUNEL and Caspase-3-positive apoptotic cells, and an increase in the number of oligodendroglial cells staining positive for PDGFRα, Olig2, Sox10 and CC-1 antibody in the brains of cuprizone-induced mice after progesterone administration. These results indicate that progesterone can alleviate neural behavioral deficits and demyelination against oligodendroglial cell degeneration in cuprizone-induced mice.
Collapse
Affiliation(s)
- Jian-Ning Ye
- Department of Neurology, Xin Qiao Hospital, Third Military Medical University, Chongqing, China
| | - Xing-Shu Chen
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Le Su
- Squadron 9 of Cadet Brigade, Third Military Medical University, Chongqing, China
| | - Yun-Lai Liu
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Qi-Yan Cai
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Xiao-Li Zhan
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Yan Xu
- Department of Physiology, Third Military Medical University, Chongqing, China
| | - Shi-Fu Zhao
- Department of Neurology, Xin Qiao Hospital, Third Military Medical University, Chongqing, China
- * E-mail: (Z-XY) (SZ); (S-FZ) (ZY)
| | - Zhong-Xiang Yao
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
- Department of Physiology, Third Military Medical University, Chongqing, China
- * E-mail: (Z-XY) (SZ); (S-FZ) (ZY)
| |
Collapse
|