1
|
Pasam T, Padhy HP, Dandekar MP. Lactobacillus Helveticus Improves Controlled Cortical Impact Injury-Generated Neurological Aberrations by Remodeling of Gut-Brain Axis Mediators. Neurochem Res 2024; 50:3. [PMID: 39541016 DOI: 10.1007/s11064-024-04251-4] [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: 06/04/2024] [Revised: 08/15/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Considerable studies augured the potential of gut microbiota-based interventions in brain injury-associated complications. Based on our earlier study results, we envisaged the sex-specific neuroprotective effect of Lactobacillus helveticus by remodeling of gut-brain axis. In this study, we investigated the effect of L. helveticus on neurological complications in a mouse model of controlled cortical impact (CCI). Adult, male and female, C57BL/6 mice underwent CCI surgery and received L. helveticus treatment for six weeks. Sensorimotor function was evaluated via neurological severity score and rotarod test. Long-term effects on anxiety-like behavior and cognition were assessed using the elevated-zero maze (EZM) and novel object recognition test (NORT). Brain perilesional area, blood, colon, and fecal samples were collected post-CCI for molecular biology analysis. CCI-operated mice displayed significant neurological impairments at 1-, 3-, 5-, and 7-days post-injury (dpi) and exhibited altered behavior in EZM and NORT compared to sham-operated mice. However, these behavioral changes were ameliorated in mice receiving L. helveticus. GFAP, Iba-1, TNF-α, and IL-1β expressions and corticotrophin-releasing hormone (CRH) levels were elevated in the perilesional cortex of CCI-operated male/female mice. These elevated biomarkers and decreased BDNF levels in both male/female mice were modified by L. helveticus treatment. Additionally, L. helveticus treatment restored altered short-chain fatty acids (SCFAs) levels in fecal samples and improved intestinal integrity but did not affect decreased plasma levels of progesterone and testosterone in CCI mice. These results indicate that L. helveticus exerts beneficial effects in the CCI mouse model by mitigating inflammation and remodeling of gut microbiota-brain mediators.
Collapse
Affiliation(s)
- Tulasi Pasam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Hara Prasad Padhy
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Manoj P Dandekar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
2
|
Gaynor JW, Moldenhauer JS, Zullo EE, Burnham NB, Gerdes M, Bernbaum JC, D’Agostino JA, Linn RL, Klepczynski B, Randazzo I, Gionet G, Choi GH, Karaj A, Russell WW, Zackai EH, Johnson MP, Gebb JS, Soni S, DeBari SE, Szwast AL, Ahrens-Nicklas RC, Drivas TG, Jacobwitz M, Licht DJ, Vossough A, Nicolson SC, Spray TL, Rychik J, Putt ME. Progesterone for Neurodevelopment in Fetuses With Congenital Heart Defects: A Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2412291. [PMID: 38805228 PMCID: PMC11134212 DOI: 10.1001/jamanetworkopen.2024.12291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/08/2024] [Indexed: 05/29/2024] Open
Abstract
Importance Neurodevelopmental outcomes for children with congenital heart defects (CHD) have improved minimally over the past 20 years. Objectives To assess the feasibility and tolerability of maternal progesterone therapy as well as the magnitude of the effect on neurodevelopment for fetuses with CHD. Design, Setting, and Participants This double-blinded individually randomized parallel-group clinical trial of vaginal natural progesterone therapy vs placebo in participants carrying fetuses with CHD was conducted between July 2014 and November 2021 at a quaternary care children's hospital. Participants included maternal-fetal dyads where the fetus had CHD identified before 28 weeks' gestational age and was likely to need surgery with cardiopulmonary bypass in the neonatal period. Exclusion criteria included a major genetic or extracardiac anomaly other than 22q11 deletion syndrome and known contraindication to progesterone. Statistical analysis was performed June 2022 to April 2024. Intervention Participants were 1:1 block-randomized to vaginal progesterone or placebo by diagnosis: hypoplastic left heart syndrome (HLHS), transposition of the great arteries (TGA), and other CHD diagnoses. Treatment was administered twice daily between 28 and up to 39 weeks' gestational age. Main Outcomes and Measures The primary outcome was the motor score of the Bayley Scales of Infant and Toddler Development-III; secondary outcomes included language and cognitive scales. Exploratory prespecified subgroups included cardiac diagnosis, fetal sex, genetic profile, and maternal fetal environment. Results The 102 enrolled fetuses primarily had HLHS (n = 52 [50.9%]) and TGA (n = 38 [37.3%]), were more frequently male (n = 67 [65.7%]), and without genetic anomalies (n = 61 [59.8%]). The mean motor score differed by 2.5 units (90% CI, -1.9 to 6.9 units; P = .34) for progesterone compared with placebo, a value not statistically different from 0. Exploratory subgroup analyses suggested treatment heterogeneity for the motor score for cardiac diagnosis (P for interaction = .03) and fetal sex (P for interaction = .04), but not genetic profile (P for interaction = .16) or maternal-fetal environment (P for interaction = .70). Conclusions and Relevance In this randomized clinical trial of maternal progesterone therapy, the overall effect was not statistically different from 0. Subgroup analyses suggest heterogeneity of the response to progesterone among CHD diagnosis and fetal sex. Trial Registration ClinicalTrials.gov Identifier: NCT02133573.
Collapse
Affiliation(s)
- J. William Gaynor
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Julie S. Moldenhauer
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Erin E. Zullo
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Nancy B. Burnham
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Marsha Gerdes
- Department of Psychology, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Judy C. Bernbaum
- Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jo Ann D’Agostino
- Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Rebecca L. Linn
- Division of Anatomic Pathology, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Brenna Klepczynski
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Isabel Randazzo
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Gabrielle Gionet
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Grace H. Choi
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Antoneta Karaj
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - William W. Russell
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Elaine H. Zackai
- Division of Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Mark P. Johnson
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Juliana S. Gebb
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Shelly Soni
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Suzanne E. DeBari
- Center for Fetal Diagnosis and Treatment, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Anita L. Szwast
- Division of Cardiology, Department of Pediatrics, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rebecca C. Ahrens-Nicklas
- Division of Genetics, Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Theodore G. Drivas
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Marin Jacobwitz
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel J. Licht
- Division of Neurology, Department of Pediatrics, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Arastoo Vossough
- Division of Radiology, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Susan C. Nicolson
- Division of Cardiac Anesthesia, Department of Anesthesia and Critical Medicine, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Thomas L. Spray
- Division of Cardiothoracic Surgery, Department of Surgery, Children’s Hospital of Philadelphia, and the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Jack Rychik
- Division of Cardiology, Department of Pediatrics, Children’s Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mary E. Putt
- Department of Biostatistics, Epidemiology, and Informatics, the Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|
3
|
Yatoo MI, Bahader GA, Beigh SA, Khan AM, James AW, Asmi MR, Shah ZA. Neuroprotection or Sex Bias: A Protective Response to Traumatic Brain Injury in the Females. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:906-916. [PMID: 37592792 DOI: 10.2174/1871527323666230817102125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023]
Abstract
Traumatic brain injury (TBI) is a major healthcare problem and a common cause of mortality and morbidity. Clinical and preclinical research suggests sex-related differences in short- and longterm outcomes following TBI; however, males have been the main focus of TBI research. Females show a protective response against TBI. Female animals in preclinical studies and women in clinical trials have shown comparatively better outcomes against mild, moderate, or severe TBI. This reflects a favorable protective nature of the females compared to the males, primarily attributed to various protective mechanisms that provide better prognosis and recovery in the females after TBI. Understanding the sex difference in the TBI pathophysiology and the underlying mechanisms remains an elusive goal. In this review, we provide insights into various mechanisms related to the anatomical, physiological, hormonal, enzymatic, inflammatory, oxidative, genetic, or mitochondrial basis that support the protective nature of females compared to males. Furthermore, we sought to outline the evidence of multiple biomarkers that are highly potential in the investigation of TBI's prognosis, pathophysiology, and treatment and which can serve as objective measures and novel targets for individualized therapeutic interventions in TBI treatment. Implementations from this review are important for the understanding of the effect of sex on TBI outcomes and possible mechanisms behind the favorable response in females. It also emphasizes the critical need to include females as a biological variable and in sufficient numbers in future TBI studies.
Collapse
Affiliation(s)
- Mohammad I Yatoo
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Ghaith A Bahader
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Shafayat A Beigh
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Adil M Khan
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Antonisamy William James
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Maleha R Asmi
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
4
|
Moschonas EH, Annas EM, Zamudio-Flores J, Jarvis JM, Lajud N, Bondi CO, Kline AE. Pediatric Traumatic Brain Injury: Models, Therapeutics, and Outcomes. ADVANCES IN NEUROBIOLOGY 2024; 42:147-163. [PMID: 39432041 DOI: 10.1007/978-3-031-69832-3_7] [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: 10/22/2024]
Abstract
Pediatric traumatic brain injury (TBI) is a significant healthcare issue, but potential treatments are absent despite robust investigation in several clinical trials. Factors attributed to clinical TBI, such as heterogeneity of injury and single-dose pharmacological treatments as well as timing of administration, may be reasons for the negative studies. Preclinical models of TBI can reduce some of the impediments by highlighting differences in injury depending on injury severity and location and by conducting dose response studies, thus providing better therapeutic targets and pharmacological profiles for clinical use. In this chapter, there were sufficient reports to make comparisons between the models in terms of pathophysiology, behavioral dysfunction, and the efficacy of therapeutic interventions. The models used to date include controlled cortical impact (CCI), weight drop, fluid percussion, and abusive head trauma. Several therapeutics were identified after CCI injury but none in the other models, which underscores the need for studies evaluating the therapies reported after CCI injury as well as novel potential approaches.
Collapse
Affiliation(s)
- Eleni H Moschonas
- Department of Physical Medicine & Rehabilitation, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ellen M Annas
- Department of Physical Medicine & Rehabilitation, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan Zamudio-Flores
- Centro de Investigación Biomédica de Michoacán - Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - Jessica M Jarvis
- Department of Physical Medicine & Rehabilitation, and Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Naima Lajud
- Instituto de Investigaciones sobre los Recursos Naturales - Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacan, Mexico
| | - Corina O Bondi
- Departments of Physical Medicine & Rehabilitation and Neurobiology, Center for Neuroscience, and Safar Center for Resuscitation Research, John G. Rangos Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anthony E Kline
- Departments of Physical Medicine & Rehabilitation, Critical Care Medicine, and Psychology, and Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, John G. Rangos Research Center, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Iannucci J, O’Neill K, Wang X, Mukherjee S, Wang J, Shapiro LA. Sex-Specific and Traumatic Brain Injury Effects on Dopamine Receptor Expression in the Hippocampus. Int J Mol Sci 2023; 24:16084. [PMID: 38003274 PMCID: PMC10671736 DOI: 10.3390/ijms242216084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/26/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Traumatic brain injury (TBI) is a major health concern. Each year, over 50 million individuals worldwide suffer from TBI, and this leads to a number of acute and chronic health issues. These include affective and cognitive impairment, as well as an increased risk of alcohol and drug use. The dopaminergic system, a key component of reward circuitry, has been linked to alcohol and other substance use disorders, and previous research indicates that TBI can induce plasticity within this system. Understanding how TBI modifies the dopaminergic system may offer insights into the heightened substance use and reward-seeking behavior following TBI. The hippocampus, a critical component of the reward circuit, is responsible for encoding and integrating the spatial and salient aspects of rewarding stimuli. This study explored TBI-related changes in neuronal D2 receptor expression within the hippocampus, examining the hypothesis that sex differences exist in both baseline hippocampal D2 receptor expression and its response to TBI. Utilizing D2-expressing tdTomato transgenic male and female mice, we implemented either a sham injury or the lateral fluid percussion injury (FPI) model of TBI and subsequently performed a region-specific quantification of D2 expression in the hippocampus. The results show that male mice exhibit higher baseline hippocampal D2 expression compared to female mice. Additionally, there was a significant interaction effect between sex and injury on the expression of D2 in the hippocampus, particularly in regions of the dentate gyrus. Furthermore, TBI led to significant reductions in hippocampal D2 expression in male mice, while female mice remained mostly unaffected. These results suggest that hippocampal D2 expression varies between male and female mice, with the female dopaminergic system demonstrating less susceptibility to TBI-induced plasticity.
Collapse
Affiliation(s)
- Jaclyn Iannucci
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| | - Katherine O’Neill
- Department of Biological Science, Texas A&M University, College Station, TX 77843, USA
| | - Xuehua Wang
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| | - Sanjib Mukherjee
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| | - Jun Wang
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| | - Lee A. Shapiro
- Department of Neuroscience and Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX 77807, USA; (J.I.)
| |
Collapse
|
6
|
Craine TJ, Race NS, Kutash LA, Iouchmanov AL, Moschonas EH, O'Neil DA, Sunleaf CR, Patel A, Patel N, Grobengeiser KO, Marshall IP, Magdelinic TN, Cheng JP, Bondi CO. Milnacipran Ameliorates Executive Function Impairments following Frontal Lobe Traumatic Brain Injury in Male Rats: A Multimodal Behavioral Assessment. J Neurotrauma 2023; 40:112-124. [PMID: 35979888 PMCID: PMC10024072 DOI: 10.1089/neu.2022.0289] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Traumatic brain injuries (TBIs) affect more than 10 million patients annually worldwide, causing long-term cognitive and psychosocial impairments. Frontal lobe TBIs commonly impair executive function, but laboratory models typically focus primarily on spatial learning and declarative memory. We implemented a multi-modal approach for clinically relevant cognitive-behavioral assessments of frontal lobe function in rats with TBI and assessed treatment benefits of the serotonin-norepinephrine reuptake inhibitor, milnacipran (MLN). Two attentional set-shifting tasks (AST) evaluated cognitive flexibility via the rats' ability to locate food-based rewards by learning, unlearning, and relearning sequential rule sets with shifting salient cues. Adult male rats reached stable pre-injury operant AST (oAST) performance in 3-4 weeks, then were isoflurane-anesthetized, subjected to a unilateral frontal lobe controlled cortical impact (2.4 mm depth, 4 m/sec velocity) or Sham injury, and randomized to treatment conditions. Milnacipran (30 mg/kg/day) or vehicle (VEH; 10% ethanol in saline) was administered intraperitoneally via implanted osmotic minipumps (continuous infusions post-surgery, 60 μL/h). Rats had a 10-day recovery post-TBI/Sham before performing light/location-based oAST for 10 days and, subsequently, odor/media-based digging AST (dAST) on the last test day (26-27 days post-injury) before sacrifice. Both AST tests revealed significant deficits in TBI+VEH rats, seen as elevated total trials and errors (p < 0.05), which generally normalized in MLN-treated rats (p < 0.05). This first simultaneous dual AST assessment demonstrates oAST and dAST are sufficiently sensitive and robust to detect subtle attentional and cognitive flexibility executive impairments after frontal lobe TBI in rats. Chronic MLN administration shows promise for attenuation of post-TBI executive function deficits, thus meriting further investigation.
Collapse
Affiliation(s)
- Timothy J. Craine
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- University of Bath, Claverton Down, Bath, United Kingdom
| | - Nicholas S. Race
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Association of Academic Physiatrists Rehabilitation Medicine Scientist Training Program, Owings Mills, Maryland, USA
| | - Lindsay A. Kutash
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anna L. Iouchmanov
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Eleni H. Moschonas
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Darik A. O'Neil
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Carlson R. Sunleaf
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aarti Patel
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nima Patel
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine O. Grobengeiser
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ian P. Marshall
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Taylor N. Magdelinic
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeffrey P. Cheng
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Corina O. Bondi
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Neurobiology, and University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Children's Neuroscience Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Nasre-Nasser RG, Severo MMR, Pires GN, Hort MA, Arbo BD. Effects of Progesterone on Preclinical Animal Models of Traumatic Brain Injury: Systematic Review and Meta-analysis. Mol Neurobiol 2022; 59:6341-6362. [PMID: 35922729 DOI: 10.1007/s12035-022-02970-9] [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: 04/05/2022] [Accepted: 07/21/2022] [Indexed: 12/09/2022]
Abstract
Since the publication of two phase III clinical trials not supporting the use of progesterone in patients with traumatic brain injury (TBI), several possible explanations have been postulated, including limitations in the analysis of results from preclinical evidence. Therefore, to address this question, a systematic review and meta-analysis was performed to evaluate the effects of progesterone as a neuroprotective agent in preclinical animal models of TBI. A total of 48 studies were included for review: 29 evaluated brain edema, 21 evaluated lesion size, and 0 studies reported the survival rate. In the meta-analysis, it was found that progesterone reduced brain edema (effect size - 1.73 [- 2.02, - 1.44], p < 0.0001) and lesion volume (effect size - 0.40 [- 0.65, - 0.14], p = 0.002). Lack of details in the studies hindered the assessment of risk of bias (through the SYRCLE tool). A funnel plot asymmetry was detected, suggesting a possible publication bias. In conclusion, preclinical studies show that progesterone has an anti-edema effect in animal models of TBI, decreasing lesion volume or increasing remaining tissue. However, more studies are needed using assessing methods with lower risk of histological artifacts.
Collapse
Affiliation(s)
- Raif Gregorio Nasre-Nasser
- Programa de Pós-Graduação Em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal Do Rio Grande (FURG), Porto Alegre, Rio Grande do Sul, Brazil
| | - Maria Manoela Rezende Severo
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos 2600, Building UFRGS 21116, Room 430, Zip code, Porto Alegre - RS, 90035-003, Brazil
| | - Gabriel Natan Pires
- Departamento de Psicobiologia, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Brazilian Reproducibility Initiative in Preclinical Systematic Review and Meta-Analysis (BRISA), Rio de Janeiro, Brazil
| | - Mariana Appel Hort
- Programa de Pós-Graduação Em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal Do Rio Grande (FURG), Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruno Dutra Arbo
- Programa de Pós-Graduação Em Ciências Fisiológicas, Instituto de Ciências Biológicas, Universidade Federal Do Rio Grande (FURG), Porto Alegre, Rio Grande do Sul, Brazil.
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal Do Rio Grande Do Sul (UFRGS), Rua Ramiro Barcelos 2600, Building UFRGS 21116, Room 430, Zip code, Porto Alegre - RS, 90035-003, Brazil.
| |
Collapse
|
8
|
Minchew HM, Ferren SL, Christian SK, Hu J, Keselman P, Brooks WM, Andrews BT, Harris JL. Comparing Imaging Biomarkers of Cerebral Edema after TBI in Young Adult Male and Female Rats. Brain Res 2022; 1789:147945. [PMID: 35595066 DOI: 10.1016/j.brainres.2022.147945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 03/16/2022] [Accepted: 05/13/2022] [Indexed: 11/02/2022]
Abstract
Traumatic brain injury (TBI) is one of the leading causes of death and disability worldwide. Cerebral edema following TBI is known to play a critical role in injury severity and prognosis. In the current study we used multimodal magnetic resonance imaging (MRI) to assess cerebral edema 24 hours after unilateral contusive TBI in male and female rats. We then directly quantified brain water content in the same subjectsex vivo.We found that both males and females had similarly elevated T2 values after TBI compared with sham controls. Apparent diffusion coefficient (ADC) was more variable than T2 and did not show significant injury effects in males or females. Brain water was elevated in male TBI rats compared with sham controls, but there was no difference between female TBI and sham groups. Notably, MRI biomarkers of edema were more closely correlated with brain water in male rats; female rats did not show any relationship between brain water and T2 or ADC. These observations raise questions about the interpretation of radiological findings traditionally interpreted as edema in female TBI patients. A better understanding of sex differences and similarities in the pathophysiology of post-traumatic edema is needed to help improve patient management and the development of effective treatment strategies for men and women.
Collapse
Affiliation(s)
- Heather M Minchew
- University of Kansas School of Medicine, Kansas City, KS, United States
| | - Sadie L Ferren
- Department of Anatomy and Cell Biology, KUMC, Kansas City, KS, United States
| | - Sarah K Christian
- Department of Anatomy and Cell Biology, KUMC, Kansas City, KS, United States
| | - Jinxiang Hu
- Department of Biostatistics, KUMC, Kansas City, KS, United States
| | - Paul Keselman
- Hoglund Biomedical Imaging Center, KUMC, Kansas City, KS, United States
| | - William M Brooks
- Hoglund Biomedical Imaging Center, KUMC, Kansas City, KS, United States; Depatment of Neurology, KUMC, Kansas City, KS, United States
| | - Brian T Andrews
- Department of Otolaryngology, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Janna L Harris
- Department of Anatomy and Cell Biology, KUMC, Kansas City, KS, United States; Hoglund Biomedical Imaging Center, KUMC, Kansas City, KS, United States.
| |
Collapse
|
9
|
Lengel D, Romm ZL, Bostwick AL, Huh JW, Snyder NW, Smith G, Raghupathi R. Glucocorticoid Receptor Overexpression in the Dorsal Hippocampus Attenuates Spatial Learning and Synaptic Plasticity Deficits Following Pediatric Traumatic Brain Injury. J Neurotrauma 2022; 39:979-998. [PMID: 35293260 DOI: 10.1089/neu.2022.0012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) in children younger than 4 years old leads to long-term deficits in cognitive and learning abilities that can persist or even worsen as children age into adolescence. In this study, the role of glucocorticoid receptor (GR) function in the dorsal hippocampus (DH) in hippocampal-dependent cognitive function and synaptic plasticity were assessed following injury to the 11-day-old rat. Brain injury produced significant impairments in spatial learning and memory in the Morris water maze in male and female rats at 1-month post-injury (adolescence) which was accompanied by impairments in induction and maintenance of long-term potentiation (LTP) in the CA1 region of the DH. Brain injury resulted in a significant decrease in the expression of the glucocorticoid-inducible gene, serum- and glucocorticoid-kinase 1 (sgk1), suggestive of an impairment in GR transcriptional activity within the hippocampus. Lentiviral transfection of the human GR (hGR) in the DH improved spatial learning and memory in the Morris water maze and attenuated LTP deficits following TBI. GR overexpression in the DH was also associated with a significant increase in the mRNA expression levels of sgk1, and the glutamate receptor subunits GluA1 and GluA2 within the hippocampus. Overall, these findings support an important role of dorsal hippocampal GR function in learning and memory deficits following pediatric TBI and suggest that these effects may be related to the regulation of glutamate receptor subunit expression in the DH.
Collapse
Affiliation(s)
- Dana Lengel
- Drexel University College of Medicine, 12312, Philadelphia, Pennsylvania, United States.,Mount Sinai School of Medicine, 5925, Neuroscience, New York, New York, United States;
| | - Zoe L Romm
- Drexel University College of Medicine, 12312, Neurobiology and Anatomy, Philadelphia, Pennsylvania, United States;
| | - Anna L Bostwick
- Temple University, 6558, Microbiology and Immunology, Philadelphia, Pennsylvania, United States;
| | - Jimmy W Huh
- Childrens Hospital of Philadelphia, Anesthesiology and Critical Care, Critical Care Office-7C26, 34th Street & Civic Center Blvd., Philadelphia, Pennsylvania, United States, 19104;
| | - Nathaniel W Snyder
- Temple University, 6558, Microbiology and Immunology, Philadelphia, Pennsylvania, United States;
| | - George Smith
- Temple University, 6558, Pediatric Research Center, Philadelphia, Pennsylvania, United States;
| | - Ramesh Raghupathi
- Drexel University, 6527, Neurobiology and Anatomy, 2900 Queen Lane, Philadelphia, Philadelphia, Pennsylvania, United States, 19104-2816;
| |
Collapse
|
10
|
Wali B, Sayeed I, Stein DG, Raper J. Prophylactic progesterone prevents adverse behavioural and neurocognitive effects of neonatal anaesthesia exposure in rat. Br J Anaesth 2021; 128:301-310. [PMID: 34920856 DOI: 10.1016/j.bja.2021.10.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 10/02/2021] [Accepted: 10/28/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Evidence from animal models and human studies suggests an association between early general anaesthesia exposure and development of long-lasting neurocognitive problems including learning and memory impairments and an anxious phenotype. Because millions of children each year undergo procedures that require anaesthesia, it is important to investigate ways to protect the vulnerable developing brain. We evaluated whether progesterone treatment administered before general anaesthesia exposure could prevent long-term anaesthesia-induced neurocognitive and behavioural changes. METHODS Female and male Long-Evans rat pups were repeatedly exposed to 2 h of sevoflurane or control procedures at postnatal days 7, 10, and 13. Subcutaneous injections of progesterone or vehicle were administered immediately before general anaesthesia exposure or control procedures. Neurobehavioural and cognitive outcomes were evaluated using elevated plus maze and Morris water maze tests. RESULTS Prophylactic progesterone treatment attenuated the chemokine (C-X-C motif) ligand 1 (CXCL1) response to sevoflurane exposure. Rats given vehicle treatment with general anaesthesia exposure exhibited increased anxiety on the elevated plus maze and learning and memory impairments on the Morris water maze. However, rats treated with progesterone before general anaesthesia lacked these impairments and performed in a similar manner to controls on both tasks. CONCLUSIONS Progesterone attenuated the anaesthesia-induced, acute peripheral inflammatory response and prevented cognitive and behavioural alterations associated with early repeated general anaesthesia exposure. Importantly, our results suggest that progesterone treatments given before general anaesthesia may help to protect the developing brain.
Collapse
Affiliation(s)
- Bushra Wali
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald G Stein
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA; Neuroscience and Behavioral Biology Program, Emory College of Arts and Sciences, Atlanta, GA, USA
| | - Jessica Raper
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA; Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA.
| |
Collapse
|
11
|
Du YL, Sergeeva EG, Stein DG. Visual recovery following optic nerve crush in male and female wild-type and TRIF-deficient mice. Restor Neurol Neurosci 2021; 38:355-368. [PMID: 32986632 DOI: 10.3233/rnn-201019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is growing evidence that the TIR-domain-containing adapter-inducing interferon-β (TRIF) pathway is implicated in the modulation of neuroinflammation following injuries to the brain and retina. After exposure to injury or to excitotoxic pathogens, toll-like receptors (TLR) activate the innate immune system signaling cascade and stimulate the release of inflammatory cytokines. Inhibition of the TLR4 receptor has been shown to enhance retinal ganglion cell (RGC) survival in optic nerve crush (ONC) and in ischemic injury to other parts of the brain. OBJECTIVE Based on this evidence, we tested the hypothesis that mice with the TRIF gene knocked out (TKO) will demonstrate decreased inflammatory responses and greater functional recovery after ONC. METHODS Four experimental groups -TKO ONC (12 males and 8 females), WT ONC (10 males and 8 females), TKO sham (9 males and 5 females), and WT sham (7 males and 5 females) -were used as subjects. Visual evoked potentials (VEP) were recorded in the left and right primary visual cortices and optomotor response were assessed in all mice at 14, 30, and 80 days after ONC. GFAP and Iba-1 were used as markers for astrocytes and microglial cells respectively at 7 days after ONC, along with NF-kB to measure inflammatory effects downstream of TRIF activation; RMPBS marker was used to visualize RGC survival and GAP-43 was used as a marker of regenerating optic nerve axons at 30 days after ONC. RESULTS We found reduced inflammatory response in the retina at 7 days post-ONC, less RGC loss and greater axonal regeneration 30 days post-ONC, and better recovery of visual function 80 days post-ONC in TKO mice compared to WT mice. CONCLUSIONS Our study showed that the TRIF pathway is involved in post-ONC inflammatory response and gliosis and that deletion of TRIF induces better RGC survival and regeneration and better functional recovery in mice. Our results suggest the TRIF pathway as a potential therapeutic target for reducing the inflammatory damage caused by nervous system injury.
Collapse
Affiliation(s)
- Yimeng Lina Du
- Emory University College of Arts and Sciences, Neuroscience and Behavioral Biology Program, GA, USA
| | - Elena G Sergeeva
- Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald G Stein
- Emory University College of Arts and Sciences, Neuroscience and Behavioral Biology Program, GA, USA.,Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
12
|
Amirkhosravi L, Khaksari M, Sheibani V, Shahrokhi N, Ebrahimi MN, Amiresmaili S, Salmani N. Improved spatial memory, neurobehavioral outcomes, and neuroprotective effect after progesterone administration in ovariectomized rats with traumatic brain injury: Role of RU486 progesterone receptor antagonist. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:349-359. [PMID: 33995946 PMCID: PMC8087858 DOI: 10.22038/ijbms.2021.50973.11591] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 12/22/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVES The contribution of classic progesterone receptors (PR) in interceding the neuroprotective efficacy of progesterone (P4) on the prevention of brain edema and long-time behavioral disturbances was assessed in traumatic brain injury (TBI). MATERIALS AND METHODS Female Wistar rats were ovariectomized and apportioned into 6 groups: sham, TBI, oil, P4, vehicle, and RU486. P4 or oil was injected following TBI. The antagonist of PR (RU486) or DMSO was administered before TBI. The brain edema and destruction of the blood-brain barrier (BBB) were determined. Intracranial pressure (ICP), cerebral perfusion pressure (CPP), and beam walk (BW) task were evaluated previously and at various times post-trauma. Long-time locomotor and cognitive consequences were measured one day before and on days 3, 7, 14, and 21 after the trauma. RESULTS RU486 eliminated the inhibitory effects of P4 on brain edema and BBB leakage (P<0.05, P<0.001, respectively). RU486 inhibited the decremental effect of P4 on ICP as well as the increasing effect of P4 on CPP (P<0.001) after TBI. Also, RU486 inhibited the effect of P4 on the increase in traversal time and reduction in vestibulomotor score in the BW task (P<0.001). TBI induced motor, cognitive, and anxiety-like disorders, which lasted for 3 weeks after TBI; but, P4 prevented these cognitive and behavioral abnormalities (P<0.05), and RU486 opposed this P4 effect (P<0.001). CONCLUSION The classic progesterone receptors have neuroprotective effects and prevent long-time behavioral and memory deficiency after brain trauma.
Collapse
Affiliation(s)
- Ladan Amirkhosravi
- Neuroscience Research and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Nader Shahrokhi
- Physiology Research Centers, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Navid Ebrahimi
- Neuroscience Research and Physiology Research Centers, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Neda Salmani
- Department of Psychology, Genetic Institute, Islamic Azad University- Zarand Branch, Kerman, Iran
| |
Collapse
|
13
|
Lengel D, Sevilla C, Romm ZL, Huh JW, Raghupathi R. Stem Cell Therapy for Pediatric Traumatic Brain Injury. Front Neurol 2020; 11:601286. [PMID: 33343501 PMCID: PMC7738475 DOI: 10.3389/fneur.2020.601286] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/10/2020] [Indexed: 12/11/2022] Open
Abstract
There has been a growing interest in the potential of stem cell transplantation as therapy for pediatric brain injuries. Studies in pre-clinical models of pediatric brain injury such as Traumatic Brain Injury (TBI) and neonatal hypoxia-ischemia (HI) have contributed to our understanding of the roles of endogenous stem cells in repair processes and functional recovery following brain injury, and the effects of exogenous stem cell transplantation on recovery from brain injury. Although only a handful of studies have evaluated these effects in models of pediatric TBI, many studies have evaluated stem cell transplantation therapy in models of neonatal HI which has a considerable overlap of injury pathology with pediatric TBI. In this review, we have summarized data on the effects of stem cell treatments on histopathological and functional outcomes in models of pediatric brain injury. Importantly, we have outlined evidence supporting the potential for stem cell transplantation to mitigate pathology of pediatric TBI including neuroinflammation and white matter injury, and challenges that will need to be addressed to incorporate these therapies to improve functional outcomes following pediatric TBI.
Collapse
Affiliation(s)
- Dana Lengel
- Graduate Program in Neuroscience, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Cruz Sevilla
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Zoe L Romm
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ramesh Raghupathi
- Graduate Program in Neuroscience, Drexel University College of Medicine, Philadelphia, PA, United States.,Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
14
|
Lengel D, Huh JW, Barson JR, Raghupathi R. Progesterone treatment following traumatic brain injury in the 11-day-old rat attenuates cognitive deficits and neuronal hyperexcitability in adolescence. Exp Neurol 2020; 330:113329. [PMID: 32335121 DOI: 10.1016/j.expneurol.2020.113329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/13/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) in children younger than 4 years old results in cognitive and psychosocial deficits in adolescence and adulthood. At 4 weeks following closed head injury on postnatal day 11, male and female rats exhibited impairment in novel object recognition memory (NOR) along with an increase in open arm time in the elevated plus maze (EPM), suggestive of risk-taking behaviors. This was accompanied by an increase in intrinsic excitability and frequency of spontaneous excitatory post-synaptic currents (EPSCs), and a decrease in the frequency of spontaneous inhibitory post-synaptic currents in layer 2/3 neurons within the medial prefrontal cortex (PFC), a region that is implicated in both object recognition and risk-taking behaviors. Treatment with progesterone for the first week after brain injury improved NOR memory at the 4-week time point in both sham and brain-injured rats and additionally attenuated the injury-induced increase in the excitability of neurons and the frequency of spontaneous EPSCs. The effect of progesterone on cellular excitability changes after injury may be related to its ability to decrease the mRNA expression of the β3 subunit of the voltage-gated sodium channel and increase the expression of the neuronal excitatory amino acid transporter 3 in the medial PFC in sham- and brain-injured animals and also increase glutamic acid decarboxylase mRNA expression in sham- but not brain-injured animals. Progesterone treatment did not affect injury-induced changes in the EPM test. These results demonstrate that administration of progesterone immediately after TBI in 11-day-old rats reduces cognitive deficits in adolescence, which may be mediated by progesterone-mediated regulation of excitatory signaling mechanisms within the medial PFC.
Collapse
Affiliation(s)
- Dana Lengel
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Jessica R Barson
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Ramesh Raghupathi
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States of America.
| |
Collapse
|
15
|
Rubin TG, Lipton ML. Sex Differences in Animal Models of Traumatic Brain Injury. J Exp Neurosci 2019; 13:1179069519844020. [PMID: 31205421 PMCID: PMC6537488 DOI: 10.1177/1179069519844020] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 03/25/2019] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) is highly prevalent and there is currently no adequate treatment. Understanding the underlying mechanisms governing TBI and recovery remains an elusive goal. The heterogeneous nature of injury and individual's response to injury have made understanding risk and susceptibility to TBI of great importance. Epidemiologic studies have provided evidence of sex-dependent differences following TBI. However, preclinical models of injury have largely focused on adult male animals. Here, we review 50 studies that have investigated TBI in both sexes using animal models. Results from these studies are highly variable and model dependent, but largely show females to have a protective advantage in behavioral outcomes and pathology following TBI. Further research of both sexes using newer models that better recapitulate mild and repetitive TBI is needed to characterize the nature of sex-dependent injury and recovery, and ultimately identifies targets for enhanced recovery.
Collapse
Affiliation(s)
- Todd G Rubin
- The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, Bronx, NY, USA.,Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael L Lipton
- The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Rose F. Kennedy Center, Bronx, NY, USA.,Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx NY, USA.,Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
16
|
Dong S, Zhang Q, Kong D, Zhou C, Zhou J, Han J, Zhou Y, Jin G, Hua X, Wang J, Hua F. Gender difference in the effect of progesterone on neonatal hypoxic/ischemic brain injury in mouse. Exp Neurol 2018; 306:190-198. [DOI: 10.1016/j.expneurol.2018.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 04/29/2018] [Accepted: 05/12/2018] [Indexed: 12/27/2022]
|
17
|
Huh JW, Raghupathi R. Therapeutic strategies to target acute and long-term sequelae of pediatric traumatic brain injury. Neuropharmacology 2018; 145:153-159. [PMID: 29933010 DOI: 10.1016/j.neuropharm.2018.06.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/08/2018] [Accepted: 06/19/2018] [Indexed: 12/15/2022]
Abstract
Pediatric traumatic brain injury (TBI) remains one of the leading causes of morbidity and mortality in children. Experimental and clinical studies demonstrate that the developmental age, the type of injury (diffuse vs. focal) and sex may play important roles in the response of the developing brain to a traumatic injury. Advancements in acute neurosurgical interventions and neurocritical care have improved and led to a decrease in mortality rates over the past decades. However, survivors are left with life-long behavioral deficits underscoring the need to better define the cellular mechanisms underlying these functional changes. A better understanding of these mechanisms some of which begin in the acute post-traumatic period may likely lead to targeted treatment strategies. Key considerations in designing pre-clinical experiments to test therapeutic strategies in pediatric TBI include the use of age-appropriate and pathologically-relevant models, functional outcomes that are tested as animals age into adolescence and beyond, sex as a biological variable and the recognition that doses and dosing strategies that have been demonstrated to be effective in animal models of adult TBI may not be effective in the developing brain. This article is part of the Special Issue entitled "Novel Treatments for Traumatic Brain Injury".
Collapse
Affiliation(s)
- Jimmy W Huh
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Ramesh Raghupathi
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
18
|
Kawarai Y, Tanaka H, Kobayashi T, Shozu M. Progesterone as a Postnatal Prophylactic Agent for Encephalopathy Caused by Prenatal Hypoxic Ischemic Insult. Endocrinology 2018; 159:2264-2274. [PMID: 29648595 PMCID: PMC5946846 DOI: 10.1210/en.2018-00148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/30/2018] [Indexed: 12/24/2022]
Abstract
Brain damage caused by hypoxic ischemic insult during the perinatal period causes hypoxic ischemic encephalopathies (HIEs). Therapeutic hypothermia is indicated for HIE, but because the therapeutic burden is large for its limited therapeutic effectiveness, another strategy is needed. Progesterone (P4) plays a neuroprotective role through the actions of its metabolite, allopregnanolone (Allo), on P4 receptor, γ-aminobutyric acid type A receptors or both. We examined the therapeutic potential of P4 using a newborn rat model of HIE. Fetal rats were exposed to transient ischemic hypoxia by 30-minute bilateral uterine artery clamping on gestational day 18. After spontaneous birth, newborn pups were subcutaneously injected with P4 (0.10 or 0.01 mg), medroxyprogesterone acetate (MPA; 0.12 mg), or Allo (0.10 mg) through postnatal days (PDs) 1 to 9. Brain damage in the rats was assessed using the rotarod test at PD50. The HIE insult reduced the rats' ability in the rotarod task, which was completely reversed by P4 and Allo, but not by MPA. Histological examination revealed that the HIE insult decreased neuronal (the cortex and the hippocampal CA1 region) and oligodendroglial cell density (the corpus callosum) through PD0 to PD50. The axon fiber density and myelin sheath thickness in the corpus callosum were also reduced at PD50. The time-course study revealed that P4 restored oligodendroglial cells by PD5, which was followed by neuroprotective action of P4 that lasted long over the injection period. These results suggest that P4 protects the neonatal brain from HIE insult via restoration of oligodendroglial cells.
Collapse
Affiliation(s)
- Yoshimasa Kawarai
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hirokazu Tanaka
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Obstetrics and Gynecology, School of Medicine, International University of Health and Welfare, Narita, Japan
| | - Tatsuya Kobayashi
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Makio Shozu
- Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
- Correspondence: Makio Shozu, MD, PhD, Department of Reproductive Medicine, Graduate School of Medicine, Chiba University, Japan, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8670, Japan. E-mail:
| |
Collapse
|
19
|
Turan N, Miller BA, Heider RA, Nadeem M, Sayeed I, Stein DG, Pradilla G. Neurobehavioral testing in subarachnoid hemorrhage: A review of methods and current findings in rodents. J Cereb Blood Flow Metab 2017; 37:3461-3474. [PMID: 27677672 PMCID: PMC5669338 DOI: 10.1177/0271678x16665623] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The most important aspect of a preclinical study seeking to develop a novel therapy for neurological diseases is whether the therapy produces any clinically relevant functional recovery. For this purpose, neurobehavioral tests are commonly used to evaluate the neuroprotective efficacy of treatments in a wide array of cerebrovascular diseases and neurotrauma. Their use, however, has been limited in experimental subarachnoid hemorrhage studies. After several randomized, double-blinded, controlled clinical trials repeatedly failed to produce a benefit in functional outcome despite some improvement in angiographic vasospasm, more rigorous methods of neurobehavioral testing became critical to provide a more comprehensive evaluation of the functional efficacy of proposed treatments. While several subarachnoid hemorrhage studies have incorporated an array of neurobehavioral assays, a standardized methodology has not been agreed upon. Here, we review neurobehavioral tests for rodents and their potential application to subarachnoid hemorrhage studies. Developing a standardized neurobehavioral testing regimen in rodent studies of subarachnoid hemorrhage would allow for better comparison of results between laboratories and a better prediction of what interventions would produce functional benefits in humans.
Collapse
Affiliation(s)
- Nefize Turan
- 1 Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Brandon A Miller
- 1 Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Robert A Heider
- 1 Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Maheen Nadeem
- 1 Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Iqbal Sayeed
- 2 Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Donald G Stein
- 2 Department of Emergency Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Gustavo Pradilla
- 1 Department of Neurosurgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
20
|
Wali B, Stein DG, Sayeed I. Intralipid Vehicle Does Not Interfere with the Efficacy of Progesterone in Attenuating Edema following Traumatic Brain Injury. J Neurotrauma 2017; 34:2183-2186. [DOI: 10.1089/neu.2016.4845] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Bushra Wali
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| | - Donald G. Stein
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
21
|
Geddes RI, Hayashi K, Bongers Q, Wehber M, Anderson IM, Jansen AD, Nier C, Fares E, Farquhar G, Kapoor A, Ziegler TE, VadakkadathMeethal S, Bird IM, Atwood CS. Conjugated Linoleic Acid Administration Induces Amnesia in Male Sprague Dawley Rats and Exacerbates Recovery from Functional Deficits Induced by a Controlled Cortical Impact Injury. PLoS One 2017; 12:e0169494. [PMID: 28125600 PMCID: PMC5268708 DOI: 10.1371/journal.pone.0169494] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 12/16/2016] [Indexed: 12/05/2022] Open
Abstract
Long-chain polyunsaturated fatty acids like conjugated linoleic acids (CLA) are required for normal neural development and cognitive function and have been ascribed various beneficial functions. Recently, oral CLA also has been shown to increase testosterone (T) biosynthesis, which is known to diminish traumatic brain injury (TBI)-induced neuropathology and reduce deficits induced by stroke in adult rats. To test the impact of CLA on cognitive recovery following a TBI, 5-6 month old male Sprague Dawley rats received a focal injury (craniectomy + controlled cortical impact (CCI; n = 17)) or Sham injury (craniectomy alone; n = 12) and were injected with 25 mg/kg body weight of Clarinol® G-80 (80% CLA in safflower oil; n = 16) or saline (n = 13) every 48 h for 4 weeks. Sham surgery decreased baseline plasma progesterone (P4) by 64.2% (from 9.5 ± 3.4 ng/mL to 3.4 ± 0.5 ng/mL; p = 0.068), T by 74.6% (from 5.9 ± 1.2 ng/mL to 1.5 ± 0.3 ng/mL; p < 0.05), 11-deoxycorticosterone (11-DOC) by 37.5% (from 289.3 ± 42.0 ng/mL to 180.7 ± 3.3 ng/mL), and corticosterone by 50.8% (from 195.1 ± 22.4 ng/mL to 95.9 ± 2.2 ng/mL), by post-surgery day 1. CCI injury induced similar declines in P4, T, 11-DOC and corticosterone (58.9%, 74.6%, 39.4% and 24.6%, respectively) by post-surgery day 1. These results suggest that both Sham surgery and CCI injury induce hypogonadism and hypoadrenalism in adult male rats. CLA treatment did not reverse hypogonadism in Sham (P4: 2.5 ± 1.0 ng/mL; T: 0.9 ± 0.2 ng/mL) or CCI-injured (P4: 2.2 ± 0.9 ng/mL; T: 1.0 ± 0.2 ng/mL, p > 0.05) animals by post-injury day 29, but rapidly reversed by post-injury day 1 the hypoadrenalism in Sham (11-DOC: 372.6 ± 36.6 ng/mL; corticosterone: 202.6 ± 15.6 ng/mL) and CCI-injured (11-DOC: 384.2 ± 101.3 ng/mL; corticosterone: 234.6 ± 43.8 ng/mL) animals. In Sham surgery animals, CLA did not alter body weight, but did markedly increase latency to find the hidden Morris Water Maze platform (40.3 ± 13.0 s) compared to saline treated Sham animals (8.8 ± 1.7 s). In CCI injured animals, CLA did not alter CCI-induced body weight loss, CCI-induced cystic infarct size, or deficits in rotarod performance. However, like Sham animals, CLA injections exacerbated the latency of CCI-injured rats to find the hidden MWM platform (66.8 ± 10.6 s) compared to CCI-injured rats treated with saline (30.7 ± 5.5 s, p < 0.05). These results indicate that chronic treatment of CLA at a dose of 25 mg/kg body weight in adult male rats over 1-month 1) does not reverse craniectomy- and craniectomy + CCI-induced hypogonadism, but does reverse craniectomy- and craniectomy + CCI-induced hypoadrenalism, 2) is detrimental to medium- and long-term spatial learning and memory in craniectomized uninjured rats, 3) limits cognitive recovery following a moderate-severe CCI injury, and 4) does not alter body weight.
Collapse
Affiliation(s)
- Rastafa I. Geddes
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Kentaro Hayashi
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Quinn Bongers
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Marlyse Wehber
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Icelle M. Anderson
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Alex D. Jansen
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Chase Nier
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Emily Fares
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Gabrielle Farquhar
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Amita Kapoor
- Assay Services Unit and Institute for Clinical and Translational Research Core Laboratory, National Primate Research Center, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Toni E. Ziegler
- Assay Services Unit and Institute for Clinical and Translational Research Core Laboratory, National Primate Research Center, University of Wisconsin-Madison, Wisconsin, United States of America
| | - Sivan VadakkadathMeethal
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Ian M. Bird
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
| | - Craig S. Atwood
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Wisconsin, United States of America
- Geriatric Research, Education and Clinical Center, Veterans Administration Hospital, Madison, Wisconsin, United States of America
- School of Exercise, Biomedical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
22
|
Ma J, Huang S, Qin S, You C, Zeng Y, Cochrane Injuries Group. Progesterone for acute traumatic brain injury. Cochrane Database Syst Rev 2016; 12:CD008409. [PMID: 28005271 PMCID: PMC6463867 DOI: 10.1002/14651858.cd008409.pub4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is a leading cause of death and disability, and the identification of effective, inexpensive and widely practicable treatments for brain injury is of great public health importance worldwide. Progesterone is a naturally produced hormone that has well-defined pharmacokinetics, is widely available, inexpensive, and has steroidal, neuroactive and neurosteroidal actions in the central nervous system. It is, therefore, a potential candidate for treating TBI patients. However, uncertainty exists regarding the efficacy of this treatment. This is an update of our previous review of the same title, published in 2012. OBJECTIVES To assess the effects of progesterone on neurologic outcome, mortality and disability in patients with acute TBI. To assess the safety of progesterone in patients with acute TBI. SEARCH METHODS We updated our searches of the following databases: the Cochrane Injuries Group's Specialised Register (30 September 2016), the Cochrane Central Register of Controlled Trials (CENTRAL; Issue 9, 2016), MEDLINE (Ovid; 1950 to 30 September 2016), Embase (Ovid; 1980 to 30 September 2016), Web of Science Core Collection: Conference Proceedings Citation Index-Science (CPCI-S; 1990 to 30 September 2016); and trials registries: Clinicaltrials.gov (30 September 2016) and the World Health Organization (WHO) International Clinical Trials Registry Platform (30 September 2016). SELECTION CRITERIA We included randomised controlled trials (RCTs) of progesterone versus no progesterone (or placebo) for the treatment of people with acute TBI. DATA COLLECTION AND ANALYSIS Two review authors screened search results independently to identify potentially relevant studies for inclusion. Independently, two review authors selected trials that met the inclusion criteria from the results of the screened searches, with no disagreement. MAIN RESULTS We included five RCTs in the review, with a total of 2392 participants. We assessed one trial to be at low risk of bias; two at unclear risk of bias (in one multicentred trial the possibility of centre effects was unclear, whilst the other trial was stopped early), and two at high risk of bias, due to issues with blinding and selective reporting of outcome data.All included studies reported the effects of progesterone on mortality and disability. Low quality evidence revealed no evidence of a difference in overall mortality between the progesterone group and placebo group (RR 0.91, 95% CI 0.65 to 1.28, I² = 62%; 5 studies, 2392 participants, 2376 pooled for analysis). Using the GRADE criteria, we assessed the quality of the evidence as low, due to the substantial inconsistency across studies.There was also no evidence of a difference in disability (unfavourable outcomes as assessed by the Glasgow Outcome Score) between the progesterone group and placebo group (RR 0.98, 95% CI 0.89 to 1.06, I² = 37%; 4 studies; 2336 participants, 2260 pooled for analysis). We assessed the quality of this evidence to be moderate, due to inconsistency across studies.Data were not available for meta-analysis for the outcomes of mean intracranial pressure, blood pressure, body temperature or adverse events. However, data from three studies showed no difference in mean intracranial pressure between the groups. Data from another study showed no evidence of a difference in blood pressure or body temperature between the progesterone and placebo groups, although there was evidence that intravenous progesterone infusion increased the frequency of phlebitis (882 participants). There was no evidence of a difference in the rate of other adverse events between progesterone treatment and placebo in the other three studies that reported on adverse events. AUTHORS' CONCLUSIONS This updated review did not find evidence that progesterone could reduce mortality or disability in patients with TBI. However, concerns regarding inconsistency (heterogeneity among participants and the intervention used) across included studies reduce our confidence in these results.There is no evidence from the available data that progesterone therapy results in more adverse events than placebo, aside from evidence from a single study of an increase in phlebitis (in the case of intravascular progesterone).There were not enough data on the effects of progesterone therapy for our other outcomes of interest (intracranial pressure, blood pressure, body temperature) for us to be able to draw firm conclusions.Future trials would benefit from a more precise classification of TBI and attempts to optimise progesterone dosage and scheduling.
Collapse
Affiliation(s)
- Junpeng Ma
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | - Siqing Huang
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | - Shu Qin
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | - Chao You
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | - Yunhui Zeng
- West China Hospital, Sichuan UniversityDepartment of NeurosurgeryNo. 37, Guo Xue XiangChengduSichuanChina610041
| | | |
Collapse
|
23
|
Osier ND, Dixon CE. The Controlled Cortical Impact Model: Applications, Considerations for Researchers, and Future Directions. Front Neurol 2016; 7:134. [PMID: 27582726 PMCID: PMC4987613 DOI: 10.3389/fneur.2016.00134] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/02/2016] [Indexed: 12/26/2022] Open
Abstract
Controlled cortical impact (CCI) is a mechanical model of traumatic brain injury (TBI) that was developed nearly 30 years ago with the goal of creating a testing platform to determine the biomechanical properties of brain tissue exposed to direct mechanical deformation. Initially used to model TBIs produced by automotive crashes, the CCI model rapidly transformed into a standardized technique to study TBI mechanisms and evaluate therapies. CCI is most commonly produced using a device that rapidly accelerates a rod to impact the surgically exposed cortical dural surface. The tip of the rod can be varied in size and geometry to accommodate scalability to difference species. Typically, the rod is actuated by a pneumatic piston or electromagnetic actuator. With some limits, CCI devices can control the velocity, depth, duration, and site of impact. The CCI model produces morphologic and cerebrovascular injury responses that resemble certain aspects of human TBI. Commonly observed are graded histologic and axonal derangements, disruption of the blood-brain barrier, subdural and intra-parenchymal hematoma, edema, inflammation, and alterations in cerebral blood flow. The CCI model also produces neurobehavioral and cognitive impairments similar to those observed clinically. In contrast to other TBI models, the CCI device induces a significantly pronounced cortical contusion, but is limited in the extent to which it models the diffuse effects of TBI; a related limitation is that not all clinical TBI cases are characterized by a contusion. Another perceived limitation is that a non-clinically relevant craniotomy is performed. Biomechanically, this is irrelevant at the tissue level. However, craniotomies are not atraumatic and the effects of surgery should be controlled by including surgical sham control groups. CCI devices have also been successfully used to impact closed skulls to study mild and repetitive TBI. Future directions for CCI research surround continued refinements to the model through technical improvements in the devices (e.g., minimizing mechanical sources of variation). Like all TBI models, publications should report key injury parameters as outlined in the NIH common data elements (CDEs) for pre-clinical TBI.
Collapse
Affiliation(s)
- Nicole D. Osier
- Department of Acute and Tertiary Care, University of Pittsburgh School of Nursing, Pittsburgh, PA, USA
- Safar Center for Resuscitation Research, Pittsburgh, PA, USA
| | - C. Edward Dixon
- Safar Center for Resuscitation Research, Pittsburgh, PA, USA
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- VA Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|