1
|
Haque A, Zaman V, Drasites KP, Matzelle D, Sawant S, Vertegel A, Varma A, Banik NL. Induction of Neural Differentiation and Protection by a Novel Slow-Release Nanoparticle Estrogen Construct in a Rat Model of Spinal Cord Injury. Neurochem Res 2024; 50:41. [PMID: 39613948 PMCID: PMC11607007 DOI: 10.1007/s11064-024-04289-4] [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: 08/01/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024]
Abstract
Spinal cord injury (SCI) is a complex debilitating condition leading to permanent life-long neurological deficits. Estrogen (E2) treatment is known to be neuroprotectant in SCI. This hormone is highly pleiotropic and has been shown to decrease apoptosis, modulate calcium signaling, regulate growth factor expression, act as an anti-inflammatory, and drive angiogenesis. These beneficial effects were found in our earlier study at the low dose of 10 µg/kg E2 in rats. However, the dose remains non-physiologic, which poses a safety hurdle for clinical use. Thus, we recently devised/constructed a fast release nanoparticle (NP) estrogen embedded (FNP-E2) construct and tested a focal delivery system in a contused SCI rat model which showed protection in the short run. In the current study, we have developed a novel slow-release NP estrogen (SNP-E2) delivery system that shows sustained release of E2 in the injured spinal cord and no systemic exposure in the host. The study of E2 release and kinetics of this SNP-E2 construct in vitro and in vivo supported this claim. Delivery of E2 to the injured spinal cord via this approach reduced inflammation and gliosis, and induced microglial differentiation of M1 to M2 in rats after SCI. Analysis of spinal cord samples showed improved myelination and survival signals (AKT) as demonstrated by western blot analysis. SNP-E2 treatment also induced astrocytic differentiation into neuron-like (MAP2/NeuN) cells, supported the survival of oligodendrocyte precursor cells (OPC), and improved bladder and locomotor function in rats following SCI. These data suggest that this novel delivery strategy of SNP-E2 to the injured spinal cord may provide a safe and effective therapeutic approach to treat individuals suffering from SCI.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA.
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
| | - Kelsey P Drasites
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
| | - Sushant Sawant
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Alexey Vertegel
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Abhay Varma
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA.
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
| |
Collapse
|
2
|
Coyoy-Salgado A, Segura-Uribe J, Salgado-Ceballos H, Castillo-Mendieta T, Sánchez-Torres S, Freyermuth-Trujillo X, Orozco-Barrios C, Orozco-Suarez S, Feria-Romero I, Pinto-Almazán R, Moralí de la Brena G, Guerra-Araiza C. Evaluating Sex Steroid Hormone Neuroprotection in Spinal Cord Injury in Animal Models: Is It Promising in the Clinic? Biomedicines 2024; 12:1478. [PMID: 39062051 PMCID: PMC11274729 DOI: 10.3390/biomedicines12071478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/11/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The primary mechanism of traumatic spinal cord injury (SCI) comprises the initial mechanical trauma due to the transmission of energy to the spinal cord, subsequent deformity, and persistent compression. The secondary mechanism of injury, which involves structures that remained undamaged after the initial trauma, triggers alterations in microvascular perfusion, the liberation of free radicals and neurotransmitters, lipid peroxidation, alteration in ionic concentrations, and the consequent cell death by necrosis and apoptosis. Research in the treatment of SCI has sought to develop early therapeutic interventions that mitigate the effects of these pathophysiological mechanisms. Clinical and experimental evidence has demonstrated the therapeutic benefits of sex-steroid hormone administration after traumatic brain injury and SCI. The administration of estradiol, progesterone, and testosterone has been associated with neuroprotective effects, better neurological recovery, and decreased mortality after SCI. This review evaluated evidence supporting hormone-related neuroprotection over SCI and the possible underlying mechanisms in animal models. As neuroprotection has been associated with signaling pathways, the effects of these hormones are observed on astrocytes and microglia, modulating the inflammatory response, cerebral blood flow, and metabolism, mediating glutamate excitotoxicity, and their antioxidant effects. Based on the current evidence, it is essential to analyze the benefit of sex steroid hormone therapy in the clinical management of patients with SCI.
Collapse
Affiliation(s)
- Angélica Coyoy-Salgado
- CONAHCyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Julia Segura-Uribe
- Subdirección de Gestión de la Investigación, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City 06720, Mexico;
| | - Hermelinda Salgado-Ceballos
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Tzayaka Castillo-Mendieta
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Stephanie Sánchez-Torres
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Ximena Freyermuth-Trujillo
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Carlos Orozco-Barrios
- CONAHCyT-Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Sandra Orozco-Suarez
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Iris Feria-Romero
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico; (H.S.-C.); (T.C.-M.); (S.S.-T.); (S.O.-S.)
| | - Rodolfo Pinto-Almazán
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Mexico City 11340, Mexico
| | - Gabriela Moralí de la Brena
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Christian Guerra-Araiza
- Unidad de Investigación Médica en Farmacología, Hospital de Especialidades Dr. Bernardo Sepúlveda, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| |
Collapse
|
3
|
Gao L, Xie J, Zhang H, Zheng H, Zheng W, Pang C, Cai Y, Deng B. Neuron-specific enolase in hypertension patients with acute ischemic stroke and its value forecasting long-term functional outcomes. BMC Geriatr 2023; 23:294. [PMID: 37189072 DOI: 10.1186/s12877-023-03986-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Neuron Specific Enolase (NSE), a neuro-biochemical protein marker, may correlate with the prognosis of stroke patients. Moreover, hypertension is the most common comorbidities in patients with acute ischemic stroke (AIS), and the relationship between NSE levels and long-term functional outcomes in such an increasingly large population is unclear. The aim of the study was to investigate the relationships mentioned above and optimize the prediction models. METHODS From 2018 to 2020, 1086 admissions for AIS were grouped as hypertension and non-hypertension, while hypertension group was randomly divided into development and validation cohorts for internal validation. The severity of the stroke was staged by National Institutes of Health Stroke Scale (NIHSS) score. Stroke prognosis after 1 year of follow up was documented by modified Rankin Scale (mRS) score. RESULTS Analysis revealed the following findings:(i) Serum NSE levels increased greatly in hypertension subjects with poor functional outcomes(p = 0.046). However, there was no association in non-hypertension individuals(p = 0.386). (ii) In addition to the conventional factors (age and NIHSS score), NSE (OR:1.241, 95% CI: 1.025-1.502) and prothrombin time were significantly related to the incidence of unfavorable outcomes. (iii)Based on the above four indicators, a novel nomogram was established to predict the prognosis of stoke in hypertension patients with the c-index values of 0.8851. CONCLUSIONS Overall, high baseline NSE is associated with poor 1-year AIS outcomes in hypertension patients, suggesting NSE may be a potential prognostic and therapeutic target for stroke in hypertension patients.
Collapse
Affiliation(s)
- Lingfei Gao
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- First Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Jiali Xie
- Department of Neurology, Shanghai East Hospital, Tongji University, Shanghai, PR China
| | - Haiqin Zhang
- First Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Hangqi Zheng
- First Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Wanjun Zheng
- First Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Chunyang Pang
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- First Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Yunlei Cai
- Anyang District Hospital, Dengta Road, Beiguan District, Anyang City, Henan Province, PR China
| | - Binbin Deng
- Department of Neurology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
4
|
The neuroprotective effects of estrogen and estrogenic compounds in spinal cord injury. Neurosci Biobehav Rev 2023; 146:105074. [PMID: 36736846 DOI: 10.1016/j.neubiorev.2023.105074] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
Spinal cord injury (SCI) occurs when the spinal cord is damaged from either a traumatic event or disease. SCI is characterised by multiple injury phases that affect the transmission of sensory and motor signals and lead to temporary or long-term functional deficits. There are few treatments for SCI. Estrogens and estrogenic compounds, however, may effectively mitigate the effects of SCI and therefore represent viable treatment options. This review systematically examines the pre-clinical literature on estrogen and estrogenic compound neuroprotection after SCI. Several estrogens were examined by the included studies: estrogen, estradiol benzoate, Premarin, isopsoralen, genistein, and selective estrogen receptor modulators. Across these pharmacotherapies, we find significant evidence that estrogens indeed offer protection against myriad pathophysiological effects of SCI and lead to improvements in functional outcomes, including locomotion. A STRING functional network analysis of proteins modulated by estrogen after SCI demonstrated that estrogen simultaneously upregulates known neuroprotective pathways, such as HIF-1, and downregulates pro-inflammatory pathways, including IL-17. These findings highlight the strong therapeutic potential of estrogen and estrogenic compounds after SCI.
Collapse
|
5
|
Sungura R, Shirima G, Spitsbergen J, Mpolya E, Vianney JM. A case-control study on the driving factors of childhood brain volume loss: What pediatricians must explore. PLoS One 2022; 17:e0276433. [PMID: 36584214 PMCID: PMC9803277 DOI: 10.1371/journal.pone.0276433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 10/07/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The brain volume loss also known as brain atrophy is increasingly observed among children in the course of performing neuroimaging using CT scan and MRI brains. While severe forms of brain volume loss are frequently associated with neurocognitive changes due to effects on thought processing speed, reasoning and memory of children that eventually alter their general personality, most clinicians embark themselves in managing the neurological manifestations of brain atrophy in childhood and less is known regarding the offending factors responsible for developing pre-senile brain atrophy. It was therefore the goal of this study to explore the factors that drive the occurrence of childhood brain volume under the guidance of brain CT scan quantitative evaluation. METHODS This study was a case-control study involving 168 subjects with brain atrophy who were compared with 168 age and gender matched control subjects with normal brains on CT scan under the age of 18 years. All the children with brain CT scan were subjected to an intense review of their birth and medical history including laboratory investigation reports. RESULTS Results showed significant and influential risk factors for brain atrophy in varying trends among children including age between 14-17(OR = 1.1), male gender (OR = 1.9), birth outside facility (OR = 0.99), immaturity (OR = 1.04), malnutrition (OR = 0.97), head trauma (OR = 1.02), maternal alcoholism (OR = 1.0), antiepileptic drugs & convulsive disorders (OR = 1.0), radiation injury (OR = 1.06), space occupying lesions and ICP (OR = 1.01) and birth injury/asphyxia (OR = 1.02). CONCLUSIONS Pathological reduction of brain volume in childhood exhibits a steady trend with the increase in pediatric age, with space occupying lesions & intracranial pressure being the most profound causes of brain atrophy.
Collapse
Affiliation(s)
- Richard Sungura
- Department of Health and Biomedical Sciences, School of Life Science, Nelson Mandela- African Institution of Science and Technology, Arusha, Tanzania
- * E-mail:
| | - Gabriel Shirima
- Department of Health and Biomedical Sciences, School of Life Science, Nelson Mandela- African Institution of Science and Technology, Arusha, Tanzania
| | - John Spitsbergen
- Department of Neuroscience, Western Michigan University, Kalamazoo, MI, United States of America
| | - Emmanuel Mpolya
- Department of Health and Biomedical Sciences, School of Life Science, Nelson Mandela- African Institution of Science and Technology, Arusha, Tanzania
| | - John-Mary Vianney
- Department of Health and Biomedical Sciences, School of Life Science, Nelson Mandela- African Institution of Science and Technology, Arusha, Tanzania
| |
Collapse
|
6
|
Premarin Reduces Neurodegeneration and Promotes Improvement of Function in an Animal Model of Spinal Cord Injury. Int J Mol Sci 2022; 23:ijms23042384. [PMID: 35216504 PMCID: PMC8875481 DOI: 10.3390/ijms23042384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 01/27/2023] Open
Abstract
Spinal cord injury (SCI) causes significant mortality and morbidity. Currently, no FDA-approved pharmacotherapy is available for treating SCI. Previously, low doses of estrogen (17β-estradiol, E2) were shown to improve the post-injury outcome in a rat SCI model. However, the range of associated side effects makes advocating its therapeutic use difficult. Therefore, this study aimed at investigating the therapeutic efficacy of Premarin (PRM) in SCI. PRM is an FDA-approved E2 (10%) formulation, which is used for hormone replacement therapy with minimal risk of serious side effects. The effects of PRM on SCI were examined by magnetic resonance imaging, immunofluorescent staining, and western blot analysis in a rat model. SCI animals treated with vehicle alone, PRM, E2 receptor antagonist (ICI), or PRM + ICI were graded in a blinded way for locomotor function by using the Basso–Beattie–Bresnahan (BBB) locomotor scale. PRM treatment for 7 days decreased post-SCI lesion volume and attenuated neuronal cell death, inflammation, and axonal damage. PRM also altered the balance of pro- and anti-apoptotic proteins in favor of cell survival and improved angiogenesis and microvascular growth. Increased expression of estrogen receptors (ERs) ERα and ERβ following PRM treatment and their inhibition by ER inhibitor indicated that the neuroprotection associated with PRM treatment might be E2-receptor mediated. The attenuation of glial activation with decreased inflammation and cell death, and increased angiogenesis by PRM led to improved functional outcome as determined by the BBB locomotor scale. These results suggest that PRM treatment has significant therapeutic implications for the improvement of post-SCI outcome.
Collapse
|
7
|
Haque A, Drasites KP, Cox A, Capone M, Myatich AI, Shams R, Matzelle D, Garner DP, Bredikhin M, Shields DC, Vertegel A, Banik NL. Protective Effects of Estrogen via Nanoparticle Delivery to Attenuate Myelin Loss and Neuronal Death after Spinal Cord Injury. Neurochem Res 2021; 46:2979-2990. [PMID: 34269965 PMCID: PMC9723545 DOI: 10.1007/s11064-021-03401-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 11/24/2022]
Abstract
Spinal cord injury (SCI) is associated with devastating neurological deficits affecting more than 11,000 Americans each year. Although several therapeutic agents have been proposed and tested, no FDA-approved pharmacotherapy is available for SCI treatment. We have recently demonstrated that estrogen (E2) acts as an antioxidant and anti-inflammatory agent, attenuating gliosis in SCI. We have also demonstrated that nanoparticle-mediated focal delivery of E2 to the injured spinal cord decreases lesion size, reactive gliosis, and glial scar formation. The current study tested in vitro effects of E2 on reactive oxygen species (ROS) and calpain activity in microglia, astroglia, macrophages, and fibroblasts, which are believed to participate in the inflammatory events and glial scar formation after SCI. E2 treatment decreased ROS production and calpain activity in these glial cells, macrophages, and fibroblast cells in vitro. This study also tested the efficacy of fast- and slow-release nanoparticle-E2 constructs in a rat model of SCI. Focal delivery of E2 via nanoparticles increased tissue distribution of E2 over time, attenuated cell death, and improved myelin preservation in injured spinal cord. Specifically, the fast-release nanoparticle-E2 construct reduced the Bax/Bcl-2 ratio in injured spinal cord tissues, and the slow-release nanoparticle-E2 construct prevented gliosis and penumbral demyelination distal to the lesion site. These data suggest this novel E2 delivery strategy to the lesion site may decrease inflammation and improve functional outcomes following SCI.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
| | - Kelsey P Drasites
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
| | - April Cox
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
| | - Mollie Capone
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
| | - Ali I Myatich
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
| | - Ramsha Shams
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
| | - Dena P Garner
- Department of Health and Human Performance, The Citadel, 171 Moultrie St, Charleston, SC, 29409, USA
| | | | - Donald C Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA
| | - Alexey Vertegel
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, USA.
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
- Department of Neurosurgery and Neurology, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA.
| |
Collapse
|
8
|
Kirshblum S, Snider B, Eren F, Guest J. Characterizing Natural Recovery after Traumatic Spinal Cord Injury. J Neurotrauma 2021; 38:1267-1284. [PMID: 33339474 PMCID: PMC8080912 DOI: 10.1089/neu.2020.7473] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The predominant tool used to predict outcomes after traumatic spinal cord injury (SCI) is the International Standards for Neurological Classification of Spinal Cord Injury (ISNCSCI), in association with the American Spinal Injury Association (ASIA) Impairment Scale (AIS). These measures have evolved based on analyses of large amounts of longitudinal neurological recovery data published in numerous separate studies. This article reviews and synthesizes published data on neurological recovery from multiple sources, only utilizing data in which the sacral sparing definition was applied for determination of completeness. Conversion from a complete to incomplete injury is more common in tetraplegia than paraplegia. The majority of AIS conversion and motor recovery occurs within the first 6-9 months, with the most rapid rate of motor recovery occurring in the first three months after injury. Motor score changes, as well as recovery of motor levels, are described with the initial strength of muscles as well as the levels of the motor zone of partial preservation influencing the prognosis. Total motor recovery is greater for patients with initial AIS B than AIS A, and greater after initial AIS C than with motor complete injuries. Older age has a negative impact on neurological and functional recovery after SCI; however, the specific age (whether >50 or >65 years) and underlying reasons for this impact are unclear. Penetrating injury is more likely to lead to a classification of a neurological complete injury compared with blunt trauma and reduces the likelihood of AIS conversion at one year. There are insufficient data to support gender having a major effect on neurological recovery after SCI.
Collapse
Affiliation(s)
- Steven Kirshblum
- Kessler Institute for Rehabilitation, West Orange, New Jersy, USA
- Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Kessler Foundation, West Orange, New Jersey, USA
| | - Brittany Snider
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, Minnesota, USA
| | - Fatma Eren
- Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Kessler Foundation, West Orange, New Jersey, USA
| | - James Guest
- Neurological Surgery, Miller School of Medicine, Miami, Florida, USA
- The Miami Project to Cure Paralysis, Miami, Florida, USA
| |
Collapse
|
9
|
Cox A, Capone M, Matzelle D, Vertegel A, Bredikhin M, Varma A, Haque A, Shields DC, Banik NL. Nanoparticle-Based Estrogen Delivery to Spinal Cord Injury Site Reduces Local Parenchymal Destruction and Improves Functional Recovery. J Neurotrauma 2021; 38:342-352. [PMID: 32680442 PMCID: PMC11864116 DOI: 10.1089/neu.2020.7047] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Spinal cord injury (SCI) patients sustain significant functional impairments; this is causally related to restricted neuronal regeneration after injury. The ensuing reactive gliosis, inflammatory cascade, and glial scar formation impede axonal regrowth. Although systemic anti-inflammatory agents (steroids) have been previously administered to counteract this, no current therapeutic is approved for post-injury neuronal regeneration, in part because of related side effects. Likewise, therapeutic systemic estrogen levels exhibit neuroprotective properties, but dose-dependent side effects are prohibitive. The current study thus uses low-dose estrogen delivery to the spinal cord injury (SCI) site using an agarose gel patch embedded with estrogen-loaded nanoparticles. Compared to controls, spinal cords from rodents treated with nanoparticle site-directed estrogen demonstrated significantly decreased post-injury lesion size, reactive gliosis, and glial scar formation. However, axonal regeneration, vascular endothelial growth factor production, and glial-cell-derived neurotrophic factor levels were increased with estrogen administration. Concomitantly improved locomotor and bladder functional recovery were observed with estrogen administration after injury. Therefore, low-dose site-directed estrogen may provide a future approach for enhanced neuronal repair and functional recovery in SCI patients.
Collapse
Affiliation(s)
- April Cox
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Mollie Capone
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Alexey Vertegel
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Mikhail Bredikhin
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Abhay Varma
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Donald C. Shields
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Naren L. Banik
- Department of Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
10
|
Shields DC, Haque A, Banik NL. Neuroinflammatory responses of microglia in central nervous system trauma. J Cereb Blood Flow Metab 2020; 40:S25-S33. [PMID: 33086921 PMCID: PMC7687037 DOI: 10.1177/0271678x20965786] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Although relatively few in number compared to astrocytes and neurons, microglia demonstrate multiple, varied neuroimmunological functions in the central nervous system during normal and pathological states. After injury to the brain or spinal cord, microglia express beneficial pro- and anti-inflammatory phenotypes at various stages of recovery. However, prolonged microglial activation following injury has been linked to impaired parenchymal healing and functional restoration. The nature and magnitude of microglial response to injury relates in part to peripheral immune cell invasion, extent of tissue damage, and the local microenvironment.
Collapse
Affiliation(s)
- Donald C Shields
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Naren L Banik
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA.,Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
11
|
Rezk S, Althani A, Abd-elmaksoud A, Kassab M, Farag A, Lashen S, Cenciarelli C, Caceci T, Marei H. Effects of estrogen on Survival and Neuronal Differentiation of adult human olfactory bulb neural stem Cells Transplanted into Spinal Cord Injured Rats.. [DOI: 10.1101/571950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractIn the present study we developed an excitotoxic spinal cord injury (SCI) model using kainic acid (KA) to evaluate of the therapeutic potential of human olfactory bulb neural stem cells (h-OBNSCs) for spinal cord injury (SCI). In a previous study, we assessed the therapeutic potential of these cells for SCI; all transplanted animals showed successful engraftment. These cells differentiated predominantly as astrocytes, not motor neurons, so no improvement in motor functions was detected. In the current study we used estrogen as neuroprotective therapy before transplantation of OBNSCs to preserve some of endogenous neurons and enhance the differentiation of these cells towards neurons. The present work demonstrated that the h-GFP-OBNSCs were able to survive for more than eight weeks after sub-acute transplantation into injured spinal cord. Stereological quantification of OBNSCs showed approximately a 2.38-fold increase in the initial cell population transplanted. 40.91% of OBNSCs showed differentiation along the neuronal lineages, which was the predominant fate of these cells. 36.36% of the cells differentiated into mature astrocytes; meanwhile 22.73% of the cells differentiated into oligodendrocytes. Improvement in motor functions was also detected after cell transplantation.
Collapse
|
12
|
Namjoo Z, Mortezaee K, Joghataei MT, Moradi F, Piryaei A, Abbasi Y, Hosseini A, Majidpoor J. Targeting axonal degeneration and demyelination using combination administration of 17β‐estradiol and Schwann cells in the rat model of spinal cord injury. J Cell Biochem 2018; 119:10195-10203. [DOI: 10.1002/jcb.27361] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/26/2018] [Indexed: 12/26/2022]
Affiliation(s)
- Zeinab Namjoo
- Department of Anatomy School of Medicine, Iran University of Medical Sciences Tehran Iran
| | - Keywan Mortezaee
- Department of Anatomy School of Medicine, Kurdistan University of Medical Sciences Sanandaj Iran
| | - Mohammad T. Joghataei
- Department of Anatomy School of Medicine, Iran University of Medical Sciences Tehran Iran
- Cellular and Molecular Research Center Faculty of Medicine, Iran University of Medical Sciences Tehran Iran
| | - Fateme Moradi
- Department of Anatomy School of Medicine, Iran University of Medical Sciences Tehran Iran
- Cellular and Molecular Research Center Faculty of Medicine, Iran University of Medical Sciences Tehran Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences School of Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Yusef Abbasi
- Department of Anatomy School of Medicine, Iran University of Medical Sciences Tehran Iran
| | - Amir Hosseini
- Department of Anatomy School of Medicine, Iran University of Medical Sciences Tehran Iran
| | - Jamal Majidpoor
- Department of Anatomy School of Medicine, Iran University of Medical Sciences Tehran Iran
| |
Collapse
|
13
|
Haque A, Polcyn R, Matzelle D, Banik NL. New Insights into the Role of Neuron-Specific Enolase in Neuro-Inflammation, Neurodegeneration, and Neuroprotection. Brain Sci 2018; 8:E33. [PMID: 29463007 PMCID: PMC5836052 DOI: 10.3390/brainsci8020033] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 01/18/2023] Open
Abstract
Neurodegeneration is a complex process that leads to irreversible neuronal damage and death in spinal cord injury (SCI) and various neurodegenerative diseases, which are serious, debilitating conditions. Despite exhaustive research, the cause of neuronal damage in these degenerative disorders is not completely understood. Elevation of cell surface α-enolase activates various inflammatory pathways, including the production of pro-inflammatory cytokines, chemokines, and some growth factors that are detrimental to neuronal cells. While α-enolase is present in all neurological tissues, it can also be converted to neuron specific enolase (NSE). NSE is a glycolytic enzyme found in neuronal and neuroendocrine tissues that may play a dual role in promoting both neuroinflammation and neuroprotection in SCI and other neurodegenerative events. Elevated NSE can promote ECM degradation, inflammatory glial cell proliferation, and actin remodeling, thereby affecting migration of activated macrophages and microglia to the injury site and promoting neuronal cell death. Thus, NSE could be a reliable, quantitative, and specific marker of neuronal injury. Depending on the injury, disease, and microenvironment, NSE may also show neurotrophic function as it controls neuronal survival, differentiation, and neurite regeneration via activation of phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling pathways. This review discusses possible implications of NSE expression and activity in neuroinflammation, neurodegeneration, and neuroprotection in SCI and various neurodegenerative diseases for prognostic and therapeutic potential.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29401, USA.
| | - Rachel Polcyn
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29401, USA.
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29401, USA.
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, USA.
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29401, USA.
- Department of Neurosurgery, Medical University of South Carolina, Charleston, SC 29401, USA.
- Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC 29401, USA.
| |
Collapse
|
14
|
Adhya D, Annuario E, Lancaster MA, Price J, Baron‐Cohen S, Srivastava DP. Understanding the role of steroids in typical and atypical brain development: Advantages of using a "brain in a dish" approach. J Neuroendocrinol 2018; 30:e12547. [PMID: 29024164 PMCID: PMC5838783 DOI: 10.1111/jne.12547] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/14/2017] [Accepted: 10/03/2017] [Indexed: 01/02/2023]
Abstract
Steroids have an important role in growth, development, sexual differentiation and reproduction. All four classes of steroids, androgens, oestrogens, progestogens and glucocorticoids, have varying effects on the brain. Androgens and oestrogens are involved in the sexual differentiation of the brain, and also influence cognition. Progestogens such as progesterone and its metabolites have been shown to be involved in neuroprotection, although their protective effects are timing-dependent. Glucocorticoids are linked with stress and memory performance, also in a dose- and time-dependent manner. Importantly, dysfunction in steroid function has been implicated in the pathogenesis of disease. Moreover, regulating steroid-signalling has been suggested as potential therapeutic avenue for the treatment of a number of neurodevelopmental, psychiatric and neurodegenerative disorders. Therefore, clarifying the role of steroids in typical and atypical brain function is essential for understanding typical brain functions, as well as determining their potential use for pharmacological intervention in the atypical brain. However, the majority of studies have thus far have been conducted using animal models, with limited work using native human tissue or cells. Here, we review the effect of steroids in the typical and atypical brain, focusing on the cellular, molecular functions of these molecules determined from animal models, and the therapeutic potential as highlighted by human studies. We further discuss the promise of human-induced pluripotent stem cells, including advantages of using three-dimensional neuronal cultures (organoids) in high-throughput screens, in accelerating our understanding of the role of steroids in the typical brain, and also with respect to their therapeutic value in the understanding and treatment of the atypical brain.
Collapse
Affiliation(s)
- D. Adhya
- Department of PsychiatryAutism Research CentreUniversity of CambridgeCambridgeUK
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Laboratory of Molecular BiologyCambridgeUK
| | - E. Annuario
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | | | - J. Price
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
- National Institute for Biological Standards and ControlSouth MimmsUK
| | - S. Baron‐Cohen
- Department of PsychiatryAutism Research CentreUniversity of CambridgeCambridgeUK
| | - D. P. Srivastava
- Department of Basic and Clinical NeuroscienceMaurice Wohl Clinical Neuroscience InstituteInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
- MRC Centre for Neurodevelopmental DisordersKing's College LondonLondonUK
| |
Collapse
|
15
|
Yan S, Zhang L, Wang S, Wu T, Gong Z. Inhibition of the Ras/Raf/extracellular signal-regulated kinase 1/2 signaling pathway by compounds of natural origin for possible treatment of spinal cord injury: An in silico approach. Exp Ther Med 2018; 15:2860-2868. [PMID: 29456689 PMCID: PMC5795380 DOI: 10.3892/etm.2018.5734] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 07/27/2017] [Indexed: 01/09/2023] Open
Abstract
Spinal cord injury (SCI) is a severe disease associated with permanent neurological deficit. Recent studies in the treatment of SCI have demonstrated that the Ras/Raf/extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway serves an important role in the disease etiology, and that upregulation of this signaling pathway is associated with the development of SCI. In the present study, inhibition of Ras protein was employed in order to downregulate the Ras/Raf/ERK1/2 signaling pathway using compounds of natural origin from the Interbioscreen natural compound database. To the best of our knowledge, this is the first study using a chemical-computational approach in order to identify novel small molecule inhibitors for Ras. A database of ~50,000 compounds was selected for virtual screening, setting a free energy binding bias of −7 kcal/mol to limit the number of compounds. The subset of compounds generated by virtual screening was further limited by subjecting these to the Lipinski's rule of five parameters. A total of five shortlisted compounds were subjected to molecular docking simulation. The compounds were docked into the GTP binding site of Ras, and the inhibition of this site was examined as a promising strategy for the downregulation of Ras/Raf/ERK1/2 signaling pathway. The compounds bound to the GTP binding site through hydrogen bonds and hydrophobic interactions. The identified lead compound was then subjected to molecular dynamics simulation, and the results revealed that GLY60 in the GTP binding site of Ras protein was the optimal binding site during a 100 nsec run.
Collapse
Affiliation(s)
- Shilei Yan
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Li Zhang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Shuai Wang
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Tianhao Wu
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| | - Zhixin Gong
- Department of Orthopedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
16
|
Colón JM, González PA, Cajigas Á, Maldonado WI, Torrado AI, Santiago JM, Salgado IK, Miranda JD. Continuous tamoxifen delivery improves locomotor recovery 6h after spinal cord injury by neuronal and glial mechanisms in male rats. Exp Neurol 2018; 299:109-121. [PMID: 29037533 PMCID: PMC5723542 DOI: 10.1016/j.expneurol.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 10/03/2017] [Accepted: 10/05/2017] [Indexed: 12/13/2022]
Abstract
No treatment is available for patients with spinal cord injury (SCI). Patients often arrive to the hospital hours after SCI suggesting the need of a therapy that can be used on a clinically relevant window. Previous studies showed that Tamoxifen (TAM) treatment 24h after SCI benefits locomotor recovery in female rats. Tamoxifen exerts beneficial effects in male and female rodents but a gap of knowledge exists on: the therapeutic window of TAM, the spatio-temporal mechanisms activated and if this response is sexually dimorphic. We hypothesized that TAM will favor locomotor recovery when administered up-to 24h after SCI in male Sprague-Dawley rats. Rats received a thoracic (T10) contusion using the MACSIS impactor followed by placebo or TAM (15mg/21days) pellets in a therapeutic window of 0, 6, 12, or 24h. Animals were sacrificed at 2, 7, 14, 28 or 35days post injury (DPI) to study the molecular and cellular changes in the acute and chronic stages. Immediate or delayed therapy (t=6h) improved locomotor function, increased white matter spared tissue, and neuronal survival. TAM reduced reactive gliosis during chronic stages and increased the expression of Olig-2. A significant difference was observed in estrogen receptor alpha between male and female rodents from 2 to 28 DPI suggesting a sexually dimorphic characteristic that could be related to the behavioral differences observed in the therapeutic window of TAM. This study supports the use of TAM in the SCI setting due to its neuroprotective effects but with a significant sexually dimorphic therapeutic window.
Collapse
Affiliation(s)
- Jennifer M Colón
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Pablo A González
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Ámbar Cajigas
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Wanda I Maldonado
- University of Puerto Rico Carolina Campus, Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, Carolina, PR 00984, USA.
| | - Aranza I Torrado
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - José M Santiago
- University of Puerto Rico Carolina Campus, Neuroregeneration Division, Neuroscience Research Laboratory, Natural Sciences Department, Carolina, PR 00984, USA.
| | - Iris K Salgado
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| | - Jorge D Miranda
- University of Puerto Rico Medical Sciences Campus, Department of Physiology, San Juan, PR 00936, USA.
| |
Collapse
|
17
|
Expression of aromatase and estrogen receptors in lumbar motoneurons of mice. Neurosci Lett 2017; 653:7-11. [PMID: 28501695 DOI: 10.1016/j.neulet.2017.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/12/2022]
Abstract
Estrogen exerts protective roles in amyotrophic lateral sclerosis (ALS). However, the expression of aromatase (ARO) and estrogen receptors (ERs) in the motoneurons of spinal cord, has not yet been elucidated. By immunohistochemistry, we found that ARO and ERs were present in the ventral horn of adult mice lumbar spinal cord, and colocalized with SMI-32, a motoneuron specific marker. Within motoneurons, we observed that ARO is detected primarily in the cytoplasm, with fewer ARO in the nucleus; ERα and ERβ mainly localized in the nucleus with less in the cytoplasm; while GPR30 is located in soma and processes. In conclusion, we found that ERs and ARO are expressed in the motoneurons of lumbar spinal cord in adult mice. These findings suggest that estrogen may be useful as a promising therapeutic agent for prevention of damage and improvement of locomotor function in ALS.
Collapse
|
18
|
Danford ID, Verkuil LD, Choi DJ, Collins DW, Gudiseva HV, Uyhazi KE, Lau MK, Kanu LN, Grant GR, Chavali VRM, O'Brien JM. Characterizing the "POAGome": A bioinformatics-driven approach to primary open-angle glaucoma. Prog Retin Eye Res 2017; 58:89-114. [PMID: 28223208 PMCID: PMC5464971 DOI: 10.1016/j.preteyeres.2017.02.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/03/2017] [Accepted: 02/10/2017] [Indexed: 01/10/2023]
Abstract
Primary open-angle glaucoma (POAG) is a genetically, physiologically, and phenotypically complex neurodegenerative disorder. This study addressed the expanding collection of genes associated with POAG, referred to as the "POAGome." We used bioinformatics tools to perform an extensive, systematic literature search and compiled 542 genes with confirmed associations with POAG and its related phenotypes (normal tension glaucoma, ocular hypertension, juvenile open-angle glaucoma, and primary congenital glaucoma). The genes were classified according to their associated ocular tissues and phenotypes, and functional annotation and pathway analyses were subsequently performed. Our study reveals that no single molecular pathway can encompass the pathophysiology of POAG. The analyses suggested that inflammation and senescence may play pivotal roles in both the development and perpetuation of the retinal ganglion cell degeneration seen in POAG. The TGF-β signaling pathway was repeatedly implicated in our analyses, suggesting that it may be an important contributor to the manifestation of POAG in the anterior and posterior segments of the globe. We propose a molecular model of POAG revolving around TGF-β signaling, which incorporates the roles of inflammation and senescence in this disease. Finally, we highlight emerging molecular therapies that show promise for treating POAG.
Collapse
Affiliation(s)
- Ian D Danford
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lana D Verkuil
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daniel J Choi
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David W Collins
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Harini V Gudiseva
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katherine E Uyhazi
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Marisa K Lau
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Levi N Kanu
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gregory R Grant
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA, Penn Center for Bioinformatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Venkata R M Chavali
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Joan M O'Brien
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
19
|
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: 164] [Impact Index Per Article: 20.5] [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
|
20
|
|
21
|
Samantaray S, Das A, Matzelle DC, Yu SP, Wei L, Varma A, Ray SK, Banik NL. Administration of low dose estrogen attenuates persistent inflammation, promotes angiogenesis, and improves locomotor function following chronic spinal cord injury in rats. J Neurochem 2016; 137:604-17. [PMID: 26998684 DOI: 10.1111/jnc.13610] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/22/2016] [Accepted: 02/17/2016] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) causes loss of neurological function and, depending upon the severity of injury, may lead to paralysis. Currently, no FDA-approved pharmacotherapy is available for SCI. High-dose methylprednisolone is widely used, but this treatment is controversial. We have previously shown that low doses of estrogen reduces inflammation, attenuates cell death, and protects axon and myelin in SCI rats, but its effectiveness in recovery of function is not known. Therefore, the goal of this study was to investigate whether low doses of estrogen in post-SCI would reduce inflammation, protect cells and axons, and improve locomotor function during the chronic phase of injury. Injury (40 g.cm force) was induced at thoracic 10 in young adult male rats. Rats were treated with 10 or 100 μg 17β-estradiol (estrogen) for 7 days following SCI and compared with vehicle-treated injury and laminectomy (sham) controls. Histology (H&E staining), immunohistofluorescence, Doppler laser technique, and Western blotting were used to monitor tissue integrity, gliosis, blood flow, angiogenesis, the expression of angiogenic factors, axonal degeneration, and locomotor function (Basso, Beattie, and Bresnahan rating) following injury. To assess the progression of recovery, rats were sacrificed at 7, 14, or 42 days post injury. A reduction in glial reactivity, attenuation of axonal and myelin damage, protection of cells, increased expression of angiogenic factors and microvessel growth, and improved locomotor function were found following estrogen treatment compared with vehicle-treated SCI rats. These results suggest that treatment with a very low dose of estrogen has significant therapeutic implications for the improvement of locomotor function in chronic SCI. Experimental studies with low dose estrogen therapy in chronic spinal cord injury (SCI) demonstrated the potential for multi-active beneficial outcomes that could ameliorate the degenerative pathways in chronic SCI as shown in (a). Furthermore, the alterations in local spinal blood flow could be significantly alleviated with low dose estrogen therapy. This therapy led to the preservation of the structural integrity of the spinal cord (b), which in turn led to the improved functional recovery as shown (c).
Collapse
Affiliation(s)
- Supriti Samantaray
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Arabinda Das
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Denise C Matzelle
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shan P Yu
- Department of Anesthesia, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ling Wei
- Department of Anesthesia, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Abhay Varma
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Naren L Banik
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA.,Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
22
|
Chakrabarti M, Das A, Samantaray S, Smith JA, Banik NL, Haque A, Ray SK. Molecular mechanisms of estrogen for neuroprotection in spinal cord injury and traumatic brain injury. Rev Neurosci 2016; 27:271-81. [PMID: 26461840 DOI: 10.1515/revneuro-2015-0032] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 08/27/2015] [Indexed: 03/14/2025]
Abstract
Estrogen (EST) is a steroid hormone that exhibits several important physiological roles in the human body. During the last few decades, EST has been well recognized as an important neuroprotective agent in a variety of neurological disorders in the central nervous system (CNS), such as spinal cord injury (SCI), traumatic brain injury (TBI), Alzheimer's disease, and multiple sclerosis. The exact molecular mechanisms of EST-mediated neuroprotection in the CNS remain unclear due to heterogeneity of cell populations that express EST receptors (ERs) in the CNS as well as in the innate and adaptive immune system. Recent investigations suggest that EST protects the CNS from injury by suppressing pro-inflammatory pathways, oxidative stress, and cell death, while promoting neurogenesis, angiogenesis, and neurotrophic support. In this review, we have described the currently known molecular mechanisms of EST-mediated neuroprotection and neuroregeneration in SCI and TBI. At the same time, we have emphasized on the recent in vitro and in vivo findings from our and other laboratories, implying potential clinical benefits of EST in the treatment of SCI and TBI.
Collapse
|
23
|
Karabey-Akyurek Y, Gurcay AG, Gurcan O, Turkoglu OF, Yabanoglu-Ciftci S, Eroglu H, Sargon MF, Bilensoy E, Oner L. Localized delivery of methylprednisolone sodium succinate with polymeric nanoparticles in experimental injured spinal cord model. Pharm Dev Technol 2016; 22:972-981. [PMID: 26895158 DOI: 10.3109/10837450.2016.1143002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
With important social and economic consequences, spinal cord injuries (SCIs) still exist among major health problems. Although many therapeutic agents and methods investigated for the treatment of acute SCI, only high dose methylprednisolone (MP) is being used currently in practice. Due to the serious side effects, high dose systemic MP administration after SCI is a critical issue that is mostly considered controversial. In our study, it is aimed to develop a nanoparticle-gel combined drug delivery system for localization of MP on trauma site and eliminating dose-dependent side effects by lowering the administered dose. For this purpose, methyl prednisolone sodium succinate (MPSS) loaded polycaprolactone based nanoparticles were developed and embedded in an implantable fibrin gel. The effects of MPSS delivery system are evaluated on an acute SCI rat model, by quantification the levels of three inflammatory cytokines (interleukin-1β, interleukin-6 and caspase-3) and assessment of the damage on ultrastructural level by transmission electron microscopy. Developed NP-gel system showed very similar results with systemic high dose of MPSS. It is believed that developed system may be used as a tool for the safe and effective localized delivery of several other therapeutic molecules on injured spinal cord cases.
Collapse
Affiliation(s)
- Yasemin Karabey-Akyurek
- a Department of Pharmaceutical Technology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| | - Ahmet Gurhan Gurcay
- b Department of Neurosurgery , Ankara Ataturk Research & Education Hospital , Ankara , Turkey
| | - Oktay Gurcan
- b Department of Neurosurgery , Ankara Ataturk Research & Education Hospital , Ankara , Turkey
| | - Omer Faruk Turkoglu
- b Department of Neurosurgery , Ankara Ataturk Research & Education Hospital , Ankara , Turkey
| | - Samiye Yabanoglu-Ciftci
- c Department of Biochemistry , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey , and
| | - Hakan Eroglu
- a Department of Pharmaceutical Technology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| | - Mustafa Fevzi Sargon
- d Department of Anatomy , Faculty of Medicine, Hacettepe University , Ankara , Turkey
| | - Erem Bilensoy
- a Department of Pharmaceutical Technology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| | - Levent Oner
- a Department of Pharmaceutical Technology , Faculty of Pharmacy, Hacettepe University , Ankara , Turkey
| |
Collapse
|
24
|
Samantaray S, Das A, Matzelle DC, Yu SP, Wei L, Varma A, Ray SK, Banik NL. Administration of low dose estrogen attenuates gliosis and protects neurons in acute spinal cord injury in rats. J Neurochem 2016; 136:1064-73. [PMID: 26662641 DOI: 10.1111/jnc.13464] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 11/23/2015] [Accepted: 11/25/2015] [Indexed: 11/29/2022]
Abstract
Spinal cord injury (SCI) is a debilitating condition with neurological deficits and loss of motor function that, depending on the severity, may lead to paralysis. The only treatment currently available is methylprednisolone, which is widely used and renders limited efficacy in SCI. Therefore, other therapeutic agents must be developed. The neuroprotective efficacy of estrogen in SCI was studied with a pre-clinical and pro-translational perspective. Acute SCI was induced in rats that were treated with low doses of estrogen (1, 5, 10, or 100 μg/kg) and compared with vehicle-treated injured rats or laminectomy control (sham) rats at 48 h post-SCI. Changes in gliosis and other pro-inflammatory responses, expression and activity of proteolytic enzymes (e.g., calpain, caspase-3), apoptosis of neurons in SCI, and cell death were monitored via Western blotting and immunohistochemistry. Negligible pro-inflammatory responses or proteolytic events and very low levels of neuronal death were found in sham rats. In contrast, vehicle-treated SCI rats showed profound pro-inflammatory responses with reactive gliosis, elevated expression and activity of calpain and caspase-3, elevated Bax:Bcl-2 ratio, and high levels of neuronal death in lesion and caudal regions of the injured spinal cord. Estrogen treatment at each dose reduced pro-inflammatory and proteolytic activities and protected neurons in the caudal penumbra in acute SCI. Estrogen treatment at 10 μg was found to be as effective as 100 μg in ameliorating the above parameters in injured animals. Results from this investigation indicated that estrogen at a low dose could be a promising therapeutic agent for treating acute SCI. Experimental studies with low dose estrogen therapy in acute spinal cord injury (SCI) demonstrated the potential for multi-active beneficial outcomes. Estrogen has been found to ameliorate several degenerative pathways following SCI. Thus, such early protective effects may even lead to functional recovery in long term injury. Studies are underway in chronic SCI in a follow up manuscript.
Collapse
Affiliation(s)
- Supriti Samantaray
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Arabinda Das
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Denise C Matzelle
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Shan P Yu
- Department of Anesthesia, Emory University School of Medicine, Atlanta, GA, USA
| | - Ling Wei
- Department of Anesthesia, Emory University School of Medicine, Atlanta, GA, USA
| | - Abhay Varma
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Naren L Banik
- Department of Neurology and Neurosurgery, Medical University of South Carolina, Charleston, South Carolina, USA.,Ralph H. Johnson Veterans Administration Medical Center, Charleston, South Carolina, USA
| |
Collapse
|
25
|
Cox A, Varma A, Barry J, Vertegel A, Banik N. Nanoparticle Estrogen in Rat Spinal Cord Injury Elicits Rapid Anti-Inflammatory Effects in Plasma, Cerebrospinal Fluid, and Tissue. J Neurotrauma 2015; 32:1413-21. [PMID: 25845398 DOI: 10.1089/neu.2014.3730] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Persons with spinal cord injury (SCI) are in need of effective therapeutics. Estrogen (E2), as a steroid hormone, is a highly pleiotropic agent; with anti-inflammatory, anti-apoptotic, and neurotrophic properties, it is ideal for use in treatment of patients with SCI. Safety concerns around the use of high doses of E2 have limited clinical application, however. To address these concerns, low doses of E2 (25 μg and 2.5 μg) were focally delivered to the injured spinal cord using nanoparticles. A per-acute model (6 h after injury) was used to assess nanoparticle release of E2 into damaged spinal cord tissue; in addition, E2 was evaluated as a rapid anti-inflammatory. To assess inflammation, 27-plex cytokine/chemokine arrays were conducted in plasma, cerebrospinal fluid (CSF), and spinal cord tissue. A particular focus was placed on IL-6, GRO-KC, and MCP-1 as these have been identified from CSF in human studies as potential biomarkers in SCI. S100β, an additional proposed biomarker, was also assessed in spinal cord tissue only. Tissue concentrations of E2 were double those found in the plasma, indicating focal release. E2 showed rapid anti-inflammatory effects, significantly reducing interleukin (IL)-6, GRO-KC, MCP-1, and S100β in one or all compartments. Numerous additional targets of rapid E2 modulation were identified including: leptin, MIP-1α, IL-4, IL-2, IL-10, IFNγ, tumor necrosis factor-α, etc. These data further elucidate the rapid anti-inflammatory effects E2 exerts in an acute rat SCI model, have identified additional targets of estrogen efficacy, and suggest nanoparticle delivered estrogen may provide a safe and efficacious treatment option in persons with acute SCI.
Collapse
Affiliation(s)
- April Cox
- 1 Department of Neurology and Neurosurgery, Medical University of South Carolina , Charleston, South Carolina
| | - Abhay Varma
- 1 Department of Neurology and Neurosurgery, Medical University of South Carolina , Charleston, South Carolina
| | - John Barry
- 2 Department of Bioengineering, Clemson University , Clemson, South Carolina
| | - Alexey Vertegel
- 2 Department of Bioengineering, Clemson University , Clemson, South Carolina
| | - Naren Banik
- 1 Department of Neurology and Neurosurgery, Medical University of South Carolina , Charleston, South Carolina.,3 Ralph H. Johnson VA Medical Center , Charleston, South Carolina
| |
Collapse
|
26
|
Tang P, Zhang Y, Chen C, Ji X, Ju F, Liu X, Gan WB, He Z, Zhang S, Li W, Zhang L. In vivo two-photon imaging of axonal dieback, blood flow, and calcium influx with methylprednisolone therapy after spinal cord injury. Sci Rep 2015; 5:9691. [PMID: 25989524 PMCID: PMC4437044 DOI: 10.1038/srep09691] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/17/2015] [Indexed: 12/30/2022] Open
Abstract
Severe spinal cord injury (SCI) can cause neurological dysfunction and paralysis. However, the early dynamic changes of neurons and their surrounding environment after SCI are poorly understood. Although methylprednisolone (MP) is currently the standard therapeutic agent for treating SCI, its efficacy remains controversial. The purpose of this project was to investigate the early dynamic changes and MP's efficacy on axonal damage, blood flow, and calcium influx into axons in a mouse SCI model. YFP H-line and Thy1-GCaMP transgenic mice were used in this study. Two-photon microscopy was used for imaging of axonal dieback, blood flow, and calcium influx post-injury. We found that MP treatment attenuated progressive damage of axons, increased blood flow, and reduced calcium influx post-injury. Furthermore, microglia/macrophages accumulated in the lesion site after SCI and expressed the proinflammatory mediators iNOS, MCP-1 and IL-1β. MP treatment markedly inhibited the accumulation of microglia/macrophages and reduced the expression of the proinflammatory mediators. MP treatment also improved the recovery of behavioral function post-injury. These findings suggest that MP exerts a neuroprotective effect on SCI treatment by attenuating progressive damage of axons, increasing blood flow, reducing calcium influx, and inhibiting the accumulation of microglia/macrophages after SCI.
Collapse
Affiliation(s)
- Peifu Tang
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China, 100853
| | - Yiling Zhang
- 1] Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China, 100853 [2] Key Laboratory of Chemical Genomics, Shenzhen Graduate School, Peking University, Shenzhen, China, 518055
| | - Chao Chen
- 1] Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China, 100853 [2] Key Laboratory of Chemical Genomics, Shenzhen Graduate School, Peking University, Shenzhen, China, 518055
| | - Xinran Ji
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China, 100853
| | - Furong Ju
- School of Life Sciences, Lanzhou University, Lanzhou, China, 73000
| | - Xingyu Liu
- Beijing YouAn Hospital, Capital Medical University, Beijing, China, 100069
| | - Wen-Biao Gan
- 1] Key Laboratory of Chemical Genomics, Shenzhen Graduate School, Peking University, Shenzhen, China, 518055 [2] Skirball Institute, Department of Neuroscience and Physiology, New York University School of Medicine, New York, USA, 10016
| | - Zhigang He
- F.M. Kirby Program in Neuroscience, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts, USA, 02115
| | - Shengxiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou, China, 73000
| | - Wei Li
- Key Laboratory of Chemical Genomics, Shenzhen Graduate School, Peking University, Shenzhen, China, 518055
| | - Lihai Zhang
- Department of Orthopedics, the General Hospital of Chinese People's Liberation Army, Beijing, China, 100853
| |
Collapse
|
27
|
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
|
28
|
Cox A, Varma A, Banik N. Recent advances in the pharmacologic treatment of spinal cord injury. Metab Brain Dis 2015; 30:473-82. [PMID: 24833553 PMCID: PMC4233197 DOI: 10.1007/s11011-014-9547-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/10/2014] [Indexed: 12/16/2022]
Abstract
A need exists for the effective treatment of individuals suffering from spinal cord injury (SCI). Recent advances in the understanding of the pathophysiological mechanisms occurring in SCI have resulted in an expansion of new therapeutic targets. This review summarizes both preclinical and clinical findings investigating the mechanisms and cognate pharmacologic therapeutics targeted to modulate hypoxia, ischemia, excitotoxicity, inflammation, apoptosis, epigenetic alterations, myelin regeneration and scar remodeling. Successful modulation of these targets has been demonstrated in both preclinical and clinical studies with agents such as Oxycyte, Minocycline, Riluzole, Premarin, Cethrin, and ATI-355. The translation of these agents into clinical studies highlights the progress the field has made in the past decade. SCI proves to be a complex condition; the numerous pathophysiological mechanisms occurring at varying time points suggests that a single agent approach to the treatment of SCI may not be optimal. As the field continues to mature, the hope is that the knowledge gained from these studies will be applied to the development of an effective multi-pronged treatment strategy for SCI.
Collapse
Affiliation(s)
- April Cox
- Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas ST. MSC606, Charleston, SC, 29425, USA,
| | | | | |
Collapse
|
29
|
Shivers KY, Amador N, Abrams L, Hunter D, Jenab S, Quiñones-Jenab V. Estrogen alters baseline and inflammatory-induced cytokine levels independent from hypothalamic-pituitary-adrenal axis activity. Cytokine 2015; 72:121-9. [PMID: 25647266 DOI: 10.1016/j.cyto.2015.01.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 12/18/2014] [Accepted: 01/07/2015] [Indexed: 11/30/2022]
Abstract
Although estrogen reduces inflammatory-mediated pain responses, the mechanisms behind its effects are unclear. This study investigated if estrogen modulates inflammatory signaling by reducing baseline or inflammation-induced cytokine levels in the injury-site, serum, dorsal root ganglia (DRG) and/or spinal cord. We further tested whether estrogen effects on cytokine levels are in part mediated through hypothalamic-pituitary-adrenal (HPA) axis activation. Lumbar DRG, spinal cord, serum, and hind paw tissue were analyzed for cytokine levels in 17β-estradiol-(20%) or vehicle-(100% cholesterol) treated female rats following ovariectomy/sham adrenalectomy (OVX), adrenalectomy/sham ovariectomy (ADX) or ADX+OVX operation at baseline and post formalin injection. Formalin significantly increased pro-inflammatory interleukin (IL)-6 levels in the paw, as well as pro- and anti-inflammatory cytokine levels in the DRG, spinal cord and serum in comparison to naïve conditions. Estrogen replacement significantly increased anti-inflammatory IL-10 levels in the DRG. Centrally, estradiol significantly decreased pro-inflammatory tumor necrosis factor (TNF)-α and IL-1β levels, as well as IL-10 levels, in the spinal cord in comparison to cholesterol treatment. At both sites, most estradiol modulatory effects occurred irrespective of pain or surgical condition. Estradiol alone had no influence on cytokine release in the paw or serum, indicating that estrogen effects were site-specific. Although cytokine levels were altered between surgical conditions at baseline and following formalin administration, ADX operation did not significantly reverse estradiol's modulation of cytokine levels. These results suggest that estrogen directly regulates cytokines independent of HPA axis activity in vivo, in part by reducing cytokine levels in the spinal cord.
Collapse
Affiliation(s)
- Kai-Yvonne Shivers
- Hunter College and the Graduate Center, The City University of New York, 695 Park Avenue, New York, NY 10065, USA.
| | - Nicole Amador
- Hunter College and the Graduate Center, The City University of New York, 695 Park Avenue, New York, NY 10065, USA
| | - Lisa Abrams
- Hunter College and the Graduate Center, The City University of New York, 695 Park Avenue, New York, NY 10065, USA; Rowan University, 201 Mullica Hill Road, Glassboro, NJ 08028, USA
| | - Deirtra Hunter
- Hunter College and the Graduate Center, The City University of New York, 695 Park Avenue, New York, NY 10065, USA; Marymount Manhattan College, 221 71st Street, New York, NY 10021, USA
| | - Shirzad Jenab
- Hunter College and the Graduate Center, The City University of New York, 695 Park Avenue, New York, NY 10065, USA
| | - Vanya Quiñones-Jenab
- Hunter College and the Graduate Center, The City University of New York, 695 Park Avenue, New York, NY 10065, USA
| |
Collapse
|
30
|
Naderi A, Asgari AR, Zahed R, Ghanbari A, Samandari R, Jorjani M. Estradiol attenuates spinal cord injury-related central pain by decreasing glutamate levels in thalamic VPL nucleus in male rats. Metab Brain Dis 2014; 29:763-70. [PMID: 24879046 DOI: 10.1007/s11011-014-9570-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/20/2014] [Indexed: 12/13/2022]
Abstract
Central neuropathic pain (CNP) is a complicated medical problem that involves both the spinal and supraspinal regions of the central nervous system. Estrogen, a neuroprotective agent, has been considered a possible candidate for CNP treatment. In this study, we examined the effects of a single dose of 17β-estradiol on glutamate levels in the ventral posterolateral (VPL) nucleus of the rat thalamus. Furthermore, we determined whether there was a correlation between glutamate levels and neuropathic pain induced by unilateral electrolytic spinothalamic tract (STT) lesion. STT lesioning was performed in male Wistar rats at the T8-T9 vertebrae; rats were then administered 17β-estradiol (4 mg/kg, i.p.) 30 min after injury. Glutamate samples were collected using a microdialysis probe and quantified by high performance liquid chromatography. Mechanical allodynia (MA) and thermal hyperalgesia (TH) thresholds were measured pre-injury and 7, 14, and 28 days post-injury. We found that STT lesion significantly increased glutamate levels in the ipsilateral VPL nucleus 14 and 28 days post-injury; this was accompanied by allodynia and hyperalgesia in the hind paws of the rats. Administering 17β-estradiol to the rats decreased glutamate levels in the ipsilateral VPL nucleus and significantly increased MA and TH thresholds. These results suggest that glutamate in the VPL nucleus of the thalamus is involved in the pathology of neuropathic pain after STT injury; furthermore, 17β-estradiol may attenuate this neuropathic pain by decreasing glutamate levels.
Collapse
Affiliation(s)
- Asieh Naderi
- Department of Physiology & Biophysics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran,
| | | | | | | | | | | |
Collapse
|
31
|
Mosquera L, Colón JM, Santiago JM, Torrado AI, Meléndez M, Segarra AC, Rodríguez-Orengo JF, Miranda JD. Tamoxifen and estradiol improved locomotor function and increased spared tissue in rats after spinal cord injury: their antioxidant effect and role of estrogen receptor alpha. Brain Res 2014; 1561:11-22. [PMID: 24637260 DOI: 10.1016/j.brainres.2014.03.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 02/20/2014] [Accepted: 03/05/2014] [Indexed: 12/17/2022]
Abstract
17β-Estradiol is a multi-active steroid that imparts neuroprotection via diverse mechanisms of action. However, its role as a neuroprotective agent after spinal cord injury (SCI), or the involvement of the estrogen receptor-alpha (ER-α) in locomotor recovery, is still a subject of much debate. In this study, we evaluated the effects of estradiol and of Tamoxifen (an estrogen receptor mixed agonist/antagonist) on locomotor recovery following SCI. To control estradiol cyclical variability, ovariectomized female rats received empty or estradiol filled implants, prior to a moderate contusion to the spinal cord. Estradiol improved locomotor function at 7, 14, 21, and 28 days post injury (DPI), when compared to control groups (measured with the BBB open field test). This effect was ER-α mediated, because functional recovery was blocked with an ER-α antagonist. We also observed that ER-α was up-regulated after SCI. Long-term treatment (28 DPI) with estradiol and Tamoxifen reduced the extent of the lesion cavity, an effect also mediated by ER-α. The antioxidant effects of estradiol were seen acutely at 2 DPI but not at 28 DPI, and this acute effect was not receptor mediated. Rats treated with Tamoxifen recovered some locomotor activity at 21 and 28 DPI, which could be related to the antioxidant protection seen at these time points. These results show that estradiol improves functional outcome, and these protective effects are mediated by the ER-α dependent and independent-mechanisms. Tamoxifen׳s effects during late stages of SCI support the use of this drug as a long-term alternative treatment for this condition.
Collapse
Affiliation(s)
- Laurivette Mosquera
- Department of Physiology, University of Puerto Rico-School of Medicine, San Juan, PR 00936, USA
| | - Jennifer M Colón
- Department of Physiology, University of Puerto Rico-School of Medicine, San Juan, PR 00936, USA
| | - José M Santiago
- University of Puerto Rico Carolina Campus, Department of Natural Sciences, Carolina, PR 00984, USA
| | - Aranza I Torrado
- Department of Physiology, University of Puerto Rico-School of Medicine, San Juan, PR 00936, USA
| | | | - Annabell C Segarra
- Department of Physiology, University of Puerto Rico-School of Medicine, San Juan, PR 00936, USA
| | - José F Rodríguez-Orengo
- Department of Biochemistry, University of Puerto Rico-School of Medicine, San Juan, PR 00936, USA
| | - Jorge D Miranda
- Department of Physiology, University of Puerto Rico-School of Medicine, San Juan, PR 00936, USA.
| |
Collapse
|
32
|
Elkabes S, Nicot AB. Sex steroids and neuroprotection in spinal cord injury: a review of preclinical investigations. Exp Neurol 2014; 259:28-37. [PMID: 24440641 DOI: 10.1016/j.expneurol.2014.01.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/25/2013] [Accepted: 01/04/2014] [Indexed: 11/30/2022]
Abstract
Spinal cord injury (SCI) is a debilitating condition that affects motor, sensory and autonomic functions. Subsequent to the first mechanical trauma, secondary events, which include inflammation and glial activation, exacerbate tissue damage and worsen functional deficits. Although these secondary injury mechanisms are amenable to therapeutic interventions, the efficacy of current approaches is inadequate. Further investigations are necessary to implement new therapies that can protect neural cells and attenuate some of the detrimental effects of inflammation while promoting regeneration. Studies on different animal models of SCI indicated that sex steroids, especially 17β-estradiol and progesterone, exert neuroprotective, anti-apoptotic and anti-inflammatory effects, ameliorate tissue sparing and improve functional deficits in SCI. As sex steroid receptors are expressed in a variety of cells including neurons, glia and immune system-related cells which infiltrate the injury epicenter, sex steroids could impact multiple processes simultaneously and in doing so, influence the outcomes of SCI. However, the translation of these pre-clinical findings into the clinical setting presents challenges such as the narrow therapeutic time window of sex steroid administration, the diversity of treatment regimens that have been employed in animal studies and the lack of sufficient information regarding the persistence of the effects in chronic SCI. The current review will summarize some of the major findings in this field and will discuss the challenges associated with the implementation of sex steroids as a promising treatment in human SCI.
Collapse
Affiliation(s)
- Stella Elkabes
- The Reynolds Family Spine Laboratory, Department of Neurological Surgery, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA.
| | - Arnaud B Nicot
- UMR 1064, INSERM, Nantes, France; Faculté de Médecine, Université de Nantes, France; ITUN, CHU de Nantes, France
| |
Collapse
|
33
|
Park S, Nozaki K, Smith JA, Krause JS, Banik NL. Cross-talk between IGF-1 and estrogen receptors attenuates intracellular changes in ventral spinal cord 4.1 motoneuron cells because of interferon-gamma exposure. J Neurochem 2013; 128:904-18. [PMID: 24188094 DOI: 10.1111/jnc.12520] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 10/16/2013] [Accepted: 10/21/2013] [Indexed: 12/26/2022]
Abstract
Insulin-like growth factor-1 (IGF-1) is a neuroprotective growth factor that promotes neuronal survival by inhibition of apoptosis. To examine whether IGF-1 exerts cytoprotective effects against extracellular inflammatory stimulation, ventral spinal cord 4.1 (VSC4.1) motoneuron cells were treated with interferon-gamma (IFN-γ). Our data demonstrated apoptotic changes, increased calpain:calpastatin and Bax:Bcl-2 ratios, and expression of apoptosis-related proteases (caspase-3 and -12) in motoneurons rendered by IFN-γ in a dose-dependent manner. Post-treatment with IGF-1 attenuated these changes. In addition, IGF-1 treatment of motoneurons exposed to IFN-γ decreased expression of inflammatory markers (cyclooxygenase-2 and nuclear factor-kappa B:inhibitor of kappa B ratio). Furthermore, IGF-1 attenuated the loss of expression of IGF-1 receptors (IGF-1Rα and IGF-1Rβ) and estrogen receptors (ERα and ERβ) induced by IFN-γ. To determine whether the protective effects of IGF-1 are associated with ERs, ERs antagonist ICI and selective siRNA targeted against ERα and ERβ were used in VSC4.1 motoneurons. Distinctive morphological changes were observed following siRNA knockdown of ERα and ERβ. In particular, apoptotic cell death assessed by TUNEL assay was enhanced in both ERα and ERβ-silenced VSC4.1 motoneurons following IFN-γ and IGF-1 exposure. These results suggest that IGF-1 protects motoneurons from inflammatory insult by a mechanism involving pivotal interactions with ERα and ERβ.
Collapse
Affiliation(s)
- Sookyoung Park
- Department of Neurosciences, Division of Neurology, College of Health Professions, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | |
Collapse
|
34
|
Varma AK, Das A, Wallace G, Barry J, Vertegel AA, Ray SK, Banik NL. Spinal cord injury: a review of current therapy, future treatments, and basic science frontiers. Neurochem Res 2013; 38:895-905. [PMID: 23462880 DOI: 10.1007/s11064-013-0991-6] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 01/17/2013] [Accepted: 01/29/2013] [Indexed: 12/12/2022]
Abstract
The incidence of acute and chronic spinal cord injury (SCI) in the United States is more than 10,000 per year, resulting in 720 cases per million persons enduring permanent disability each year. The economic impact of SCI is estimated to be more than 4 billion dollars annually. Preclinical studies, case reports, and small clinical trials suggest that early treatment may improve neurological recovery. To date, no proven therapeutic modality exists that has demonstrated a positive effect on neurological outcome. Emerging data from recent preclinical and clinical studies offer hope for this devastating condition. This review gives an overview of current basic research and clinical studies for the treatment of SCI.
Collapse
Affiliation(s)
- Abhay K Varma
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Smith JA, Park S, Krause JS, Banik NL. Oxidative stress, DNA damage, and the telomeric complex as therapeutic targets in acute neurodegeneration. Neurochem Int 2013; 62:764-75. [PMID: 23422879 DOI: 10.1016/j.neuint.2013.02.013] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 02/04/2013] [Accepted: 02/08/2013] [Indexed: 01/19/2023]
Abstract
Oxidative stress has been identified as an important contributor to neurodegeneration associated with acute CNS injuries and diseases such as spinal cord injury (SCI), traumatic brain injury (TBI), and ischemic stroke. In this review, we briefly detail the damaging effects of oxidative stress (lipid peroxidation, protein oxidation, etc.) with a particular emphasis on DNA damage. Evidence for DNA damage in acute CNS injuries is presented along with its downstream effects on neuronal viability. In particular, unchecked oxidative DNA damage initiates a series of signaling events (e.g. activation of p53 and PARP-1, cell cycle re-activation) which have been shown to promote neuronal loss following CNS injury. These findings suggest that preventing DNA damage might be an effective way to promote neuronal survival and enhance neurological recovery in these conditions. Finally, we identify the telomere and telomere-associated proteins (e.g. telomerase) as novel therapeutic targets in the treatment of neurodegeneration due to their ability to modulate the neuronal response to both oxidative stress and DNA damage.
Collapse
Affiliation(s)
- Joshua A Smith
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas St., Clinical Sciences Building Room 309, Charleston, SC 29425, USA.
| | | | | | | |
Collapse
|
36
|
Saghaei E, Abbaszadeh F, Naseri K, Ghorbanpoor S, Afhami M, Haeri A, Rahimi F, Jorjani M. Estradiol attenuates spinal cord injury-induced pain by suppressing microglial activation in thalamic VPL nuclei of rats. Neurosci Res 2013; 75:316-23. [PMID: 23419864 DOI: 10.1016/j.neures.2013.01.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 01/21/2013] [Accepted: 01/29/2013] [Indexed: 12/22/2022]
Abstract
In our previous study we showed that central pain syndrome (CPS) induced by electrolytic injury caused in the unilateral spinothalamic tract (STT) is a concomitant of glial alteration at the site of injury. Here, we investigated the activity of glial cells in thalamic ventral posterolateral nuclei (VPL) and their contribution to CPS. We also examined whether post-injury administration of a pharmacological dose of estradiol can attenuate CPS and associated molecular changes. Based on the results,in the ipsilateral VPL the microglial phenotype switched o hyperactive mode and Iba1 expression was increased significantly on days 21 and 28 post-injury. The same feature was observed in contralateral VPL on day 28 (P<.05). These changes were strongly correlated with the onset of CPS (r(2)=0.670). STT injury did not induce significant astroglial response in both ipsilateral and contralateral VPL. Estradiol attenuated bilateral mechanical hypersensitivity 14 days after STT lesion (P<.05). Estradiol also suppressed microglial activation in the VPL. Taken together, these findings indicate that selective STT lesion induces bilateral microglia activation in VPL which might contribute to mechanical hypersensitivity. Furthermore, a pharmacological dose of estradiol reduces central pain possibly via suppression of glial activity in VPL region.
Collapse
Affiliation(s)
- Elham Saghaei
- Department of Pharmacology, Shahid Beheshti University of Medical Sciences, Evin, Tehran, Iran.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Chan WM, Mohammed Y, Lee I, Pearse DD. Effect of gender on recovery after spinal cord injury. Transl Stroke Res 2013; 4:447-61. [PMID: 24323341 DOI: 10.1007/s12975-012-0249-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 12/26/2012] [Indexed: 11/24/2022]
Abstract
Spinal cord injury (SCI) is a debilitating condition that affects thousands of new individuals each year, the majority of which are males. Males with SCI tend to be injured at an earlier age, mostly during sports or motor vehicle accidents, whereas females tend be injured later in life, particularly in the age group 65 and older. In both experimental and clinical studies, the question as to whether gender affects outcome has been addressed in a variety of patient groups and animal models. Results from experimental paradigms have suggested that a gender bias in outcome exists that favors females and appears to involve the advantageous or disadvantageous effects of the gonadal sex hormones estrogen and progesterone or testosterone, respectively. However, other studies have shown an absence of gender differences in outcome in specific SCI models and work has also questioned the involvement of female sex hormones in the observed outcome improvements in females. Similar controversy exists clinically, in studies that have examined gender disparities in outcome after SCI. The current review examines the experimental and clinical evidence for a gender bias in outcome following SCI and discusses issues that have made it difficult to conclusively answer this question.
Collapse
Affiliation(s)
- Wai-Man Chan
- Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | | | | | | |
Collapse
|