Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Physiol Behav ; 278: 114511, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38479582

ABSTRACT

Successive negative contrast (SNC) has been used to study reward relativity, reward loss, and frustration for decades. In instrumental SNC (iSNC), the anticipatory performance of animals downshifted from a large reward to a small reward is compared to that of animals always reinforced with the small reward. iSNC involves a transient deterioration of anticipatory behavior in downshifted animals compared to unshifted controls. There is scattered information on the optimal parameters to produce this effect and even less information about its neural basis. Five experiments with rats trained in a runway to collect food pellets explored the effects of trial distribution (massed or spaced), amount of preshift training, reward disparity, and reward magnitude on the development of an iSNC effect. Start, run, and goal latencies were measured. Using spaced trials (one trial per day), evidence of the iSNC effect was observed with 24 preshift trials and a 32-to-4 pellet disparity. With massed trials (4 trials per session separated by 30-s intertrial intervals), evidence of iSNC was found with 12 preshift sessions (a total of 48 trials) and a 16-to-2 pellet disparity. The massed-training procedure was then used to assess neural activity in three prefrontal cortex areas using c-Fos expression in animals perfused after the first downshift session. There was evidence of increased activation in the anterior cingulate cortex and a trend toward increased activation in the infralimbic and prelimbic cortices. These procedures open a venue for studying the neural basis of the instrumental behavior of animals that experience reward loss.


Subject(s)
Conditioning, Operant , Reward , Rats , Animals , Conditioning, Operant/physiology , Motivation , Prefrontal Cortex
2.
J Neuroendocrinol ; 35(1): e13228, 2023 01.
Article in English | MEDLINE | ID: mdl-36690381

ABSTRACT

Hippocampal neuropathology is a recognized feature of the spontaneously hypertensive rat (SHR). The hippocampal alterations associate with cognitive impairment. We have shown that hippocampal abnormalities are reversed by 17ß-estradiol, a steroid binding to intracellular receptors (estrogen receptor α and ß subtypes) or the membrane-located G-protein coupled estradiol receptor. Genistein (GEN) is a neuroprotective phytoestrogen which binds to estrogen receptor ß and G-protein coupled estradiol receptor. Here, we investigated whether GEN neuroprotection extends to SHR. For this purpose, we treated 5-month-old SHR for 2 weeks with 10 mg kg-1 daily s.c injections of GEN. We analyzed the expression of doublecortin+ neuronal progenitors, glial fibrillary acidic protein+ astrocytes and ionized calcium-binding adapter molecule 1+ microglia in the CA1 region and dentate gyrus of the hippocampus using immunocytochemistry, whereas a quantitative real-time polymerase chain reaction was used to measure the expression of pro- and anti-inflammatory factors tumor necrosis factor α, cyclooxygenase-2 and transforming growth factor ß. We also evaluated hippocampal dependent memory using the novel object recognition test. The results showed a decreased number of doublecortin+ neural progenitors in the dentate gyrus of SHR that was reversed with GEN. The number of glial fibrillary acidic protein+ astrocytes in the dentate gyrus and CA1 was increased in SHR but significantly decreased by GEN treatment. Additionally, GEN shifted microglial morphology from the predominantly activated phenotype present in SHR, to the more surveillance phenotype found in normotensive rats. Furthermore, treatment with GEN decreased the mRNA of the pro-inflammatory factors tumor necrosis factor α and cyclooxygenase-2 and increased the mRNA of the anti-inflammatory factor transforming growth factor ß. Discrimination index in the novel object recognition test was decreased in SHR and treatment with GEN increased this parameter. Our results indicate important neuroprotective effects of GEN at the neurochemical and behavioral level in SHR. Our data open an interesting possibility for proposing this phytoestrogen as an alternative therapy in hypertensive encephalopathy.


Subject(s)
Genistein , Phytoestrogens , Rats , Animals , Rats, Inbred SHR , Genistein/pharmacology , Phytoestrogens/pharmacology , Phytoestrogens/metabolism , Glial Fibrillary Acidic Protein/metabolism , Receptors, Estradiol/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cyclooxygenase 2/metabolism , Rats, Inbred WKY , Hippocampus/metabolism , Transforming Growth Factor beta/metabolism , Doublecortin Domain Proteins , RNA, Messenger/metabolism
3.
Neural Regen Res ; 18(2): 253-257, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35900399

ABSTRACT

Although little attention has been paid to cognitive and emotional dysfunctions observed in patients after spinal cord injury, several reports have described impairments in cognitive abilities. Our group also has contributed significantly to the study of cognitive impairments in a rat model of spinal cord injury. These findings are very significant because they demonstrate that cognitive and mood deficits are not induced by lifestyle changes, drugs of abuse, and combined medication. They are related to changes in brain structures involved in cognition and emotion, such as the hippocampus. Chronic spinal cord injury decreases neurogenesis, enhances glial reactivity leading to hippocampal neuroinflammation, and triggers cognitive deficits. These brain distal abnormalities are recently called tertiary damage. Given that there is no treatment for Tertiary Damage, insulin growth factor 1 gene therapy emerges as a good candidate. Insulin growth factor 1 gene therapy recovers neurogenesis and induces the polarization from pro-inflammatory towards anti-inflammatory microglial phenotypes, which represents a potential strategy to treat the neuroinflammation that supports tertiary damage. Insulin growth factor 1 gene therapy can be extended to other central nervous system pathologies such as traumatic brain injury where the neuroinflammatory component is crucial. Insulin growth factor 1 gene therapy could emerge as a new therapeutic strategy for treating traumatic brain injury and spinal cord injury.

4.
Cell Mol Neurobiol ; 42(1): 197-215, 2022 Jan.
Article in English | MEDLINE | ID: mdl-32537668

ABSTRACT

The hippocampus encodes spatial and contextual information involved in memory and learning. The incorporation of new neurons into hippocampal networks increases neuroplasticity and enhances hippocampal-dependent learning performances. Only few studies have described hippocampal abnormalities after spinal cord injury (SCI) although cognitive deficits related to hippocampal function have been reported in rodents and even humans. The aim of this study was to characterize in further detail hippocampal changes in the acute and chronic SCI. Our data suggested that neurogenesis reduction in the acute phase after SCI could be due to enhanced death of amplifying neural progenitors (ANPs). In addition, astrocytes became reactive and microglial cells increased their number in almost all hippocampal regions studied. Glial changes resulted in a non-inflammatory response as the mRNAs of the major pro-inflammatory cytokines (IL-1ß, TNFα, IL-18) remained unaltered, but CD200R mRNA levels were downregulated. Long-term after SCI, astrocytes remained reactive but on the other hand, microglial cell density decreased. Also, glial cells induced a neuroinflammatory environment with the upregulation of IL-1ß, TNFα and IL-18 mRNA expression and the decrease of CD200R mRNA. Neurogenesis reduction may be ascribed at later time points to inactivation of neural stem cells (NSCs) and inhibition of ANP proliferation. The number of granular cells and CA1 pyramidal neurons decreased only in the chronic phase. The release of pro-inflammatory cytokines at the chronic phase might involve neurogenesis reduction and neurodegeneration of hippocampal neurons. Therefore, SCI led to hippocampal changes that could be implicated in cognitive deficits observed in rodents and humans.


Subject(s)
Neural Stem Cells , Spinal Cord Injuries , Hippocampus/metabolism , Humans , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neuroglia/metabolism , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
5.
Mol Neurobiol ; 58(12): 6186-6202, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34463925

ABSTRACT

The hippocampus is implicated in the generation of memory and learning, processes which involve extensive neuroplasticity. The generation of hippocampal adult-born neurons is particularly regulated by glial cells of the neurogenic niche and the surrounding microenvironment. Interestingly, recent evidence has shown that spinal cord injury (SCI) in rodents leads to hippocampal neuroinflammation, neurogenesis reduction, and cognitive impairments. In this scenario, the aim of this work was to evaluate whether an adenoviral vector expressing IGF1 could reverse hippocampal alterations and cognitive deficits after chronic SCI. SCI caused neurogenesis reduction and impairments of both recognition and working memories. We also found that SCI increased the number of hypertrophic arginase-1 negative microglia concomitant with the decrease of the number of ramified surveillance microglia in the hilus, molecular layer, and subgranular zone of the dentate gyrus. RAd-IGF1 treatment restored neurogenesis and improved recognition and working memory impairments. In addition, RAd-IGF1 gene therapy modulated differentially hippocampal regions. In the hilus and molecular layer, IGF1 gene therapy recovered the number of surveillance microglia coincident with a reduction of hypertrophic microglia cell number. However, in the neurogenic niche, IGF1 reduced the number of ramified microglia and increased the number of hypertrophic microglia, which as a whole expressed arginase-1. In summary, RAd-IGF1 gene therapy might surge as a new therapeutic strategy for patients with hippocampal microglial alterations and cognitive deficits such as those with spinal cord injury and other neurodegenerative diseases.


Subject(s)
Cognition/physiology , Cognitive Dysfunction/therapy , Genetic Therapy , Hippocampus/metabolism , Insulin-Like Growth Factor I/genetics , Neurogenesis/physiology , Spinal Cord Injuries/therapy , Animals , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Male , Microglia/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/complications , Spinal Cord Injuries/metabolism
6.
eNeuro ; 8(2)2021.
Article in English | MEDLINE | ID: mdl-33593735

ABSTRACT

In many preclinical spinal cord injury (SCI) studies, assessment of locomotion recovery is key to understanding the effectiveness of the experimental intervention. In such rat SCI studies, the most basic locomotor recovery scoring system is a subjective observation of animals freely roaming in an open field, the Basso Beattie Bresnahan (BBB) score. In comparison, CatWalk is an automated gait analysis system, providing further parameter specifications. Although together the CatWalk parameters encompass gait, studies consistently report single parameters, which differ in significance from other behavioral assessments. Therefore, we believe no single parameter produced by the CatWalk can represent the fully-coordinated motion of gait. Typically, other locomotor assessments, such as the BBB score, combine several locomotor characteristics into a representative score. For this reason, we ranked the most distinctive CatWalk parameters between uninjured and SC injured rats. Subsequently, we combined nine of the topmost parameters into an SCI gait index score based on linear discriminant analysis (LDA). The resulting combination was applied to assess gait recovery in SCI experiments comprising of three thoracic contusions, a thoracic dorsal hemisection, and a cervical dorsal column lesion model. For thoracic lesions, our unbiased machine learning model revealed gait differences in lesion type and severity. In some instances, our LDA was found to be more sensitive in differentiating recovery than the BBB score alone. We believe the newly developed gait parameter combination presented here should be used in CatWalk gait recovery work with preclinical thoracic rat SCI models.


Subject(s)
Spinal Cord Injuries , Animals , Disease Models, Animal , Gait , Locomotion , Rats , Recovery of Function , Spinal Cord
7.
Steroids ; 153: 108525, 2020 01.
Article in English | MEDLINE | ID: mdl-31634489

ABSTRACT

Progesterone has been shown to exert a wide range of remarkable protective actions in experimental models of central nervous system injury or disease. However, the intimate mechanisms involved in each of these beneficial effects are not fully depicted. In this review, we intend to give the readers a thorough revision on what is known about the participation of diverse receptors and signaling pathways in progesterone-mediated neuroprotective, pro-myelinating and anti-inflammatory outcomes, as well as point out to novel regulatory mechanisms that could open new perspectives in steroid-based therapies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Central Nervous System Diseases/drug therapy , Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Receptors, Progesterone/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Central Nervous System Diseases/metabolism , Central Nervous System Diseases/pathology , Humans , Neuroprotective Agents/chemistry , Progesterone/chemistry
8.
Cell Mol Neurobiol ; 40(5): 711-723, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31784921

ABSTRACT

It is known that spontaneously hypertensive rats (SHR) present a marked encephalopathy, targeting vulnerable regions such as the hippocampus. Abnormalities of the hippocampus of SHR include decreased neurogenesis in the dentate gyrus (DG), partial loss of neurons in the hilus of the DG, micro and astrogliosis and inflammation. It is also known that 17ß-estradiol (E2) exert neuroprotective effects and prevent hippocampal abnormalities of SHR. The effects of E2 may involve a variety of mechanisms, including intracellular receptors of the ERα and ERß subtypes or membrane-located receptors, such as the G protein-coupled estradiol receptor (GPER). We have now investigated the protective role of GPER in SHR employing its synthetic agonist G1. To accomplish this objective, 5 month-old male SHR received 150 µg/day of G1 during 2 weeks. At the end of this period, we analyzed neuronal progenitors by staining for doublecortin (DCX), and counted the number of glial fibrillary acidic protein (GFAP)-labeled astrocytes and Iba1-stained microglial cells by computerized image analysis. We found that G1 activation of GPER increased DCX+ cells in the DG and reduced GFAP+ astrogliosis and Iba1+ microgliosis in the CA1 region of hippocampus. We also found that the high expression of proinflammatory makers IL1ß and cyclooxygenase 2 (COX2) of SHR was decreased after G1 treatment, which correlated with a change of microglia phenotype from the activated to a resting morphology. Additionally, G1 treatment increased the anti-inflammatory factor TGFß in SHR hippocampus. Altogether, our results suggest that activation of GPER plays a neuroprotective role on the encephalopathy of SHR, an outcome resembling E2 effects but avoiding secondary effects of the natural hormone.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Hippocampus/abnormalities , Hippocampus/pathology , Hypertensive Encephalopathy/metabolism , Inflammation/metabolism , Neurogenesis , Receptors, G-Protein-Coupled/metabolism , Animals , Astrocytes/metabolism , Doublecortin Protein , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/agonists , Estrogen Receptor beta/genetics , Glial Fibrillary Acidic Protein , Hypertensive Encephalopathy/drug therapy , Male , Microglia/metabolism , Quinolines/pharmacology , Quinolines/therapeutic use , Rats , Rats, Inbred SHR , Receptors, Estradiol/agonists , Receptors, Estradiol/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics
9.
Neural Regen Res ; 14(12): 2029-2034, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31397329

ABSTRACT

A new role has emerged for progesterone after discovering its potent actions away from reproduction in both the central and the peripheral nervous system. The aim of the present report is to discuss progesterone's mechanisms of action involved in myelination, remyelination and neuroinflammation. The pivotal role of the classic progesterone receptor is described and evidence is compiled about progesterone's direct effects on oligodendrocyte linage and its indirect effects on oligodendrocyte precursor cell differentiation by decreasing the neuroinflammatory environment.

10.
Brain Res ; 1708: 36-46, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30527678

ABSTRACT

Spinal cord lesions result in chronic demyelination as a consequence of secondary injury. Although oligodendrocyte precursor cells proliferate the differentiation program fails. Successful differentiation implies progressive decrease of transcriptional inhibitors followed by upregulation of activators. Progesterone emerges as an anti-inflammatory and pro-myelinating agent which improves locomotor outcome after spinal cord injury. In this study, we have demonstrated that spinal cord injury enhanced oligodendrocyte precursor cell number and decreased mRNA expression of transcriptional inhibitors (Id2, Id4, hes5). However, mRNA expression of transcriptional activators (Olig2, Nkx2.2, Sox10 and Mash1) was down-regulated 3 days post injury. Interestingly, a differentiation factor such as progesterone increased transcriptional activator mRNA levels and the density of Olig2- expressing oligodendrocyte precursor cells. The differentiation program is regulated by extracellular signals which modify transcriptional factors and epigenetic players. As TGFß1 is a known oligodendrocyte differentiation factor which is regulated by progesterone in reproductive tissues, we assessed whether TGFß1 could mediate progesterone remyelinating actions after the lesion. Notwithstanding that astrocyte, oligodendrocyte precursor and microglial cell density increased after spinal cord injury, the number of these cells which expressed TGFß1 remained unchanged regarding sham operated rats. However, progesterone treatment increased TGFß1 mRNA expression and the number of astrocytes and microglial TGFß1 expressing cells which would indirectly enhance oligodendrocyte differentiation. Therefore, TGFß1 arises as a potential mediator of progesterone differentiating effects on oligodendrocyte linage.


Subject(s)
Oligodendroglia/drug effects , Progesterone/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Astrocytes/metabolism , Cell Count , Cell Differentiation/drug effects , Demyelinating Diseases/metabolism , Gene Expression Regulation/drug effects , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Male , Microglia/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Nuclear Proteins , Oligodendroglia/metabolism , Progesterone/metabolism , Progesterone/physiology , Rats , Rats, Sprague-Dawley , Spinal Cord/physiology , Spinal Cord Injuries/pathology , Stem Cells/physiology , Transcription Factors/metabolism , Transforming Growth Factor beta1/metabolism
11.
Neural Regen Res ; 12(7): 1044-1047, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28852380

ABSTRACT

Only a few studies have considered changes in brain structures other than sensory and motor cortex after spinal cord injury, although cognitive impairments have been reported in these patients. Spinal cord injury results in chronic brain neuroinflammation with consequent neurodegeneration and cognitive decline in rodents. Regarding the hippocampus, neurogenesis is reduced and reactive gliosis increased. These long-term abnormalities could explain behavioral impairments exhibited in humans patients suffering from spinal cord trauma.

12.
Neurochem Res ; 42(8): 2178-2190, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28290135

ABSTRACT

The incorporation of newborn neurons with increased synaptic remodeling and activity-dependent plasticity in the dentate gyrus enhances hippocampal-dependent learning performances. Astrocytes and microglial cells are components of the neurogenic niche and regulate neurogenesis under normal and neurophatological conditions leading to functional consequences for learning and memory. Although cognitive impairments were reported in patients after spinal cord injury (SCI), only few studies have considered remote changes in brain structures which are not related with sensory and motor cortex. Thus, we examined neurogenesis and glial reactivity by stereological assessment in dentate gyrus sub-regions after three different intensities of thoracic spinal cord compression in rats. Sixty days after injury we observed a decrease in the Basso-Bresnahan-Beattie locomotor scale scores, rotarod performance and volume of spare tissue that correlated with the severity of the compression. Regarding the hippocampus, we observed that neurogenesis and hilar neurons were reduced after severe SCI, while only neurogenesis decreased in the moderately injured group. In addition, severe SCI induced reactive microglia and astrogliosis in all dentate gyrus sub-regions. Furthermore, the density of reactive microglia increased in the hilus whereas astrogliosis developed in the molecular layer after moderate SCI. No changes were observed in the mildly injured rats. These results suggest glial response and neurogenesis are associated with injury intensity. Interestingly, hippocampal neurogenesis is more sensitive to SCI than astrocytes or microglia reaction, as moderate injury impairs the generation of new neurons without changing glial response in the subgranular zone.


Subject(s)
Hippocampus/pathology , Neurogenesis/physiology , Neuroglia/physiology , Spinal Cord Injuries/pathology , Animals , Hippocampus/metabolism , Locomotion/physiology , Male , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/metabolism
13.
Neurosci Lett ; 624: 23-8, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27150077

ABSTRACT

Neuropathic pain is a frequent complication of spinal cord injury (SCI), still refractory to conventional treatment. The presence and biological activity of steroidogenic regulatory proteins and enzymes in the spinal cord suggests that neurosteroids locally generated could modulate pain messages. In this study we explored temporal changes in the spinal expression of the 18kDa translocator protein TSPO, the steroidogenic acute regulatory protein (StAr) and the steroidogenic enzyme 5α-reductase (5α-RI/II) in an experimental model of central chronic pain. Male Sprague-Dawley rats were subjected to a SCI and sacrificed at different time points (1, 14 or 28days). The development of mechanical and cold allodynia was assessed. Injured animals showed an early increase in the mRNA levels of TSPO and 5α-RII, whereas in the chronic phase a significant decrease in the expression of 5α-RI and 5α-RII was observed, coinciding with the presence of allodynic behaviors. Furthermore, since we have shown that progesterone (PG) administration may offer a promising perspective in pain modulation, we also evaluated the expression of steroidogenic proteins and enzymes in injured animals receiving daily injections of the steroid. PG-treated did not develop allodynia and showed a marked increase in the mRNA levels of TSPO, StAR, 5α-RI and 5α-RII 28days after injury. Our results suggest that in the acute phase after SCI, the increased expression of TSPO and 5α-RII may represent a protective endogenous response against tissue injury, which is not maintained in the chronic allodynic phase. PG may favor local steroidogenesis and the production of its reduced metabolites, which could contribute to the antiallodynic effects observed after PG treatment.


Subject(s)
Carrier Proteins/metabolism , Cholestenone 5 alpha-Reductase/metabolism , Neuralgia/metabolism , Progesterone/administration & dosage , Receptors, GABA-A/metabolism , Spinal Cord Injuries/metabolism , Animals , Hyperalgesia/enzymology , Hyperalgesia/etiology , Hyperalgesia/metabolism , Male , Neuralgia/enzymology , Neuralgia/etiology , Neuralgia/prevention & control , Pain Threshold/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/complications , Spinal Cord Injuries/enzymology
14.
Steroids ; 110: 77-87, 2016 06.
Article in English | MEDLINE | ID: mdl-27091763

ABSTRACT

The present review discusses the potential role of neurosteroids/neuroactive steroids in the regulation of nociceptive and neuropathic pain, and recapitulates the current knowledge on the main mechanisms involved in the reduction of pain, especially those occurring at the dorsal horn of the spinal cord, a crucial site for nociceptive processing. We will make special focus on progesterone and its derivative allopregnanolone, which have been shown to exert remarkable actions in order to prevent or reverse the maladaptive changes and pain behaviors that arise after nervous system damage in various experimental neuropathic conditions.


Subject(s)
Neuralgia/metabolism , Neurotransmitter Agents/metabolism , Animals , Humans , Neuralgia/pathology , Nociception/physiology , Pregnanolone/metabolism , Progesterone/metabolism , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/pathology
15.
J Neuroimmunol ; 292: 85-92, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26943964

ABSTRACT

Neuropathic pain is a frequent complication of spinal cord injury (SCI), still refractory to conventional treatment. Glial cell activation and cytokine production contribute to the pathology of central neuropathic syndromes. In this study we evaluated the effects of progesterone, a neuroactive steroid, on pain development and the spinal expression of IL-1ß, its receptors (IL-1RI and IL-1RII) and antagonist (IL-1ra), IL-6 and TNFα, and NR1 subunit of NMDAR. Our results show that progesterone, by modulating the expression of pro-inflammatory cytokines and neuronal IL-1RI/NR1 colocalization, emerges as a promising agent to prevent chronic pain after SCI.


Subject(s)
Cytokines/metabolism , Progesterone/therapeutic use , Progestins/therapeutic use , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Animals , Cytokines/genetics , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Male , Neuralgia/etiology , Pain Threshold/drug effects , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord Injuries/complications , Time Factors
16.
J Steroid Biochem Mol Biol ; 154: 274-84, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26369614

ABSTRACT

The anti-inflammatory effects of progesterone have been increasingly recognized in several neuropathological models, including spinal cord inflammation. In the present investigation, we explored the regulation of proinflammatory factors and enzymes by progesterone at several time points after spinal cord injury (SCI) in male rats. We also demonstrated the role of the progesterone receptor (PR) in inhibiting inflammation and reactive gliosis, and in enhancing the survival of oligodendrocyte progenitors cells (OPC) in injured PR knockout (PRKO) mice receiving progesterone. First, after SCI in rats, progesterone greatly attenuated the injury-induced hyperexpression of the mRNAs of interleukin 1ß (IL1ß), IL6, tumor necrosis factor alpha (TNFα), inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2), all involved in oligodendrocyte damage. Second, the role of the PR was investigated in PRKO mice after SCI, in which progesterone failed to reduce the high expression of IL1ß, IL6, TNFα and IκB-α mRNAs, the latter being considered an index of reduced NF-κB transactivation. These effects occurred in a time framework coincident with a reduction in the astrocyte and microglial responses. In contrast to wild-type mice, progesterone did not increase the density of OPC and did not prevent apoptotic death of these cells in PRKO mice. Our results support a role of PR in: (a) the anti-inflammatory effects of progesterone; (b) the modulation of astrocyte and microglial responses and (c) the prevention of OPC apoptosis, a mechanism that would enhance the commitment of progenitors to the remyelination pathway in the injured spinal cord.


Subject(s)
Cell Survival , Gliosis/pathology , Oligodendroglia/pathology , Receptors, Progesterone/physiology , Spinal Cord Injuries/pathology , Animals , Cytokines/genetics , Gliosis/immunology , Inflammation Mediators/metabolism , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/genetics , Spinal Cord Injuries/immunology
17.
Steroids ; 103: 42-57, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26301525

ABSTRACT

Levels of steroids in the adult central nervous system (CNS) show marked changes in response to stress, degenerative disorders and injury. However, their analysis in complex matrices such as fatty brain and spinal cord tissues, and even in plasma, requires accurate and precise analytical methods. Radioimmunoassays (RIA) and enzyme-linked immunosorbent assays, even with prepurification steps, do not provide sufficient specificity, and they are at the origin of many inconsistent results in the literature. The analysis of steroids by mass spectrometric methods has become the gold standard for accurate and sensitive steroid analysis. However, these technologies involve multiple purification steps prone to errors, and they only provide accurate reference values when combined with careful sample workup. In addition, the interpretation of changes in CNS steroid levels is not an easy task because of their multiple sources: the endocrine glands and the local synthesis by neural cells. In the CNS, decreased steroid levels may reflect alterations of their biosynthesis, as observed in the case of chronic stress, post-traumatic stress disorders or depressive episodes. In such cases, return to normalization by administering exogenous hormones or by stimulating their endogenous production may have beneficial effects. On the other hand, increases in CNS steroids in response to acute stress, degenerative processes or injury may be part of endogenous protective or rescue programs, contributing to the resistance of neural cells to stress and insults. The aim of this review is to encourage a more critical reading of the literature reporting steroid measures, and to draw attention to the absolute need for well-validated methods. We discuss reported findings concerning changing steroid levels in the nervous system by insisting on methodological issues. An important message is that even recent mass spectrometric methods have their limits, and they only become reliable tools if combined with careful sample preparation.


Subject(s)
Brain Injuries/physiopathology , Central Nervous System/physiopathology , Neurodegenerative Diseases/physiopathology , Steroids/analysis , Stress, Physiological , Adult , Humans , Radioimmunoassay , Tandem Mass Spectrometry
18.
Neural Regen Res ; 9(15): 1422-4, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25317151

ABSTRACT

There is currently no standard pharmacological treatment for spinal cord injury. Here, we suggest that progesterone, a steroid hormone, may be a promising therapeutical candidate as it is already for traumatic brain injury, where it has reached phase II clinical trials. We rely on previous works showing anti-inflammatory, neuroprotective and promyelinating roles for progesterone after spinal cord injury and in our recent paper, in which we demonstrate that progesterone diminishes lesion, preserves white matter integrity and improves locomotor recovery in a clinically relevant model of spinal cord lesion.

19.
J Neurotrauma ; 31(9): 857-71, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24460450

ABSTRACT

Progesterone is an anti-inflammatory and promyelinating agent after spinal cord injury, but its effectiveness on functional recovery is still controversial. In the current study, we tested the effects of chronic progesterone administration on tissue preservation and functional recovery in a clinically relevant model of spinal cord lesion (thoracic contusion). Using magnetic resonance imaging, we observed that progesterone reduced both volume and rostrocaudal extension of the lesion at 60 days post-injury. In addition, progesterone increased the number of total mature oligodendrocytes, myelin basic protein immunoreactivity, and the number of axonal profiles at the epicenter of the lesion. Further, progesterone treatment significantly improved motor outcome as assessed using the Basso-Bresnahan-Beattie scale for locomotion and CatWalk gait analysis. These data suggest that progesterone could be considered a promising therapeutical candidate for spinal cord injury.


Subject(s)
Motor Activity/drug effects , Progesterone/pharmacology , Progestins/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/pathology , White Matter/drug effects , Animals , Disease Models, Animal , Immunohistochemistry , Magnetic Resonance Imaging , Male , Rats , Rats, Wistar
20.
CNS Neurol Disord Drug Targets ; 12(8): 1205-18, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24040821

ABSTRACT

Substantial evidence supports that progesterone exerts many functions in the central and peripheral nervous system unrelated to its classical role in reproduction. In this review we first discussed progesterone effects following binding to the classical intracellular progesterone receptors A and B and several forms of membrane progesterone receptors, the modulation of intracellular signalling cascades and the interaction of progesterone reduced metabolites with neurotransmitter receptors. We next described our results involving animal models of human neuropathologies to elucidate the protective roles of progesterone. We described: (a) the protective and promyelinating effects of progesterone in experimental spinal cord injury; (b) the progesterone protective effects exerted upon motoneurons in the degenerating spinal cord of Wobbler mouse model of amyotropic lateral sclerosis; (c) the protective and anti-inflammatory effects of progesterone in the murine experimental autoimmune encephalomyelitis model of multiple sclerosis and after lysolecithin demyelination; (d) the progesterone prevention of nociception and neuropathic pain which follow spinal cord injury; and (e) the protective effect of progesterone in experimental ischemic stroke. Whenever available, the molecular mechanisms involved in these progesterone effects were examined. The multiplicity of progesterone beneficial effects has opened new venues of research for neurological disorders. In this way, results obtained in animal models could provide the basis for novel therapeutic strategies and pre-clinical studies.


Subject(s)
Disease Models, Animal , Nervous System Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Progesterone/therapeutic use , Animals , Humans , Nervous System Diseases/metabolism , Nervous System Diseases/pathology , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...