Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Neurobiol Aging ; 32(5): 864-74, 2011 May.
Article in English | MEDLINE | ID: mdl-19482377

ABSTRACT

Administration of the neurosteroid progesterone (PROG) has been shown to be beneficial in a number of brain injury models and in two recent clinical trials. Given widespread vitamin D deficiency and increasing traumatic brain injuries (TBIs) in the elderly, we investigated the interaction of vitamin D deficiency and PROG with cortical contusion injury in aged rats. Vitamin D deficient (VitD-deficient) animals showed elevated inflammatory proteins (TNFα, IL-1ß, IL-6, NFκB p65) in the brain even without injury. VitD-deficient rats with TBI, whether given PROG or vehicle, showed increased inflammation and greater open-field behavioral deficits compared to VitD-normal animals. Although PROG was beneficial in injured VitD-normal animals, in VitD-deficient subjects neurosteroid treatment conferred no improvement over vehicle. A supplemental dose of 1,25-dihydroxyvitamin D(3) (VDH) given with the first PROG treatment dramatically improved results in VitD-deficient rats, but treatment with VDH alone did not. Our results suggest that VitD-deficiency can increase baseline brain inflammation, exacerbate the effects of TBI, and attenuate the benefits of PROG treatment; these effects may be reversed if the deficiency is corrected.


Subject(s)
Aging/drug effects , Brain Injuries/drug therapy , Progesterone/therapeutic use , Vitamin D Deficiency/metabolism , Aging/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain Injuries/metabolism , Cytokines/metabolism , Disease Models, Animal , Male , Rats , Rats, Inbred F344 , Vitamin D Deficiency/drug therapy , Vitamin D Deficiency/physiopathology
2.
J Cereb Blood Flow Metab ; 28(11): 1786-94, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18628783

ABSTRACT

Our laboratory has shown in numerous experiments that the neurosteroids progesterone (PROG) and allopregnanolone (ALLO) improve molecular and functional outcomes after traumatic brain injury (TBI). As coagulopathy is an important contributor to the secondary destruction of nervous tissue, we hypothesized that PROG and ALLO administration may also have a beneficial effect on coagulation protein expression after TBI. Adult male Sprague-Dawley rats were given bilateral contusions of the medial frontal cortex followed by treatments with PROG (16 mg/kg), ALLO (8 mg/kg), or vehicle (22.5% hydroxypropyl-beta-cyclodextrin). Controls received no injury or injections. Progesterone generally maintained procoagulant (thrombin, fibrinogen, and coagulation factor XIII), whereas ALLO increased anticoagulant protein expression (tissue-type plasminogen activator, tPA). In addition, PROG significantly increased the ratio of tPA bound to neuroserpin, a serine protease inhibitor that can reduce the activity of tPA. Our findings suggest that in a model of TBI, where blood loss may exacerbate injury, it may be preferable to treat patients with PROG, whereas it might be more appropriate to use ALLO as a treatment for thrombotic stroke, where a reduction in coagulation would be more beneficial.


Subject(s)
Brain Injuries/metabolism , Hemostasis/physiology , Pregnanolone/pharmacology , Progesterone/pharmacology , Tissue Plasminogen Activator/genetics , Animals , Blood Coagulation/drug effects , Brain Injuries/blood , Brain Injuries/genetics , Disease Models, Animal , Factor XIII/genetics , Fibrinogen/genetics , Gene Expression Regulation/drug effects , Hemostasis/drug effects , Humans , Male , Neuropeptides/metabolism , Pregnanolone/therapeutic use , Progesterone/therapeutic use , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Serpins/metabolism , Thrombin/genetics , Tissue Plasminogen Activator/metabolism , Neuroserpin
3.
J Neurotrauma ; 24(9): 1475-86, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17892409

ABSTRACT

Recent evidence has demonstrated that treatment with progesterone can attenuate many of the pathophysiological events following traumatic brain injury (TBI) in young adult rats, but this effect has not been investigated in aged animals. In this study, 20-month-old male Fischer 344 rats with bilateral contusions of the frontal cortex (n = 4 per group) or sham operations received 8, 16, or 32 mg/kg of progesterone or vehicle. Locomotor activity was measured at 72 h to assess behavioral recovery. Brain tissue was harvested at 24, 48, and 72 h, and Western blotting was performed for inflammatory and apoptotic factors. Edema was assessed at 48 h by measuring brain water content. Injured animals treated with 8 and 16 mg/kg progesterone showed decreased expression of COX-2, IL-6, and NFkappaB at all time points, indicating a reduction in the acute inflammatory process compared to vehicle. The 16 mg/kg group also showed reduced apoptosis at all time points as well as decreased edema and improved locomotor outcomes. Thus, in aged male rats, treatment with 16 mg/kg progesterone improves short-term motor recovery and attenuates edema, secondary inflammation, and cell death after TBI.


Subject(s)
Brain Injuries/drug therapy , Neuroprotective Agents/therapeutic use , Progesterone/therapeutic use , Progestins/therapeutic use , Recovery of Function/drug effects , Age Factors , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blotting, Western , Brain Edema/drug therapy , Brain Edema/pathology , Brain Injuries/pathology , Cytokines/biosynthesis , Cytokines/drug effects , Gene Expression/drug effects , Inflammation/drug therapy , Inflammation/pathology , Male , Motor Activity/drug effects , Rats , Rats, Inbred F344
4.
Neuropharmacology ; 51(6): 1078-85, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16926035

ABSTRACT

Previous work shows that neurosteroid enantiomers activate specific molecular receptors that relay neuroprotection. However, the actions of the enantiomer of progesterone (ent-PROG) at the PROG receptor (PR) are unknown. PR binding and transcriptional assays were performed to determine the actions of ent-PROG at the classical PR. Additionally, the neuroprotective effects of ent-PROG in traumatic brain injury (TBI) were investigated and compared to the actions of PROG and its metabolite allopregnanolone (ALLO), both of which have been shown to have neuroprotective properties when given after TBI. Binding studies performed in COS cells over-expressing the PR showed that ent-PROG inhibited PROG binding to the PR. In contrast, ent-PROG did not activate PR-mediated transcription. Rats received bilateral medial frontal cortex injury followed by treatments at 1, 6, 24 and 48h with PROG, ALLO or ent-PROG. Brains were processed for edema, protein and enzyme activity. ent-PROG treatment in vivo decreased cerebral edema, cell death mediators, inflammatory cytokines, and reactive gliosis, and increased antioxidant activity. These findings suggest that the progestin-mediated pro-survival response seen with TBI is regulated either independently of the classical PR or via nongenomic PR-regulated actions.


Subject(s)
Brain Injuries/drug therapy , Neuroprotective Agents , Progesterone/pharmacology , Animals , Antioxidants/metabolism , Biomarkers , Brain Edema/drug therapy , Brain Edema/metabolism , Brain Edema/pathology , Brain Injuries/pathology , COS Cells , Cell Death/drug effects , Cells, Cultured , Chlorocebus aethiops , Cytokines/analysis , Cytokines/biosynthesis , Gene Expression Regulation/drug effects , Genes, p53/physiology , Gliosis/pathology , Glutathione Reductase/metabolism , Inflammation/metabolism , Male , Nerve Tissue Proteins/metabolism , Oxidative Stress/drug effects , Progesterone/chemistry , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/drug effects , Receptors, Progesterone/genetics , Stereoisomerism
5.
Pharmacol Biochem Behav ; 84(3): 420-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16870241

ABSTRACT

The benefits of continuous progesterone release via subcutaneous silastic capsule implants were compared to daily subcutaneous injections in a rat model of traumatic brain injury (TBI). Adult male Sprague-Dawley rats received either bilateral frontal cortex contusions or sham surgery. Rats were injected with progesterone or vehicle at 1 and 6 h post-injury, then once every 24 h for six days with tapering of the dose over the final two treatments. Progesterone-packed silastic capsules were implanted post-injury while the animals were anesthetized. Behavioral assays for anxiety and locomotor activity were evaluated pre- and post-TBI. Brains were extracted eight days post-TBI and prepared for molecular assays. Decreased GABAA-4 levels complemented a decrease in anxiety behaviors on the Elevated Plus Maze for capsule compared to progesterone-injected animals prior to daily injections. All groups with implanted capsules increased locomotor activity compared to those given progesterone injections. In conclusion, steady-state progesterone treatment after TBI decreases edema and anxiety and increases activity, thus enhancing behavioral recovery. A continuous mode of pharmacological administration may prove to be more beneficial in translational and clinical testing than bolus injections over the same period of time.


Subject(s)
Brain Injuries/drug therapy , Progesterone/administration & dosage , Animals , Anxiety/drug therapy , Apoptosis , Behavior, Animal/drug effects , Edema/drug therapy , Inflammation , Male , Maze Learning/drug effects , Progesterone/metabolism , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/biosynthesis , Time Factors
6.
Exp Neurol ; 200(2): 378-85, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16797538

ABSTRACT

We previously demonstrated that after traumatic brain injury (TBI), acute progesterone withdrawal (AW) causes an increase in anxiety behaviors and cerebro-cellular inflammation compared to tapered progesterone withdrawal (TW). Our current study investigates the behavioral and cellular effects of AW two weeks after termination of treatments to determine the longer-term influence of withdrawal after injury. Adult, male Sprague-Dawley rats received either bilateral frontal cortex contusion (L) or sham (S) surgery. Rats were injected at 1 and 6 h post-injury, then every 24 h for six days. Vehicle (V)-treated rats were given 9 injections of 22.5% cyclodextrin, whereas AW rats received 9 injections of 16 mg/kg progesterone and TW rats received 7 injections of P at 16 mg/kg, followed by one at 8 mg/kg and one at 4 mg/kg. On day 8, sensory neglect and locomotor activity tests were initiated. Animals were killed 22 days post-TBI and the brains prepared for either molecular or histological analysis. Western blotting revealed increased brain-derived neurotrophic factor (BDNF) and heat shock protein 70 (HSP70) in TW vs. AW animals. P53 was increased in VL animals, whereas all progesterone-treated groups were equivalent to shams. TW animals had markedly decreased sensory neglect compared to AW animals and increased center time in locomotor activity assays. In addition, lesion reconstruction revealed a decreased lesion size for TWL over AWL over VL animals. Glial fibrillary acidic protein (GFAP) immunofluorescent staining followed this pattern as well. In conclusion, after TBI, AW affects select behaviors and molecular markers in the chronic recovery period.


Subject(s)
Anesthetics/administration & dosage , Motor Activity/drug effects , Pregnanolone/administration & dosage , Recovery of Function/drug effects , Substance Withdrawal Syndrome , Anesthetics/adverse effects , Animals , Behavior, Animal , Blotting, Western/methods , Brain Injuries/drug therapy , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Drug Administration Schedule , Glial Fibrillary Acidic Protein/metabolism , HSP70 Heat-Shock Proteins/metabolism , Immunohistochemistry/methods , Male , Peptides, Cyclic , Perceptual Disorders/etiology , Perceptual Disorders/physiopathology , Pregnanolone/adverse effects , Random Allocation , Rats , Rats, Sprague-Dawley , Time Factors , Tumor Suppressor Protein p53/metabolism , Upper Extremity/physiopathology
7.
Exp Neurol ; 195(2): 423-9, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16039652

ABSTRACT

Systemic injections of the neurosteroid progesterone improve cognitive recovery after traumatic brain injury (TBI) and stroke, and decrease molecular indicators of neuronal damage. Suddenly withdrawing progesterone after repeated dosing (PW) exacerbates ischemia and causes increased anxiety, seizure susceptibility, and excitotoxicity. Adult male Sprague-Dawley rats received either bilateral medial frontal cortex contusions or sham surgery. Injections were administered at 1 and 6 h post-injury, then every 24 h for 7 days. Vehicle-treated rats received 2-hydroxypropyl-beta-cyclodextrin (HBC). Acute PW (AW) rats received a full 16 mg/ml of progesterone for 7 days, and tapered PW (TW) rats received 5 days at full dosage, then 2 days with progressively halved dosages. Anxiety behaviors were observed pre- and post-surgery, and compared to levels at the peak of withdrawal. AW rats with lesions exhibited significantly more anxiety than any other treatment group, while both lesion- and sham-operated TW rats were indistinguishable from vehicle-treated intact animals. After behavioral tests were complete, the brains were extracted and prepared for Western blotting. TNFalpha, cFos, Caspase-3, and NFkappaB, among others, were investigated. While all progesterone treatments resulted in improved molecular recovery, TW animals had significantly fewer active markers for apoptosis and inflammation than AW animals. In conclusion, although progesterone treatment decreases inflammation and apoptosis, acute withdrawal increases activity in some apoptotic and inflammatory pathways and increases anxiety behavior during the acute healing phase. A tapered withdrawal of the hormone further enhances short-term recovery after TBI.


Subject(s)
Behavior, Animal/drug effects , Brain Injuries/drug therapy , Gene Expression Regulation/drug effects , Progesterone/administration & dosage , Recovery of Function/drug effects , Substance Withdrawal Syndrome , 2-Hydroxypropyl-beta-cyclodextrin , Animals , Blotting, Western/methods , Caspase 3 , Caspases/metabolism , Disease Models, Animal , Drug Administration Schedule , Male , Maze Learning/drug effects , Progesterone/adverse effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Tumor Necrosis Factor-alpha/metabolism , beta-Cyclodextrins/administration & dosage , NF-kappaB-Inducing Kinase
8.
Biomaterials ; 24(10): 1759-70, 2003 May.
Article in English | MEDLINE | ID: mdl-12593958

ABSTRACT

Integrin receptors mediate cell adhesion to extracellular matrices and trigger signals that direct cell function. While many integrins bind to the arginine-glycine-aspartic acid (RGD) motif present in numerous extracellular proteins, integrin alpha(5)beta(1) requires both the PHSRN synergy site in the 9th and the RGD site in the 10th type III repeat of fibronectin (FN). Binding of alpha(5)beta(1) to FN is critical to many cellular processes, including osteoblast and myoblast differentiation. This work focused on engineering integrin-specific bioadhesive surfaces by immobilizing a recombinant FN fragment (FNIII(7-10)) encompassing the alpha(5)beta(1) binding domains of FN. Model hybrid surfaces were engineered by immobilizing FNIII(7-10) onto passively adsorbed, non-adhesive albumin. Homo- and hetero-bifunctional crosslinkers of varying spacer-arm length targeting either the cysteine or lysine groups on FNIII(7-10) were investigated in ELISA and cell adhesion assays to optimize immobilization densities and activity. FN-mimetic surfaces presenting controlled densities of FNIII(7-10) were generated by varying the concentration of FNIII(7-10) in the coupling solution at a constant crosslinker concentration. Cells adhered to these functionalized surfaces via integrin alpha(5)beta(1) and blocking with integrin-specific antibodies completely eliminated adhesion. In addition, adherent cells spread and assembled focal adhesions containing alpha(5)beta(1), vinculin, and talin. This biomolecular engineering strategy represents a robust approach to increase biofunctional activity and integrin specificity of biomimetic materials.


Subject(s)
Cell Adhesion , Fibronectins/metabolism , Integrin alpha5beta1/metabolism , Tissue Engineering/methods , 3T3 Cells , Animals , Binding Sites , Biocompatible Materials , Cattle , Cross-Linking Reagents , Fibronectins/chemistry , Humans , Imidoesters , Materials Testing , Mice , Models, Molecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serum Albumin, Bovine/chemistry , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...