Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Cureus ; 14(7): e27296, 2022 Jul.
Article in English | MEDLINE | ID: mdl-36043003

ABSTRACT

This review examines various aspects of traumatic brain injury (TBI) and its potential role as a causative agent for type 2 diabetes mellitus (T2DM) in the veteran population. The pituitary glands and the hypothalamus, both housed in the intracranial space, are the most important structures for the homeostatic regulation of almost every hormone in the human body. As such, TBI not only causes psychological and cognitive impairments but can also disrupt the endocrine system. It is well established that in addition to having a high prevalence of chronic traumatic encephalopathy (CTE), veterans have a very high risk of developing various chronic medical conditions. Unfortunately, there are no measures or prophylactic agents that can have a meaningful impact on this medically complex patient population. In this review, we explore several important factors pertaining to both acute and chronic TBI that can provide additional insight into why veterans tend to develop T2DM later in life. We focus on the unique combination of risk factors in this population not typically found in civilians or other individuals with a non-military background. These include post-traumatic stress disorder, CTE, and environmental factors relating to occupation and lifestyle.

2.
Mol Neurobiol ; 58(8): 3805-3816, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33847913

ABSTRACT

A substantial literature supports the notion that cancer is a metabolic disease. Mitochondria are sexually dimorphic, and progesterone (P4) plays a key regulatory role in mitochondrial functions. We investigated the effect of P4 on mitochondrial functions in three human glioblastoma multiforme (GBM) cell lines. In dose-response and time-response studies, GBM cells were exposed to different concentrations of P4 followed by mitochondrial stress-testing with a Seahorse analyzer. Data were analyzed for oxygen consumption rate (OCR), extracellular acidification rate (ECAR), and spare respiratory capacity (SRC) to determine the effects of P4 exposure on mitochondrial respiration and rate of glycolysis. We also examined the effect of P4 on mitochondrial superoxide radical generation by confocal microscopy. As early as 1h post-P4 exposure, we found a substantial dose-dependent inhibitory effect of P4 on OCR, ECAR, and SRC in all GBM cell lines. P4 treatment altered the levels of basal respiration, maximum respiration, nonmitochondrial oxygen consumption, ATP production, and proton leak. P4 given at 80-µM concentration showed the maximum inhibitory effect compared to controls. Live imaging data showed an 11-22% increase in superoxide radical generation in all three GBM cell lines following 6h exposure to a high concentration of P4. Our data show that high-dose P4 exerts an inhibitory effect on both mitochondrial respiration and glycolysis in GBM cells. These effects would lead to decreased tumor size and rate of growth, representing a potential treatment to control the spread of GBM.


Subject(s)
Energy Metabolism/drug effects , Glioblastoma/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Progesterone/pharmacology , Animals , Cell Line, Tumor , Energy Metabolism/physiology , Glioblastoma/drug therapy , Humans , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Progesterone/therapeutic use , Smegmamorpha
3.
Mol Neurobiol ; 58(3): 950-963, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33063282

ABSTRACT

Vitamin D deficiency (Ddef) alters morphology and outcomes after a stroke. We investigated the interaction of Ddef following post-stroke systemic inflammation and evaluated whether administration of progesterone (P) or vitamin D (D) will improve outcomes. Ddef rats underwent stroke with lipopolysaccharide (LPS)-induced systemic inflammation. Rats were randomly divided into 9 groups and treated with P, D, or vehicle for 4 days. At day 4, rats were tested on different behavioral parameters. Markers of neuronal inflammation, endoplasmic reticulum stress, oxidative stress, white matter integrity, and apoptosis were measured along with immune cell populations from the spleen, thymus, and blood. Severely altered outcomes were observed in the Ddef group compared to the D-sufficient (Dsuf) group. Stroke caused peripheral immune dysfunction in the Dsuf group which was worse in the Ddef group. Systemic inflammation exacerbated injury outcomes in the Dsuf group and these were worse in the Ddef group. Monotherapy with P/D showed beneficial functional effects but the combined treatment showed better outcomes than either alone. Ddef as a comorbid condition with stroke worsens stroke outcomes and can delay functional recovery. Combination treatment with P and D might be promising for future stroke therapeutics in Ddef.


Subject(s)
Progesterone/pharmacology , Stroke/immunology , Stroke/physiopathology , Vitamin D Deficiency/immunology , Vitamin D Deficiency/physiopathology , Vitamin D/pharmacology , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Behavior, Animal/drug effects , Biomarkers/metabolism , Cyclooxygenase 2/metabolism , Endoplasmic Reticulum Stress/drug effects , Hand Strength , Inflammation/blood , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Lipopolysaccharides , Male , Myelin Basic Protein/metabolism , Neurons/drug effects , Neurons/pathology , Nitric Oxide Synthase Type II/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Reproducibility of Results , Spleen/pathology , Stroke/blood , Stroke/complications , Thymus Gland/pathology , Transcription Factor CHOP/metabolism , Vitamin D Deficiency/blood , Vitamin D Deficiency/complications , White Matter/metabolism , White Matter/pathology
4.
Neuropharmacology ; 181: 108327, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32950558

ABSTRACT

Post-stroke systemic inflammation, due to the injury itself and exacerbated by in-hospital infections, can increase morbidity and mortality in stroke patients. In this study, we examined the immunomodulatory effects of progesterone (P4) alone and in combination with vitamin D hormone (VDH) on acute phase post-stroke peripheral immune dysfunction and functional/behavioral deficits. Adult rats underwent transient middle cerebral artery occlusion/reperfusion (tMCAO) and delayed systemic inflammation was induced by injections of lipopolysaccharide (LPS) beginning 24 h post-stroke. Animals were tested for behavioral outcomes and immune function at day 4 post-stroke. We also measured infarction volume and markers of neuronal inflammation (GFAP, IL-6) and apoptosis (cleaved caspase-3) in brain post-stroke. We observed the worst stroke outcomes in the stroke + systemic inflammation group compared to the stroke-alone group. Flow cytometric analysis of different subsets of immune cells in blood, spleen and thymus revealed peripheral immune dysfunction which was restored by both P4 and VDH monotherapy. P4 monotherapy reduced infarction volume, behavioral/functional deficits, peripheral immune dysfunction, neuronal inflammation, and apoptosis induced by post-stroke systemic inflammation. Combination treatment with P4+VDH improved outcomes better than monotherapy. Our findings can be taken to suggest that the current standard of care for stroke and post-stroke infection can be substantially improved by P4 and VDH combination therapy.


Subject(s)
Immunologic Factors/pharmacology , Inflammation/prevention & control , Ischemic Stroke/pathology , Progesterone/pharmacology , Vitamin D/pharmacology , Vitamins/pharmacology , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Caspase 3/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hand Strength , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/prevention & control , Inflammation/etiology , Interleukin-6/metabolism , Ischemic Stroke/immunology , Ischemic Stroke/psychology , Male , Motor Activity/drug effects , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Treatment Outcome
5.
Int J Mol Sci ; 21(11)2020 May 26.
Article in English | MEDLINE | ID: mdl-32466385

ABSTRACT

NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome inhibition and autophagy induction attenuate inflammation and improve outcome in rodent models of cerebral ischemia. However, the impact of chronic stress on NLRP3 inflammasome and autophagic response to ischemia remains unknown. Progesterone (PROG), a neuroprotective steroid, shows promise in reducing excessive inflammation associated with poor outcome in ischemic brain injury patients with comorbid conditions, including elevated stress. Stress primes microglia, mainly by the release of alarmins such as high-mobility group box-1 (HMGB1). HMGB1 activates the NLRP3 inflammasome, resulting in pro-inflammatory interleukin (IL)-1ß production. In experiment 1, adult male Sprague-Dawley rats were exposed to social defeat stress for 8 days and then subjected to global ischemia by the 4-vessel occlusion model, a clinically relevant brain injury associated with cardiac arrest. PROG was administered 2 and 6 h after occlusion and then daily for 7 days. Animals were killed at 7 or 14 days post-ischemia. Here, we show that stress and global ischemia exert a synergistic effect in HMGB1 release, resulting in exacerbation of NLRP3 inflammasome activation and autophagy impairment in the hippocampus of ischemic animals. In experiment 2, an in vitro inflammasome assay, primary microglia isolated from neonatal brain tissue, were primed with lipopolysaccharide (LPS) and stimulated with adenosine triphosphate (ATP), displaying impaired autophagy and increased IL-1ß production. In experiment 3, hippocampal microglia isolated from stressed and unstressed animals, were stimulated ex vivo with LPS, exhibiting similar changes than primary microglia. Treatment with PROG reduced HMGB1 release and NLRP3 inflammasome activation, and enhanced autophagy in stressed and unstressed ischemic animals. Pre-treatment with an autophagy inhibitor blocked Progesterone's (PROG's) beneficial effects in microglia. Our data suggest that modulation of microglial priming is one of the molecular mechanisms by which PROG ameliorates ischemic brain injury under stressful conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Autophagy , Brain Ischemia/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Stress, Psychological/metabolism , Animals , Brain Ischemia/complications , Cells, Cultured , Hippocampus/drug effects , Hippocampus/metabolism , Interleukin-1beta/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Rats , Rats, Sprague-Dawley , Stress, Psychological/complications
6.
Sci Rep ; 9(1): 988, 2019 01 30.
Article in English | MEDLINE | ID: mdl-30700763

ABSTRACT

We examined the effect of progesterone treatments on glycolytic metabolism and senescence as possible mechanisms in controlling the growth of glioblastoma multiforme (GBM). In an orthotopic mouse model, after tumor establishment, athymic nude mice received treatment with progesterone or vehicle for 40 days. Compared to controls, high-dose progesterone administration produced a significant reduction in tumor size (~47%) and an increased survival rate (~43%) without any demonstrable toxicity to peripheral organs (liver, kidney). This was accompanied by a significant improvement in spontaneous locomotor activity and reduced anxiety-like behavior. In a follow-up in vitro study of U87MG-luc, U87dEGFR and U118MG tumor cells, we observed that high-dose progesterone inhibited expression of Glut1, which facilitated glucose transport into the cytoplasm; glyceraldehyde 3-phosphate dehydrogenase (GAPDH; a glycolysis enzyme); ATP levels; and cytoplasmic FoxO1 and Phospho-FoxO1, both of which control glycolytic metabolism through upstream PI3K/Akt/mTOR signaling in GBM. In addition, progesterone administration attenuated EGFR/PI3K/Akt/mTOR signaling, which is highly activated in grade IV GBM. High-dose progesterone also induced senescence in GBM as evidenced by changes in cell morphology and ß-galactocidase accumulation. In conclusion, progesterone inhibits the modulators of glycolytic metabolism and induces premature senescence in GBM cells and this can help to reduce/slow tumor progression.


Subject(s)
Cellular Senescence/drug effects , Glioblastoma/pathology , Glycolysis/drug effects , Progesterone/pharmacology , Adenosine Triphosphate/metabolism , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Biomarkers/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Enzyme Assays , Glioblastoma/blood supply , Luciferases/metabolism , Mice, Nude , Models, Biological , Motor Activity/drug effects , Neovascularization, Pathologic/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
7.
Exp Neurol ; 306: 138-148, 2018 08.
Article in English | MEDLINE | ID: mdl-29729249

ABSTRACT

In adult mice with unilateral optic nerve crush injury (ONC), we studied visual response plasticity in the visual cortex following stimulation with sinusoidal grating. We examined visually evoked potentials (VEP) in the primary visual cortex ipsilateral and contralateral to the crushed nerve. We found that unilateral ONC induces enhancement of visual response on the side ipsilateral to the injury that is evoked by visual stimulation to the intact eye. This enhancement was associated with supranormal spatial frequency thresholds in the intact eye when tested using optomotor response. To probe whether injury-induced disinhibition caused the potentiation, we treated animals with the neurosteroid allopregnanolone, a potent agonist of the GABAA receptor, one hour after crush and on post-injury days 3, 8, 13, and 18. Allopregnanolone diminished enhancement of the VEP and this effect was associated with the upregulated synthesis of the δ-subunit of the GABAA receptor. Our study shows a new aspect of experience-dependent plasticity following unilateral ONC. This hyper-activity in the ipsilateral visual cortex is prevented by upregulation of GABA inhibition with allopregnanolone. Our findings suggest the therapeutic potential of allopregnanolone for modulation of plasticity in certain eye and brain disorders and a possible role for disinhibition in ipsilateral hyper-activity following unilateral ONC.


Subject(s)
Optic Nerve Injuries/drug therapy , Optic Nerve Injuries/physiopathology , Pregnanolone/therapeutic use , Visual Cortex/drug effects , Animals , Electrodes, Implanted , Evoked Potentials, Visual/drug effects , Eye Movements/drug effects , Functional Laterality/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Crush , Oculomotor Muscles/drug effects , Oculomotor Muscles/innervation , Pregnanolone/pharmacology , Receptors, GABA-A/drug effects , Visual Cortex/physiopathology
9.
Horm Behav ; 96: 21-30, 2017 11.
Article in English | MEDLINE | ID: mdl-28866326

ABSTRACT

Despite improved therapeutic methods, CNS toxicity resulting from cancer treatment remains a major cause of post-treatment morbidity. More than half of adult patients with cranial irradiation for brain cancer develop neurobehavioral/cognitive deficits that severely impact quality of life. We examined the neuroprotective effects of the neurosteroid progesterone (PROG) against ionizing radiation (IR)-induced neurobehavioral/cognitive deficits in mice. Male C57/BL mice were exposed to one of two fractionated dose regimens of IR (3Gy×3 or 3Gy×5). PROG (16mg/kg; 0.16mg/g) was given as a pre-, concurrent or post-IR treatment for 14days. Mice were tested for short- and long-term effects of IR and PROG on neurobehavioral/cognitive function on days 10 and 30 after IR treatment. We evaluated both hippocampus-dependent and -independent memory functions. Locomotor activity, elevated plus maze, novel object recognition and Morris water maze tests revealed behavioral deficits following IR. PROG treatment produced improvement in behavioral performance at both time points in the mice given IR. Western blot analysis of hippocampal and cortical tissue showed that IR at both doses induced astrocytic activation (glial fibrillary acidic protein), reactive macrophages/microglia (CD68) and apoptosis (cleaved caspase-3) and PROG treatment inhibited these markers of brain injury. There was no significant difference in the degree of deficit in any test between the two dose regimens of IR at either time point. These findings could be important in the context of patients with brain tumors who may undergo radiotherapy and eventually develop cognitive deficits.


Subject(s)
Cognition Disorders/etiology , Cognition Disorders/prevention & control , Cognition/drug effects , Cranial Irradiation/adverse effects , Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Animals , Brain Injuries/complications , Brain Injuries/etiology , Brain Injuries/prevention & control , Cranial Irradiation/psychology , Hippocampus/drug effects , Male , Maze Learning/drug effects , Memory/drug effects , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/drug effects , Neurogenesis/drug effects , Quality of Life , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/prevention & control , Treatment Outcome
10.
Brain Behav Immun ; 66: 177-192, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28648389

ABSTRACT

Despite the fact that stress is associated with increased risk of stroke and worsened outcome, most preclinical studies have ignored this comorbid factor, especially in the context of testing neuroprotective treatments. Preclinical research suggests that stress primes microglia, resulting in an enhanced reactivity to a subsequent insult and potentially increasing vulnerability to stroke. Ischemia-induced activated microglia can be polarized into a harmful phenotype, M1, which produces pro-inflammatory cytokines, or a protective phenotype, M2, which releases anti-inflammatory cytokines and neurotrophic factors. Selective modulation of microglial polarization by inhibiting M1 or stimulating M2 may be a potential therapeutic strategy for treating cerebral ischemia. Our laboratory and others have shown progesterone to be neuroprotective against ischemic stroke in rodents, but it is not known whether it will be as effective under a comorbid condition of chronic stress. Here we evaluated the neuroprotective effect of progesterone on the inflammatory response in the hippocampus after exposure to stress followed by global ischemia. We focused on the effects of microglial M1/M2 polarization and pro- and anti-inflammatory mediators in stressed ischemic animals. Male Sprague-Dawley rats were exposed to 8 consecutive days of social defeat stress and then subjected to global ischemia or sham surgery. The rats received intraperitoneal injections of progesterone (8mg/kg) or vehicle at 2h post-ischemia followed by subcutaneous injections at 6h and once every 24h post-injury for 7days. The animals were killed at 7 and 14days post-ischemia, and brains were removed and processed to assess outcome measures using histological, immunohistochemical and molecular biology techniques. Pre-ischemic stress (1) exacerbated neuronal loss and neurodegeneration as well as microglial activation in the selectively vulnerable CA1 hippocampal region, (2) dysregulated microglial polarization, leading to upregulation of both M1 and M2 phenotype markers, (3) increased pro-inflammatory cytokine expression, and (4) reduced anti-inflammatory cytokine and neurotrophic factor expression in the ischemic hippocampus. Treatment with progesterone significantly attenuated stress-induced microglia priming by modulating polarized microglia and the inflammatory environment in the hippocampus, the area most vulnerable to ischemic injury. Our findings can be taken to suggest that progesterone holds potential as a candidate for clinical testing in ischemic stroke where high stress may be a contributing factor.


Subject(s)
Brain Ischemia/metabolism , Encephalitis/metabolism , Microglia/drug effects , Microglia/metabolism , Neuroprotective Agents/administration & dosage , Progesterone/administration & dosage , Stress, Psychological/metabolism , Animals , Brain Ischemia/complications , Brain Ischemia/pathology , Cell Polarity , Depression/complications , Encephalitis/complications , Encephalitis/drug therapy , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Inflammation Mediators/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Rats, Sprague-Dawley , Stress, Psychological/complications , Stress, Psychological/pathology
11.
Neuroscience ; 350: 1-12, 2017 05 14.
Article in English | MEDLINE | ID: mdl-28315443

ABSTRACT

We investigated the effect of progesterone (P4) treatment on diabetes/hyperglycemia-induced pathological changes in brain, spinal cord and sciatic nerve tissue in male rats. Animals were rendered hyperglycemic by a single dose of streptozotocin (STZ). P4 treatment was started after hyperglycemia was confirmed and body weight and blood glucose levels were monitored once/week for 5weeks. Rats underwent behavioral testing at week 5 and were then euthanized for histology. We assessed the expression of markers of angiogenesis (vascular endothelial growth factor (VEGF)), inflammation (interleukin-6 (IL-6)) and tissue injury (CD11b, NG2, COX2 and matrix metalloproteinase-2 (MMP-2)) in the brain, spinal cord and sciatic nerve. We also examined the regenerative effect of P4 on pathological changes in intra-epidermal nerve fibers (IENF) of the footpads. Diabetes/hyperglycemia led to body weight loss over 5weeks and P4 treatment reduced this loss. At week 5, blood-glucose levels were significantly lower in the P4-treated diabetic group compared to vehicle. Compared to sham or P4-treated groups, the diabetic vehicle group showed hyperactivity on the spontaneous locomotor activity test. Western blot data revealed upregulation of VEGF, IL-6, CD11b, NG2, COX2 and MMP-2 levels in the vehicle group and P4 treatment normalized these expression levels. IENF densities were reduced in the vehicle group and normalized after P4 treatment. We conclude that P4 can reduce some of the chronic pathological responses to STZ-induced diabetes.


Subject(s)
Blood Glucose/drug effects , Central Nervous System/drug effects , Diabetes Mellitus, Experimental/pathology , Hyperglycemia/metabolism , Progesterone/pharmacology , Sciatic Nerve/drug effects , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Hyperglycemia/chemically induced , Interleukin-6/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Progesterone/metabolism , Rats, Sprague-Dawley , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Spinal Cord/drug effects , Spinal Cord/metabolism , Streptozocin , Vascular Endothelial Growth Factor A/metabolism
14.
Horm Behav ; 84: 29-40, 2016 08.
Article in English | MEDLINE | ID: mdl-27283379

ABSTRACT

We investigated the neuroprotective effects of progesterone (P4) treatment in stroke-prone spontaneously hypertensive rats (SHRSPs) given 60-min transient middle cerebral artery occlusion (tMCAO). The treatment groups were: (1) Wistar-Kyoto (normotensive sham), (2) SHRSP (hypertensive sham), (3) tMCAO SHRSPs (SHRSP+tMCAO), and (4) SHRSP+tMCAO+P4. P4 (8mg/kg) was administered 1h after occlusion and then daily for 14days. We measured cerebral infarction volume, blood pressure and body weight. Behavioral outcomes were analyzed at post-stroke days 3, 9, and 14. To assess morphological protection we measured activation of microglia and astrocytes, oxidative stress, apoptosis, expression of vascular endothelial growth factor (VEGF), an angiogenic marker, and IL-1ß, a marker of inflammation, on day 14 post-stroke. There was no effect of P4 on body weight or systolic blood pressure compared to the SHRSP+tMCAO group. However, grip strength and sensory neglect measures in the P4 group were improved compared to SHRSP+tMCAO. In addition, significantly larger infarct volumes were seen in the SHRSP+tMCAO group compared to SHRSP+tMCAO+P4. Increased markers of the injury cascade such as macrophages, activated astrocytes, superoxide anion and apoptotic cells observed in the SHRSP+tMCAO group were significantly decreased by P4. We conclude that, despite hypertensive comorbidity, P4 improves functional outcomes and attenuates stroke infarct in hypertensive rats by reducing superoxide anion expression and by decreasing inflammation and neuronal apoptosis.


Subject(s)
Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/pathology , Neuroprotective Agents/pharmacology , Progesterone/pharmacology , Stroke/pathology , Animals , Apoptosis/drug effects , Blood Pressure/drug effects , Infarction, Middle Cerebral Artery/metabolism , Ischemic Attack, Transient/metabolism , Male , Microglia/drug effects , Oxidative Stress/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Stroke/metabolism , Vascular Endothelial Growth Factor A/metabolism
15.
Neuropharmacology ; 107: 317-328, 2016 08.
Article in English | MEDLINE | ID: mdl-27039043

ABSTRACT

Neonatal stroke is among the top ten causes of childhood death and permanent disability in survivors, but no safe and effective acute treatments exist. To advance understanding of its neuroprotective mechanisms, we examined the effects of progesterone (PROG) on local and systemic inflammation (IL-1ß, IL-6, TNFα), brain derived neurotrophic factor/Tropomyosin receptor kinase B (BDNF/TrkB) signaling, vascular damage (vascular endothelial growth factor (VEGF), matrix metalloproteinase-9 (MMP-9)), acute behavioral seizures and brain infarction size following neonatal arterial ischemic stroke in mice. CD1 mouse pups (postnatal day 12, mixed gender) received permanent unilateral right common carotid ligation (pUCCL) or sham surgery. Pups showing seizure activity during the first hour post-pUCCL were randomly assigned to receive PROG (8 mg/kg) or vehicle injections. PROG treatment significantly (p < 0.05) reduced seizure occurrence by ∼44% compared to vehicle and attenuated the expression of pro-inflammatory cytokines in serum and brain at different time-points. PROG differentially regulated the expression of BDNF and TrkB and the activity of VEGF and MMP-9 over the 7d period. Permanent UCCL resulted in severe hemispheric damage measured at 7 days post-pUCCL but PROG treatment produced a significant (p < 0.05) reduction in infarct volume (∼70%) compared to vehicle. A gender-based comparison of data revealed significantly greater seizure activity in males compared to females. However, we did not observe significant sex differences on any other markers of the injury at this early stage of development. PROG treatment is neuroprotective through a number of signaling pathways and can be beneficial in treating neonatal arterial ischemic stroke in CD1 mice.


Subject(s)
Brain Ischemia/metabolism , Brain-Derived Neurotrophic Factor/physiology , Progesterone/therapeutic use , Receptor, trkB/physiology , Signal Transduction/physiology , Stroke/metabolism , Animals , Animals, Newborn , Brain Ischemia/drug therapy , Female , Male , Mice , Progesterone/pharmacology , Signal Transduction/drug effects , Stroke/drug therapy , Treatment Outcome
16.
J Neurotrauma ; 33(1): 101-12, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-25970337

ABSTRACT

Patients enrolled in clinical trials for traumatic brain injury (TBI) may present with heterogeneous features over a range of injury severity, such as diffuse axonal injury, ischemia, edema, hemorrhage, oxidative damage, mitochondrial and metabolic dysfunction, excitotoxicity, inflammation, and other pathophysiological processes. To determine whether combination therapies might be more effective than monotherapy at attenuating moderate TBI or promoting recovery, the National Institutes of Health funded six preclinical studies in adult and immature male rats to evaluate promising acute treatments alone and in combination. Each of the studies had a solid rationale for its approach based on previous research, but only one reported significant improvements in long-term outcomes across a battery of behavioral tests. Four studies had equivocal results because of a lack of sensitivity of the outcome assessments. One study demonstrated worse results with the combination in comparison with monotherapies. While specific research findings are reported elsewhere, this article provides an overview of the study designs, insights, and recommendations for future research aimed at therapy development for TBI.


Subject(s)
Brain Injuries/drug therapy , Drug Therapy, Combination/methods , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Disease Models, Animal , Male , Mice , Rats
17.
PLoS One ; 10(6): e0131441, 2015.
Article in English | MEDLINE | ID: mdl-26110872

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and most aggressive malignant brain tumor. Despite optimal treatment and evolving standard of care, the median survival of patients diagnosed with GBM is only 12-15 months. In this study, we combined progesterone (PROG) and temozolomide (TMZ), a standard chemotherapeutic agent for human GBM, to test whether PROG enhances the antitumor effects of TMZ and reduces its side effects. Two WHO grade IV human GBM cells lines (U87MG and U118MG) and primary human dermal fibroblasts (HDFs) were repeatedly exposed to PROG and TMZ either alone or in combination for 3 and 6 days. Cell death was measured by MTT reduction assay. PROG and TMZ individually induced tumor cell death in a dose-dependent manner. PROG at high doses produced more cell death than TMZ alone. When combined, PROG enhanced the cell death-inducing effect of TMZ. In HDFs, PROG did not reduce viability even at the same high cytotoxic doses, but TMZ did so in a dose-dependent manner. In combination, PROG reduced TMZ toxicity in HDFs. PROG alone and in combination with TMZ suppressed the EGFR/PI3K/Akt/mTOR signaling pathway and MGMT expression in U87MG cells, thus suppressing cell proliferation. PROG and TMZ individually reduced cell migration in U87MG cells but did so more effectively in combination. PROG enhances the cytotoxic effects of TMZ in GBM cells and reduces its toxic side effects in healthy primary cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Progesterone/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , DNA Modification Methylases/biosynthesis , DNA Repair Enzymes/biosynthesis , Dacarbazine/adverse effects , Dacarbazine/therapeutic use , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Humans , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Temozolomide , Tumor Suppressor Proteins/biosynthesis
18.
Brain Inj ; 29(10): 1165-1174, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26083048

ABSTRACT

OBJECTIVE: Inflammation is an important component of the response to traumatic brain injury (TBI). Progesterone has been shown to inhibit neuroinflammation following (TBI) and may do so through Toll-like receptor (TLR)-mediated pathways. In vitro studies indicate that 1,25-dihydroxyvitamin D(3) (VDH) may also modulate the inflammatory response through the TLR4 pathway. This study tested the hypothesis that PROG and VDH would exert additive and synergistic neuroprotective effects compared with individual treatment by modulating TLR4/NF-κB-mediated inflammation pathways after TBI in rats. RESEARCH DESIGN AND METHODS: Bilateral medial frontal cortical impact injury was induced in young adult Sprague-Dawley rats. Progesterone (i.p., 16 mg kg-1 body weight) and VDH (1 µg kg-1 body weight) were injected separately or combined at 1 and 6 hours after surgery. Rats were killed 24 hours post-surgery and peri-contusional brain tissue harvested for immunostaining and protein measurement. RESULTS: TLR4, phosphorylation of NF-κB, neuronal loss and astrocyte activation were significantly reduced with combination treatment after TBI compared to each agent given individually. CONCLUSIONS: At 24 hours after TBI, combination therapy shows greater efficacy in reducing neuroinflammation compared to progesterone and VDH given separately, and does so by modulating the TLR4/NF-κB signalling pathway.

19.
Restor Neurol Neurosci ; 33(3): 251-61, 2015.
Article in English | MEDLINE | ID: mdl-25624424

ABSTRACT

PURPOSE: Most pre-clinical stroke studies address the acute phase after injury, with less attention to long-term effects of injury, treatment, and experimental testing itself. We addressed these questions: 1) Will functional deficits persist up to 8 weeks following transient stroke in older animals? 2) Will functional deficits resolve spontaneously, with time and/or repeated behavioral testing? METHODS: Male Sprague-Dawley rats (12 months) were pre-trained on behavioral tasks to provide baseline data and then underwent transient middle artery occlusion (tMCAO) or sham surgery. We measured motor, sensory, cognitive and gait impairments over 8 weeks, and the extent of hemispheric brain infarction. One cohort underwent behavioral testing once at 8 weeks post-stroke (LT); a second cohort (RLT) was tested at 3, 6 and 8 weeks post-stroke. RESULTS: Significant deficits were exhibited in all functional outcomes in both cohorts after 8 weeks. We observed some recovery in some behavioral parameters in both cohorts at 8 weeks. CONCLUSIONS: Deficits persist for at least 8 weeks after tMCAO. The greater spontaneous recovery seen in the RLT groups suggest that repeated testing did reduce the severity of these stroke-induced impairments. These findings have implications for designing future studies of agents to induce long-term functional recovery following stroke.


Subject(s)
Behavior, Animal/physiology , Brain/physiopathology , Cognition/physiology , Ischemic Attack, Transient/physiopathology , Motor Activity/physiology , Recovery of Function/physiology , Stroke/physiopathology , Animals , Gait/physiology , Male , Rats , Rats, Sprague-Dawley
20.
J Steroid Biochem Mol Biol ; 146: 62-73, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24787660

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive primary brain tumor with a mean patient survival of 13-15 months despite surgical resection, radiation therapy and standard-of-care chemotherapy. We investigated the chemotherapeutic effects of the hormone progesterone (P4) on the growth of human GBM in four genetically different cell lines (U87MG, U87dEGFR, U118MG, LN-229) in vitro and in a U87MG subcutaneous xenograft mouse model. At high concentrations (20, 40, and 80 µM), P4 significantly (P<0.05) decreased tumor cell viability in all cell lines except LN-229. This effect was not blocked by the P4 receptor antagonist RU468. Conversely, at low physiological concentrations (0.1, 1, and 5 µM) P4 showed a proliferative effect in all cell lines which was blocked by RU486. In nude mice, P4 (100 and 200 mg/kg) inhibited tumor growth significantly (P<0.05) over 5 weeks of treatment and extended survival time of tumor-bearing mice by 60% without signs of systemic toxicity. P4 suppressed tumor vascularization as indicated by the expression of CD31, vascular endothelial growth factor and matrix metalloproteinase-9. Apoptosis in tumor tissue was detected by the expression of cleaved caspase-3, BCl-2, BAD and p53 proteins and confirmed by TUNEL assay. P4 treatment also suppressed PI3K/Akt/mTOR signaling, which regulates tumor growth, as demonstrated by the suppression of proliferating cell nuclear antigen. Our data can be interpreted to suggest that P4 suppresses the growth of human GBM cells both in vitro and in vivo and enhances survival time in mice without any demonstrable side effects. This article is part of a Special Issue entitled 'Sex steroids and brain disorders'.


Subject(s)
Glioblastoma/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Progesterone/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice, Nude , PTEN Phosphohydrolase/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Progesterone/administration & dosage , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...