Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cureus ; 14(7): e27296, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36043003

RESUMEN

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.
Artículo en Inglés | MEDLINE | ID: mdl-33847913

RESUMEN

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.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Glioblastoma/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Progesterona/farmacología , Animales , Línea Celular Tumoral , Metabolismo Energético/fisiología , Glioblastoma/tratamiento farmacológico , Humanos , Consumo de Oxígeno/efectos de los fármacos , Consumo de Oxígeno/fisiología , Progesterona/uso terapéutico , Smegmamorpha
3.
Mol Neurobiol ; 58(3): 950-963, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33063282

RESUMEN

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.


Asunto(s)
Progesterona/farmacología , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/fisiopatología , Deficiencia de Vitamina D/inmunología , Deficiencia de Vitamina D/fisiopatología , Vitamina D/farmacología , Animales , Apoptosis/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Conducta Animal/efectos de los fármacos , Biomarcadores/metabolismo , Ciclooxigenasa 2/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Fuerza de la Mano , Inflamación/sangre , Inflamación/patología , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos , Masculino , Proteína Básica de Mielina/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Reproducibilidad de los Resultados , Bazo/patología , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/complicaciones , Timo/patología , Factor de Transcripción CHOP/metabolismo , Deficiencia de Vitamina D/sangre , Deficiencia de Vitamina D/complicaciones , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
4.
J Stroke Cerebrovasc Dis ; 29(11): 105249, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33066928

RESUMEN

BACKGROUND: Subarachnoid hemorrhage (SAH) results in neurocognitive dysfunction and anxiety in humans and in animal models. Neurobehavioral tests such as the Morris Water Maze (MWM) and Elevated Plus Maze (EPM) tests are validated in several models of SAH but have not been tested in the murine cisternal blood injection SAH model. METHODS: Adult C57BL/6 mice (n=16) were randomized into two groups. Group 1 (n=8) received sham surgery. Group 2 (n=8) underwent SAH with 60 µL of autologous blood injected into the cisterna magna. Mice were then tested using the Modified Garcia Score on post-operative day 2 (POD2), EPM on POD5 & POD16, and MWM on POD6-16.Brain tissues harvested on POD16 were stained with Fluoro-Jade C to identify neurodegeneration in the hippocampus and cortex and Iba-1 immunofluorescence staining for microglial activation in the dentate gyrus and CA1 region of the hippocampus. RESULTS: SAH mice showed increased escape latency on POD10. Swim distance was significantly increased on POD9-10 and swim speed was significantly decreased on POD6&POD10 in SAH mice. SAH mice exhibited a trend for lowered proportion of covered arm entries in EPM on POD16. Modified Garcia Score was not significantly different between the groups on POD2. The area of microglial activation in the dentate gyrus and CA1 region of the hippocampus was mildly increased but not significantly different at day 16 after SAH. Similarly, no significant differences were noted in the number of Fluoro-Jade C (+) cells in cortex or hippocampus. CONCLUSIONS: Cisternal single blood injection in mice produces mild neurocognitive deficits most pronounced in spatial learning and most evident 10 days after SAH.


Asunto(s)
Conducta Animal , Encéfalo/fisiopatología , Aprendizaje por Laberinto , Trastornos Neurocognitivos/etiología , Hemorragia Subaracnoidea/etiología , Animales , Encéfalo/patología , Cisterna Magna , Modelos Animales de Enfermedad , Reacción de Fuga , Inyecciones , Masculino , Ratones Endogámicos C57BL , Degeneración Nerviosa , Trastornos Neurocognitivos/patología , Trastornos Neurocognitivos/fisiopatología , Trastornos Neurocognitivos/psicología , Tiempo de Reacción , Hemorragia Subaracnoidea/patología , Hemorragia Subaracnoidea/fisiopatología , Hemorragia Subaracnoidea/psicología , Natación , Factores de Tiempo
5.
Neuropharmacology ; 181: 108327, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32950558

RESUMEN

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.


Asunto(s)
Factores Inmunológicos/farmacología , Inflamación/prevención & control , Accidente Cerebrovascular Isquémico/patología , Progesterona/farmacología , Vitamina D/farmacología , Vitaminas/farmacología , Animales , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Caspasa 3/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Fuerza de la Mano , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/prevención & control , Inflamación/etiología , Interleucina-6/metabolismo , Accidente Cerebrovascular Isquémico/inmunología , Accidente Cerebrovascular Isquémico/psicología , Masculino , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
6.
Int J Mol Sci ; 21(11)2020 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-32466385

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Autofagia , Isquemia Encefálica/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fármacos Neuroprotectores/farmacología , Progesterona/farmacología , Estrés Psicológico/metabolismo , Animales , Isquemia Encefálica/complicaciones , Células Cultivadas , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/complicaciones
7.
Sci Rep ; 9(1): 988, 2019 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-30700763

RESUMEN

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.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Glioblastoma/patología , Glucólisis/efectos de los fármacos , Progesterona/farmacología , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Biomarcadores/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Pruebas de Enzimas , Glioblastoma/irrigación sanguínea , Luciferasas/metabolismo , Ratones Desnudos , Modelos Biológicos , Actividad Motora/efectos de los fármacos , Neovascularización Patológica/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
9.
World Neurosurg ; 110: e150-e159, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29097330

RESUMEN

BACKGROUND: Subarachnoid hemorrhage (SAH) induces widespread inflammation leading to cellular injury, vasospasm, and ischemia. Evidence suggests that progesterone (PROG) can improve functional recovery in acute brain injury owing to its anti-inflammatory and neuroprotective properties, which could also be beneficial in SAH. We hypothesized that PROG treatment attenuates inflammation-mediated cerebral vasospasm and microglial activation, improves synaptic connectivity, and ameliorates functional recovery after SAH. METHODS: We investigated the effect of PROG in a cisternal SAH model in adult male C57BL/6 mice. Neurobehavioral outcomes were evaluated using rotarod latency and grip strength tests. Basilar artery perimeter, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptor 1 (GluR1)/synaptophysin colocalization, and Iba-1 immunoreactivity were quantified histologically. RESULTS: PROG (8 mg/kg) significantly improved rotarod latency at day 6 and grip strength at day 9. PROG-treated mice had significantly reduced basilar artery vasospasm at 24 hours. GluR1/synaptophysin colocalization, indicative of synaptic GluR1, was significantly reduced in the SAH+Vehicle group at 24 hours, and PROG treatment significantly attenuated this reduction. PROG treatment significantly reduced microglial cell activation and proliferation in cerebellum and cortex but not in the brainstem at 10 days. CONCLUSIONS: PROG treatment ameliorated cerebral vasospasm, reduced microglial activation, restored synaptic GluR1 localization, and improved neurobehavioral performance in a murine model of SAH. These results provide a rationale for further translational testing of PROG therapy in SAH.


Asunto(s)
Antiinflamatorios/farmacología , Fármacos Neuroprotectores/farmacología , Progesterona/farmacología , Hemorragia Subaracnoidea/tratamiento farmacológico , Vasoespasmo Intracraneal/tratamiento farmacológico , Animales , Arteria Basilar/efectos de los fármacos , Arteria Basilar/inmunología , Arteria Basilar/patología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Encéfalo/fisiopatología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/patología , Microglía/fisiología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Fuerza Muscular/efectos de los fármacos , Fuerza Muscular/fisiología , Distribución Aleatoria , Receptores AMPA/metabolismo , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Hemorragia Subaracnoidea/patología , Hemorragia Subaracnoidea/fisiopatología , Sinaptofisina/metabolismo , Vasoespasmo Intracraneal/patología , Vasoespasmo Intracraneal/fisiopatología
10.
Horm Behav ; 96: 21-30, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28866326

RESUMEN

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.


Asunto(s)
Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/prevención & control , Cognición/efectos de los fármacos , Irradiación Craneana/efectos adversos , Fármacos Neuroprotectores/farmacología , Progesterona/farmacología , Animales , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/etiología , Lesiones Encefálicas/prevención & control , Irradiación Craneana/psicología , Hipocampo/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Microglía/citología , Microglía/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Calidad de Vida , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/prevención & control , Resultado del Tratamiento
11.
Brain Behav Immun ; 66: 177-192, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28648389

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/metabolismo , Encefalitis/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Progesterona/administración & dosificación , Estrés Psicológico/metabolismo , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/patología , Polaridad Celular , Depresión/complicaciones , Encefalitis/complicaciones , Encefalitis/tratamiento farmacológico , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Mediadores de Inflamación/metabolismo , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas Sprague-Dawley , Estrés Psicológico/complicaciones , Estrés Psicológico/patología
12.
Neuroscience ; 350: 1-12, 2017 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-28315443

RESUMEN

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.


Asunto(s)
Glucemia/efectos de los fármacos , Sistema Nervioso Central/efectos de los fármacos , Diabetes Mellitus Experimental/patología , Hiperglucemia/metabolismo , Progesterona/farmacología , Nervio Ciático/efectos de los fármacos , Animales , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Modelos Animales de Enfermedad , Hiperglucemia/inducido químicamente , Interleucina-6/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Progesterona/metabolismo , Ratas Sprague-Dawley , Nervio Ciático/metabolismo , Nervio Ciático/patología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Estreptozocina , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Horm Behav ; 84: 29-40, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27283379

RESUMEN

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.


Asunto(s)
Infarto de la Arteria Cerebral Media/patología , Ataque Isquémico Transitorio/patología , Fármacos Neuroprotectores/farmacología , Progesterona/farmacología , Accidente Cerebrovascular/patología , Animales , Apoptosis/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Infarto de la Arteria Cerebral Media/metabolismo , Ataque Isquémico Transitorio/metabolismo , Masculino , Microglía/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Accidente Cerebrovascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Neuropharmacology ; 107: 317-328, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27039043

RESUMEN

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.


Asunto(s)
Isquemia Encefálica/metabolismo , Factor Neurotrófico Derivado del Encéfalo/fisiología , Progesterona/uso terapéutico , Receptor trkB/fisiología , Transducción de Señal/fisiología , Accidente Cerebrovascular/metabolismo , Animales , Animales Recién Nacidos , Isquemia Encefálica/tratamiento farmacológico , Femenino , Masculino , Ratones , Progesterona/farmacología , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Resultado del Tratamiento
16.
PLoS One ; 10(6): e0131441, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26110872

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Progesterona/uso terapéutico , Antiinflamatorios/uso terapéutico , Antineoplásicos Alquilantes/uso terapéutico , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Metilasas de Modificación del ADN/biosíntesis , Enzimas Reparadoras del ADN/biosíntesis , Dacarbazina/efectos adversos , Dacarbazina/uso terapéutico , Sinergismo Farmacológico , Receptores ErbB/antagonistas & inhibidores , Humanos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Temozolomida , Proteínas Supresoras de Tumor/biosíntesis
17.
Brain Inj ; 29(10): 1165-1174, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26083048

RESUMEN

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.

18.
Restor Neurol Neurosci ; 33(3): 251-61, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25624424

RESUMEN

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.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/fisiopatología , Cognición/fisiología , Ataque Isquémico Transitorio/fisiopatología , Actividad Motora/fisiología , Recuperación de la Función/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Marcha/fisiología , Masculino , Ratas , Ratas Sprague-Dawley
19.
J Steroid Biochem Mol Biol ; 146: 62-73, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24787660

RESUMEN

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'.


Asunto(s)
Glioblastoma/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/metabolismo , Progesterona/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Desnudos , Fosfohidrolasa PTEN/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Progesterona/administración & dosificación , Receptores de Progesterona/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Psychopharmacology (Berl) ; 231(17): 3313-23, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24752655

RESUMEN

RATIONALE: Previous studies demonstrate the neuroprotective effects of progesterone in numerous animal injury models, but a systematic dose-response study in a transient ischemic stroke model is lacking. OBJECTIVES: We investigated the effects of progesterone at different doses on post-stroke brain infarction and functional deficits in middle-aged rats. METHODS: Cerebral ischemia was induced in 13-month-old male Sprague-Dawley rats by right middle cerebral artery occlusion for 2 h followed by reperfusion. Rats received intraperitoneal injections of 8, 16, or 32 mg/kg of progesterone (P8, P16, P32) or vehicle at 2 h post-occlusion followed by subcutaneous injections at 6 h and every 24 h post-injury for 7 days. Functional recovery was evaluated at intervals over 22 days using motor, sensory, and cognitive tests. Infarct size was evaluated at 22 days post-stroke. RESULTS: Repeated-measures ANOVA showed significant group effects on grip strength, rotarod, and sensory neglect. All progesterone-treated groups had improved (p < 0.05) spatial memory performance. The P8 and P16 groups showed maximum improvement in long-term memory compared to vehicle. Significant (p < 0.05) gait impairments were observed in the vehicle group compared to shams. Animals receiving the P8 dose showed maximum gait improvement compared to vehicle. Post hoc analysis revealed that the P8 and P16 groups showed significant attenuation in infarct volume compared to vehicle. Animals receiving the P32 dose did not show any effect on infarct volume. CONCLUSIONS: Although all doses were somewhat effective, progesterone given at 8 mg/kg led to the most consistent improvements across a panel of behavioral/functional tests and reduced the severity of ischemic infarct injury.


Asunto(s)
Ataque Isquémico Transitorio/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Progesterona/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Trastornos Neurológicos de la Marcha/tratamiento farmacológico , Fuerza de la Mano , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/psicología , Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/psicología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Fármacos Neuroprotectores/administración & dosificación , Trastornos de la Percepción/tratamiento farmacológico , Trastornos de la Percepción/psicología , Equilibrio Postural/efectos de los fármacos , Progesterona/administración & dosificación , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...