Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 324(2): H212-H225, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36563009

RESUMEN

Diabetes increases the risk of poststroke cognitive impairment (PSCI). Greater hemorrhagic transformation (HT) after stroke is associated with vasoregression and cognitive decline in male diabetic rats. Iron chelator deferoxamine (DFX) prevents vasoregression and improves outcomes. Although diabetic female rats develop greater HT, its impact on poststroke cerebrovascularization and cognitive outcomes remained unknown. We hypothesized that diabetes mediates pathological neovascularization, and DFX attenuates poststroke cerebrovascular remodeling and improves neurological outcomes in female diabetic rats. Female control and diabetic animals were treated with DFX or vehicle for 7 days after stroke. Vascular indices, microglial activation, and blood-brain barrier (BBB) integrity were evaluated on day 14. Results from diabetic female rats were partially compared with our previously published findings in male counterparts. Hemin-induced programmed cell death was studied in male and female brain microvascular endothelial cell lines (BMVEC). There was no vasoregression after stroke in either control or diabetic female animals. DFX prevented diabetes-mediated gliovascular remodeling and compromised BBB integrity while improving memory function in diabetes. Comparisons of female and male rats indicated sex differences in cognitive and vascular outcomes. Hemin mediated ferroptosis in both male and female BMVECs. DFX improved survival but had differential effects on ferroptosis signaling in female and male cells. These results suggest that stroke and associated HT do not affect cerebrovascularization in diabetic female rats, but iron chelation may provide a novel therapeutic strategy in the prevention of poststroke memory impairment in females with diabetes via the preservation of gliovascular integrity and improvement of endothelial cell survival.NEW & NOTEWORTHY The current study shows for the first time that diabetes does not promote aberrant cerebrovascularization in female rats. This contrasts with what we reported in male animals in various diabetes models. Deferoxamine preserved recognition memory function in diabetic female animals after stroke. The effect(s) of stroke and deferoxamine on cerebrovascular density and microglial activation also appear(s) to be different in female diabetic rats. Lastly, deferoxamine exerts detrimental effects on animals and BMVECs under control conditions.


Asunto(s)
Diabetes Mellitus Experimental , Ferroptosis , Accidente Cerebrovascular , Ratas , Femenino , Masculino , Animales , Deferoxamina/farmacología , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Hemina/farmacología , Accidente Cerebrovascular/complicaciones
2.
Can J Physiol Pharmacol ; 100(12): 1087-1096, 2022 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-36384316

RESUMEN

Cellular senescence plays a pivotal role in the aging and progression of neurodegenerative diseases, including vascular cognitive impairment and dementia (VCID). In postmortem brains from individuals with VCID, endothelin-1 (ET-1) levels closely correlate with blood barrier breakdown and cerebral hypoperfusion. Brain microvascular endothelial cells (BMVECs), previously thought to have exclusively endothelin B receptors, also possess endothelin A (ETA) receptors; however, the functional significance of this receptor in BMVECs is not known. We hypothesize that ETA receptors mediate BMVEC senescence. Serum-starved human BMVECs (HBEC5i) were incubated with ET-1 (1 µmol/L) in the presence/absence of ETA receptor antagonist BQ-123 (20 µmol/L). Cells were collected for Western blot and quantitative real-time PCR analyses. Treatment of ET-1 increased protein expression of ETA receptor, while it was prevented by the ETA receptor antagonist. ET-1 increased p21, p16, p53, LIF1 and cyclin D1 protein levels, and ß-galactosidase accumulation, which were prevented in the presence of ETA blockade. While there was no change in tight junction proteins, ET-1 decreased adherent junction protein vascular endothelial cadherin (VE-cadherin) levels. In conclusion, ET-1 upregulates ETA receptors in BMVECs in an autocrine manner and triggers the activation of senescence. These in vitro findings need to be further studied in vivo to establish the role of ETA receptors in the progression of endothelial senescence in VCID.


Asunto(s)
Demencia Vascular , Células Endoteliales , Humanos , Receptor de Endotelina A , Encéfalo , Endotelina-1
3.
Clin Sci (Lond) ; 136(21): 1555-1570, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36314470

RESUMEN

Diabetes doubles the risk of vascular cognitive impairment, but the underlying reasons remain unclear. In the present study, we determined the temporal and spatial changes in the brain structure after microemboli (ME) injection using diffusion MRI (dMRI). Control and diabetic rats received cholesterol crystal ME (40-70 µm) injections. Cognitive tests were followed up to 16 weeks, while dMRI scans were performed at baseline and 12 weeks post-ME. The novel object recognition test had a lower d2 recognition index along with a decrease in spontaneous alternations in the Y maze test in diabetic rats with ME. dMRI showed that ME injection caused infarction in two diabetic animals (n=5) but none in controls (n=6). In diabetes, radial diffusivity (DR) was increased while fractional anisotropy (FA) was decreased in the cortex, indicating loss of tissue integrity and edema. In the dorsal hippocampus, mean diffusivity (MD), axial diffusivity (DA), and DR were significantly increased, indicating loss of axons and myelin damage. Histological analyses confirmed more tissue damage and microglial activation in diabetic rats with ME. These results suggest that ME injury and associated cerebrovascular dysfunction are greater in diabetes, which may cause cognitive deficits. Strategies to improve vascular function can be a preventive and therapeutic approach for vascular cognitive impairment.


Asunto(s)
Disfunción Cognitiva , Demencia Vascular , Diabetes Mellitus Experimental , Sustancia Blanca , Animales , Ratas , Sustancia Blanca/patología , Disfunción Cognitiva/patología , Encéfalo/patología , Imagen por Resonancia Magnética
4.
Front Physiol ; 13: 864758, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35574460

RESUMEN

Stress-induced premature senescence can contribute to the accelerated metabolic aging process in diabetes. Progressive accumulation of senescent cells in the brain, especially those displaying the harmful inflammatory senescence-associated secretory phenotype (SASP), may lead to cognitive impairment linked with metabolic disturbances. In this context, the senescence within the neurovascular unit (NVU) should be studied as much as in the neurons as emerging evidence shows that neurogliovascular communication is critical for brain health. It is also known that cerebrovascular dysfunction and decreased cerebral blood flow (CBF) precede the occurrence of neuronal pathologies and overt cognitive impairment. Various studies have shown that endothelial cells, the major component of the NVU, acquire a senescent phenotype via various molecular mediators and pathways upon exposure to high glucose and other conditions mimicking metabolic disturbances. In addition, senescence in the other cells that are part of the NVU, like pericytes and vascular smooth cells, was also triggered upon exposure to diabetic conditions. The senescence within the NVU may compromise functional and trophic coupling among glial, vascular, and neuronal cells and the resulting SASP may contribute to the chronic neurovascular inflammation observed in Alzheimer's Disease and Related Dementias (ADRD). The link between diabetes-mediated cerebral microvascular dysfunction, NVU senescence, inflammation, and cognitive impairment must be widely studied to design therapeutic strategies.

5.
Neuromolecular Med ; 23(3): 344-347, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33486699

RESUMEN

Following traumatic brain injury (TBI), increased production of reactive oxygen species (ROS) and the ensuing oxidative stress promotes the secondary brain damage that encompasses both grey matter and white matter. As this contributes to the long-term neurological deficits, decreasing oxidative stress during the acute period of TBI is beneficial. While NADPH oxidase (NOX2) is the major producer of ROS, transcription factor Nrf2 that induces antioxidant enzymes promotes efficient ROS disposal. We recently showed that treatment with an antioxidant drug combo of apocynin (NOX2 inhibitor) and TBHQ (Nrf2 activator) protects the grey matter in adult mice subjected to TBI. We currently show that this antioxidant combo therapy given at 2 h and 24 h after TBI also protects white matter in mouse brain. Thus, the better functional outcomes after TBI in the combo therapy treated mice might be due to a combination of sparing both grey matter and white matter. Hence, the antioxidant combo we tested is a potent therapeutic option for translation in future.


Asunto(s)
Acetofenonas/uso terapéutico , Antioxidantes/uso terapéutico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Hidroquinonas/uso terapéutico , Sustancia Blanca/efectos de los fármacos , Acetofenonas/administración & dosificación , Animales , Antioxidantes/administración & dosificación , Lesiones Traumáticas del Encéfalo/patología , Esquema de Medicación , Evaluación Preclínica de Medicamentos , Sinergismo Farmacológico , Quimioterapia Combinada , Sustancia Gris/efectos de los fármacos , Sustancia Gris/patología , Hidroquinonas/administración & dosificación , Masculino , Ratones , Ratones Endogámicos C57BL , NADPH Oxidasa 2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/agonistas , Estrés Oxidativo/efectos de los fármacos , Distribución Aleatoria , Sustancia Blanca/patología
6.
Brain Res ; 1749: 147132, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33002484

RESUMEN

Vascular disease plays an important role in all kinds of cognitive impairment and dementia. Diabetes increases the risk of vascular disease and dementia. However, it is not clear how existing vascular disease in the brain accelerates the development of small vessel disease and promotes cognitive dysfunction in diabetes. We used microemboli (ME) injection model in the current study to test the hypothesis that cerebrovascular dysfunction in diabetes facilitates entrapment of ME leading to inflammation and cognitive decline. We investigated cognitive function, axonal/white matter (WM) changes, neurovascular coupling, and microglial activation in control and diabetic male and female Wistar rats subjected to sham or low/high dose ME injection. Diabetic male animals had cognitive deficits, WM demyelination and greater microglial activation than the control animals even at baseline. Functional hyperemia gradually declined in diabetic male animals after ME injection. Both low and high ME injection worsened WM damage and increased microglial activation in diabetic male and female animals. Low ME did not cause cognitive decline in controls, while promoting learning/memory deficits in diabetic female rats and no further decline in diabetic male animals. High ME led to cognitive decline in control male rats and exacerbated the deficits in diabetic cohort. These results suggest that the existing cerebrovascular dysfunction in diabetes may facilitate ME-mediated demyelination leading to cognitive decline. It is important to integrate comorbidities/sex as a biological variable into experimental models for the development of preventive or therapeutic targets.


Asunto(s)
Disfunción Cognitiva/etiología , Demencia Vascular/complicaciones , Sustancia Blanca/patología , Animales , Disfunción Cognitiva/patología , Demencia Vascular/patología , Diabetes Mellitus Experimental/patología , Femenino , Masculino , Microglía/patología , Acoplamiento Neurovascular/fisiología , Ratas , Ratas Wistar
7.
Neurochem Int ; 139: 104788, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32650031

RESUMEN

Enolase inhibition is a potential therapeutic strategy currently being investigated for treatment of spinal cord injury (SCI) as it reduces pro-inflammatory cytokines and chemokines, alters metabolic factors, and reduces gliosis in acute SCI. Herein, the role of enolase in SCI has been examined to better understand the effects of this enzyme on inflammation, metabolic hormones, glial cell activation, and neuroprotection under these shorter injury conditions. Immunohistochemical analyses of inflammatory markers vimentin, Cox-2, and caspase-1 indicated that enolase inhibition attenuated the elevated levels of inflammation seen following SCI. Iba1, GFAP, NFP, and CSPG staining indicated that enolase inhibition with prolonged administration of ENOblock reduced microglia/astrocyte activation and lead to enhanced neuroprotection in SCI. An analysis of metabolic hormones revealed that ENOblock treatment significantly upregulated plasma concentrations of peptide YY, glucagon-like peptide 1, glucose-dependent insulinotropic peptide, glucagon, and insulin hormones as compared to vehicle-treated controls (Mann-Whitney, p ≤ 0.05). ENOblock did not have a significant effect on plasma concentrations of pancreatic polypeptide. Interestingly, ENOblock treatment inhibited chondroitin sulfate proteoglycan (CSPG), which is produced by activated glia and serves to block regrowth of axons across the lesion site following injury. An increased level of NeuN and MBP with reduced caspase-1 was detected in SCI tissues after ENOblock treatment, suggesting preservation of myelin and induction of neuroprotection. ENOblock also induced improved motor function in SCI rats, indicating a role for enolase in modulating inflammatory and metabolic factors in SCI with important implications for clinical consideration.


Asunto(s)
Benzamidas/uso terapéutico , Mediadores de Inflamación/antagonistas & inhibidores , Neuroprotección/efectos de los fármacos , Hormonas Peptídicas/agonistas , Fosfopiruvato Hidratasa/antagonistas & inhibidores , Traumatismos de la Médula Espinal/tratamiento farmacológico , Triazinas/uso terapéutico , Animales , Benzamidas/farmacología , Hormonas , Mediadores de Inflamación/metabolismo , Masculino , Neuroprotección/fisiología , Hormonas Peptídicas/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/enzimología , Vértebras Torácicas/lesiones , Triazinas/farmacología
8.
Exp Neurol ; 330: 113315, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32302678

RESUMEN

Parkinson's disease (PD), a debilitating progressive degenerative movement disorder associated with loss of dopaminergic (DA) neurons in the substantia nigra (SN), afflicts approximately one million people in the U.S., including a significant number of Veterans. Disease characteristics include tremor, rigidity, postural instability, bradykinesia, and at a cellular level, glial cell activation and Lewy body inclusions in DA neurons. The most potent medical/surgical treatments do not ultimately prevent disease progression. Therefore, new therapies must be developed to halt progression of the disease. While the mechanisms of the degenerative process in PD remain elusive, chronic inflammation, a common factor in many neurodegenerative diseases, has been implicated with associated accumulation of toxic aggregated α-synuclein in neurons. Calpain, a calcium-activated cysteine neutral protease, plays a pivotal role in SN and spinal cord degeneration in PD via its role in α-synuclein aggregation, activation/migration of microglia and T cells, and upregulation of inflammatory processes. Here we report an increased expression of a subset of CD4+ T cells in rodent models of PD, including MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mice and DSP-4 [N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine hydrochloride]/6-hydroxydopamine rats, which produced higher levels of perforin and granzyme B - typically found in cytotoxic T cells. Importantly, the CD4+ cytotoxic subtype was attenuated following calpain inhibition in MPTP mice, suggesting that calpain and this distinct CD4+ T cell subset may have critical roles in the inflammatory process, disease progression, and neurodegeneration in PD.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Calpaína/inmunología , Trastornos Parkinsonianos/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Calpaína/metabolismo , Modelos Animales de Enfermedad , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Trastornos Parkinsonianos/patología , Ratas , Ratas Long-Evans , Subgrupos de Linfocitos T/inmunología
9.
Neurochem Res ; 43(12): 2224-2231, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30291537

RESUMEN

The cytokine/chemokine expression signature of a 60-year-old African American male with relapsing-remitting multiple sclerosis (RRMS) was analyzed using patient blood samples obtained from two separate visits to the clinic. Thirty-six different cytokines, chemokines, and growth factors were detected in the plasma of the RRMS patient using a multiplexed bead-based immunoassay. Results indicated that at least ten of these factors with a concentration of > 100 pg/mL are identified as pro-inflammatory. Calpain inhibition led to an anti-inflammatory effect, as indicated by a decrease in expression of pro-inflammatory cytokines/chemokines such as GM-CSF, IFNγ, and IL-17A, and a relative increase in two of the anti-inflammatory cytokines (IL-13 and IL-4) in the peripheral blood mononuclear cells activated with anti-CD3/CD28. Overall, these results suggest that the unique cytokine/chemokine pattern observed in the plasma of the RRMS patient can be used as a prognostic marker and calpain inhibition may be used as a novel therapeutic strategy for treating excessive inflammatory response specific to RRMS patients.


Asunto(s)
Quimiocinas/biosíntesis , Inhibidores de Cisteína Proteinasa/uso terapéutico , Citocinas/biosíntesis , Dipéptidos/uso terapéutico , Leucocitos Mononucleares/metabolismo , Esclerosis Múltiple Recurrente-Remitente/sangre , Quimiocinas/genética , Inhibidores de Cisteína Proteinasa/farmacología , Citocinas/genética , Dipéptidos/farmacología , Expresión Génica , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico
10.
J Cereb Blood Flow Metab ; 38(10): 1818-1827, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29083257

RESUMEN

Uncontrolled oxidative stress contributes to the secondary neuronal death that promotes long-term neurological dysfunction following traumatic brain injury (TBI). Surprisingly, both NADPH oxidase 2 (NOX2) that increases and transcription factor Nrf2 that decreases reactive oxygen species (ROS) are induced after TBI. As the post-injury functional outcome depends on the balance of these opposing molecular pathways, we evaluated the effect of TBI on the motor and cognitive deficits and cortical contusion volume in NOX2 and Nrf2 knockout mice. Genetic deletion of NOX2 improved, while Nrf2 worsened the post-TBI motor function recovery and lesion volume indicating that decreasing ROS levels might be beneficial after TBI. Treatment with either apocynin (NOX2 inhibitor) or TBHQ (Nrf2 activator) alone significantly improved the motor function after TBI, but had no effect on the lesion volume, compared to vehicle control. Whereas, the combo therapy (apocynin + TBHQ) given at either 5 min/24 h or 2 h/24 h improved motor and cognitive function and decreased cortical contusion volume compared to vehicle group. Thus, both the generation and disposal of ROS are important modulators of oxidative stress, and a combo therapy that prevents ROS formation and potentiates ROS disposal concurrently is efficacious after TBI.


Asunto(s)
Antioxidantes/farmacología , Lesiones Traumáticas del Encéfalo/patología , NADPH Oxidasa 2/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/agonistas , Recuperación de la Función/efectos de los fármacos , Acetofenonas/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/metabolismo , Hidroquinonas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasa 2/deficiencia , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Recuperación de la Función/fisiología
11.
Neurochem Int ; 111: 12-22, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28131900

RESUMEN

Accumulating evidence indicates that various classes of non-coding RNAs (ncRNAs) including microRNAs (miRNAs), PIWI-interacting RNAs (piRNAs) and long non-coding RNAs (lncRNAs) play important roles in normal state as well as the diseases of the CNS. Interestingly, ncRNAs have been shown to interact with messenger RNA, DNA and proteins, and these interactions could induce epigenetic modifications and control transcription and translation, thereby adding a new layer of genomic regulation. The ncRNA expression profiles are known to be altered after acute CNS injuries including stroke, traumatic brain injury and spinal cord injury that are major contributors of morbidity and mortality worldwide. Hence, a better understanding of the functional significance of ncRNAs following CNS injuries could help in developing potential therapeutic strategies to minimize the neuronal damage in those conditions. The potential of ncRNAs in blood and CSF as biomarkers for diagnosis and/or prognosis of acute CNS injuries has also gained importance in the recent years. This review highlighted the current progress in the understanding of the role of ncRNAs in initiation and progression of secondary neuronal damage and their application as biomarkers after acute CNS injuries.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Neuroprotección , ARN no Traducido/genética , Accidente Cerebrovascular/genética , Animales , Biomarcadores/líquido cefalorraquídeo , Humanos , Isquemia/metabolismo , ARN no Traducido/líquido cefalorraquídeo , Accidente Cerebrovascular/diagnóstico
12.
Brain Inj ; 31(1): 106-119, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27819514

RESUMEN

OBJECTIVE: To investigate the effect of heterogeneity in mTBI on miRNA expression in mouse brain and to identify molecular pathways targeted by the modulated miRNAs. METHODS: A weight drop device was used to induce four increasing grades of mTBI. MiRNA expression was evaluated using TaqMan rodent miRNA arrays. Bioinformatics analysis was done using the DIANA miRPath tool and Ingenuity Pathway Analysis software. Histology of brain sections was evaluated using H&E staining. RESULTS: No histologic lesions were observed in the brains of injured mice; however, significant modulation in miRNA expression profile was observed. Global miRNA profiling indicated a trend of decrease in the number of modulated miRNAs from 24 hours to day 7 post-injury, except for the most severe grade of mTBI. Canonical pathways like calcium signalling, synaptic pathways and axon guidance pathway were the major targets of the modulated miRNAs. Network correlation analyses indicated an interaction between the modulated miRNAs and putative protein biomarkers of TBI. CONCLUSIONS: The data demonstrated that varying intensities of mTBI induced a differential miRNA expression profile in the brain post-injury. Pathways such as calcium and synaptic signalling were major targets of modulated miRNAs and may play a role in the pathophysiology of mTBI.


Asunto(s)
Conmoción Encefálica/metabolismo , Encéfalo/metabolismo , MicroARNs/metabolismo , Animales , Conmoción Encefálica/genética , Masculino , Ratones , MicroARNs/genética , Modelos Animales , Transducción de Señal/fisiología
13.
PLoS One ; 9(11): e112019, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25379886

RESUMEN

Wars in Iraq and Afghanistan have highlighted the problems of diagnosis and treatment of mild traumatic brain injury (mTBI). MTBI is a heterogeneous injury that may lead to the development of neurological and behavioral disorders. In the absence of specific diagnostic markers, mTBI is often unnoticed or misdiagnosed. In this study, mice were induced with increasing levels of mTBI and microRNA (miRNA) changes in the serum were determined. MTBI was induced by varying weight and fall height of the impactor rod resulting in four different severity grades of the mTBI. Injuries were characterized as mild by assessing with the neurobehavioral severity scale-revised (NSS-R) at day 1 post injury. Open field locomotion and acoustic startle response showed behavioral and sensory motor deficits in 3 of the 4 injury groups at day 1 post injury. All of the animals recovered after day 1 with no significant neurobehavioral alteration by day 30 post injury. Serum microRNA (miRNA) profiles clearly differentiated injured from uninjured animals. Overall, the number of miRNAs that were significantly modulated in injured animals over the sham controls increased with the severity of the injury. Thirteen miRNAs were found to identify mTBI regardless of its severity within the mild spectrum of injury. Bioinformatics analyses revealed that the more severe brain injuries were associated with a greater number of miRNAs involved in brain related functions. The evaluation of serum miRNA may help to identify the severity of brain injury and the risk of developing adverse effects after TBI.


Asunto(s)
Lesiones Encefálicas/sangre , Lesiones Encefálicas/diagnóstico , Traumatismos Cerrados de la Cabeza/complicaciones , MicroARNs/sangre , MicroARNs/genética , Animales , Conducta Animal , Encéfalo/fisiopatología , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/genética , Biología Computacional , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Reflejo de Sobresalto
14.
J Psychiatr Res ; 57: 65-73, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24998397

RESUMEN

Exposure to acute traumatic stress can cause permanent changes in neurological circuitry and may lead to the development of an anxiety disorder known as posttraumatic stress disorder (PTSD). Current diagnosis of PTSD is based on clinical or behavioral symptom assessment, however, these are not definitive due to overlapping symptoms with other psychiatric disorders or mild traumatic brain injury (mTBI). No FDA approved diagnostic tests or biomarkers are currently available for diagnosis of PTSD. Recently, circulating miRNAs have emerged as novel biomarkers of many diseases. In this study, we have examined the altered expression of serum and amygdala miRNAs in an animal model of PTSD. Differentially expressed and statistically significant miRNAs in serum were validated for their presence in amygdala of corresponding animals. A panel of nine stress-responsive miRNAs viz., miR-142-5p, miR-19b, miR-1928, miR-223-3p, miR-322∗, miR-324, miR-421-3p and miR-463∗ and miR-674∗ were identified, and may have potential as biomarker(s) for PTSD. Further validations by bioinformatics and system biology approaches indicate that five miRNAs such as miR-142-5p, miR-19b, miR-1928, miR-223 and miR-421-3p may play a potential role in the regulation of genes associated with delayed and exaggerated fear. To the best of our knowledge, this is the first report demonstrating the plausibility of using circulating miRNAs as biomarkers of PTSD.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Biomarcadores/sangre , Miedo/psicología , MicroARNs/genética , Trastornos por Estrés Postraumático/genética , Estrés Psicológico/genética , Animales , Modelos Animales de Enfermedad , Masculino , MicroARNs/sangre , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/psicología , Estrés Psicológico/complicaciones , Estrés Psicológico/psicología
15.
J Neurotrauma ; 29(7): 1379-87, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22352906

RESUMEN

Blast-induced traumatic brain injury (TBI) is of significant concern in soldiers returning from the current conflicts in Iraq and Afghanistan. Incidents of TBI have increased significantly in the current conflicts compared to previous wars, and a majority of these injuries are caused by improvised explosive devices. Currently, no specific technique or biomarker is available for diagnosing TBI when no obvious clinical symptoms are present. Micro-RNAs are small RNA (~ 22nts) molecules that are expressed endogenously and play an important role in regulating gene expression. MicroRNAs have emerged as novel serum diagnostic biomarkers for various diseases. In this study, we studied the effect of blast overpressure injury on the microRNA signatures in the serum of rats. Rats were exposed to three serial 120-kPa blast overpressure exposures through a shockwave tube. Blood and cerebrospinal fluid were collected at various time points after injury, and microRNA modulation was analyzed using real-time PCR. Five microRNAs were significantly modulated in the serum samples of these animals at three time points post-injury. Further, we also found that the levels of microRNA let-7i are also elevated in cerebrospinal fluid post-blast wave exposure. The presence of microRNA in both serum and cerebrospinal fluid immediately after injury makes microRNA let-7i an ideal candidate for further studies of biomarkers in TBI.


Asunto(s)
Traumatismos por Explosión/diagnóstico , Traumatismos por Explosión/genética , Lesiones Encefálicas/diagnóstico , MicroARNs/sangre , Animales , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Traumatismos por Explosión/sangre , Lesiones Encefálicas/sangre , Lesiones Encefálicas/líquido cefalorraquídeo , Modelos Animales de Enfermedad , Masculino , MicroARNs/biosíntesis , MicroARNs/líquido cefalorraquídeo , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA