Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neurosci ; 44(11)2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38326036

RESUMEN

Intercellular adhesion molecule-1 (ICAM-1) is identified as an initiator of neuroinflammatory responses that lead to neurodegeneration and cognitive and sensory-motor deficits in several pathophysiological conditions including traumatic brain injury (TBI). However, the underlying mechanisms of ICAM-1-mediated leukocyte adhesion and transmigration and its link with neuroinflammation and functional deficits following TBI remain elusive. Here, we hypothesize that blocking of ICAM-1 attenuates the transmigration of leukocytes to the brain and promotes functional recovery after TBI. The experimental TBI was induced in vivo by fluid percussion injury (25 psi) in male and female wild-type and ICAM-1-/- mice and in vitro by stretch injury (3 psi) in human brain microvascular endothelial cells (hBMVECs). We treated hBMVECs and animals with ICAM-1 CRISPR/Cas9 and conducted several biochemical analyses and demonstrated that CRISPR/Cas9-mediated ICAM-1 deletion mitigates blood-brain barrier (BBB) damage and leukocyte transmigration to the brain by attenuating the paxillin/focal adhesion kinase (FAK)-dependent Rho GTPase pathway. For analyzing functional outcomes, we used a cohort of behavioral tests that included sensorimotor functions, psychological stress analyses, and spatial memory and learning following TBI. In conclusion, this study could establish the significance of deletion or blocking of ICAM-1 in transforming into a novel preventive approach against the pathophysiology of TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Molécula 1 de Adhesión Intercelular , Animales , Femenino , Humanos , Masculino , Ratones , Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Sistemas CRISPR-Cas , Células Endoteliales/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Leucocitos , Paxillin , Proteínas de Unión al GTP rho/metabolismo
2.
Exp Brain Res ; 241(8): 2107-2123, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37466694

RESUMEN

MicroRNAs (miRNAs) are non-coding RNA molecules that function in RNA silencing and post-transcriptional regulation of gene expression. They are profound mediators of molecular and cellular changes in several pathophysiological conditions. Since miRNAs play major roles in regulating gene expression after traumatic brain injury (TBI), their possible role in diagnosis, prognosis, and therapy is not much explored. In this study, we aimed to identify specific miRNAs that are involved in the pathophysiological conditions in the first 24 h after mild TBI (mTBI). The genome-wide expression of miRNAs was evaluated by applying RNA sequence in the injury area of the cerebral cortex 24 after inflicting the injury using a mouse model of mild fluid percussion injury (FPI; 10 psi). Here, we identified different annotated, conserved, and novel miRNAs. A total of 978 miRNAs after 24 h of TBI were identified, and among these, 906 miRNAs were differentially expressed between control and mTBI groups. In this study, 146 miRNAs were identified as novel to mTBI and among them, 21 miRNAs were significant (p < 0.05). Using q-RT-PCR, we validated 10 differentially and significantly expressed novel miRNAs. Further, we filtered the differentially expressed miRNAs that were linked with proinflammatory cytokines, apoptosis, matrix metalloproteinases (MMPs), and tight junction and junctional adhesion molecule genes. Overall, this work shows that mTBI induces widespread changes in the expression of miRNAs that may underlie the progression of the TBI pathophysiology. The detection of several novel TBI-responsive miRNAs and their solid link with pathophysiological genes may help in identifying novel therapeutic targets.


Asunto(s)
Conmoción Encefálica , Lesiones Traumáticas del Encéfalo , MicroARNs , Humanos , Conmoción Encefálica/genética , MicroARNs/genética , MicroARNs/metabolismo , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/metabolismo , Regulación de la Expresión Génica , Corteza Cerebral/patología
3.
J Integr Neurosci ; 22(4): 81, 2023 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-37519172

RESUMEN

In this review, we discuss the possibility and feasibility of nuclear factor erythroid 2-related factor 2 (Nrf2) as a therapeutic target to minimize the devastating effects of a brain injury. To complete this review, comprehensive literature searches were conducted in MEDLINE, PubMed, Embase, and PsycINFO databases for English scientific peer-reviewed articles through December 2022. This short review addressed the different sources of oxidative stress and its effects on blood-brain barrier (BBB) dysfunction, mitochondrial damage, and changes in a variety of inflammatory molecules associated with central nervous system (CNS) injury. At last, we explained the potential efficacy of the Nrf2 transcription factor in reducing oxidative stress-mediated secondary damages after a CNS injury. The role of CPUY192018, an inhibitor of Nrf2-Keap1 protein-protein interaction in protecting the injured brain cells is given as evidence of Nrf2's role in activating antioxidant genes. Overall, the scope of Nrf2 in developing therapeutic interventions for a variety of pathophysiological conditions associated with CNS injury-induced free radical/inflammatory signaling is acknowledged. Nrf2 has a widespread application in basic and clinical neuroscience for understanding and treating free radical/inflammatory signaling disorders, including neurological diseases. The development of innovative therapeutic strategies using Nrf2-inducing agents can be applied to reduce the complications of TBI before advancing it to posttraumatic stress disorder (PTSD).

4.
Exp Neurol ; 358: 114222, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36089059

RESUMEN

After a mild traumatic brain injury (mTBI), victims often experience emotional/psychological stress such as heightened irritability, anxiety, apathy, and depression. Severe mental health complications are common in military populations following a combat-acquired TBI and intensified unhealthy alcohol use. The high prevalence of alcohol abuse among TBI victims underscores how alcohol abuse exacerbates emotional/psychological symptoms such as depression and anxiety. The experimental mTBI was induced in vivo by fluid percussion injury (15 psi) in mice and ethanol diet feeding continued for 28 days. We analyzed different biomarkers of the biochemical mechanisms and pathophysiology of neurological damage, and functional outcome of psychological stress by sucrose preference, and light-dark tests. We demonstrated that the synergistic effect of TBI and alcohol leads to psychological stress such as depression and anxiety. The studies showed that oxidative stress, amyloidogenesis, tau pathology, neuroinflammation, and neurodegeneration markers were elevated, and glial activation and blood-brain barrier (BBB) damage were exacerbated during the synergistic effect of TBI and alcohol. Further, we studied the biochemical mechanisms of psychological stress that showed the significant reduction of 5-HT1AR, neuropeptide-Y, and norepinephrine, and an increase in monoamine oxidase-a in the combined effect of TBI and alcohol. This work suggested that the combined TBI and alcohol-induced effect leads to depression and anxiety, via sequential biochemical changes that cause neuroinflammation, amyloidogenesis, tau pathology, neurodegeneration, and BBB alterations. This clinically relevant study will contribute to developing a comprehensive therapeutic approach for patients suffering from TBI and alcohol-mediated neurological damage and psychological stress.


Asunto(s)
Alcoholismo , Conmoción Encefálica , Lesiones Traumáticas del Encéfalo , Neuropéptidos , Animales , Barrera Hematoencefálica/patología , Conmoción Encefálica/complicaciones , Conmoción Encefálica/patología , Lesiones Traumáticas del Encéfalo/patología , Etanol , Ratones , Monoaminooxidasa , Enfermedades Neuroinflamatorias , Norepinefrina , Estrés Psicológico/complicaciones , Sacarosa
5.
Neuropharmacology ; 213: 109136, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35584723

RESUMEN

Oxidative signaling and inflammatory cascades are the central mechanism in alcohol-induced brain injury, which result in glial activation, neuronal and myelin loss, neuronal apoptosis, and ultimately long-term neurological deficits. While transforming growth factor-beta1 (TGF-ß1) has a significant role in inflammation and apoptosis in myriads of other pathophysiological conditions, the precise function of increased TGF-ß1 in alcohol use disorder (AUD)-induced brain damage is unknown. In this study, our objective is to study ethanol-induced activation of TGF-ß1 and associated mechanisms of neuroinflammation and apoptosis. Using a mouse model feeding with ethanol diet and an in vitro model in mouse cortical neuronal cultures, we explored the significance of TGF-ß1 activation in the pathophysiology of AUD. Our study demonstrated that the activation of TGF-ß1 in ethanol ingestion correlated with the induction of free radical generating enzyme NADPH oxidase (NOX). Further, using TGF-ß type I receptor (TGF-ßRI) inhibitor SB431542 and TGF-ß antagonist Smad7, we established that the alcohol-induced activation of TGF-ß1 impairs antioxidant signaling pathways and leads to neuroinflammation and apoptosis. Blocking of TGF-ßRI or inhibition of TGF-ß1 diminished TGF-ß1-induced inflammation and apoptosis. Further, TGF-ß1 activation increased the phosphorylation of R-Smads including Smad2 and Smad3 proteins. Using various biochemical analyses and genetic approaches, we demonstrated the up-regulation of pro-inflammatory cytokines IL-1ß and TNF-α and apoptotic cell death in neurons. In conclusion, this study significantly extends our understanding of the pathophysiology of AUD and provides a unique insight for developing various therapeutic interventions by activating antioxidant signaling pathways for the treatment of AUD-induced neurological complications.


Asunto(s)
Alcoholismo , Antioxidantes , NADPH Oxidasas , Neuronas , Factor de Crecimiento Transformador beta1 , Animales , Antioxidantes/metabolismo , Células Cultivadas , Etanol/toxicidad , Inflamación/metabolismo , Ratones , NADPH Oxidasas/metabolismo , Neuronas/efectos de los fármacos , Transducción de Señal , Factor de Crecimiento Transformador beta1/metabolismo
6.
eNeuro ; 8(4)2021.
Artículo en Inglés | MEDLINE | ID: mdl-34135004

RESUMEN

Intercellular adhesion molecule-1 (ICAM-1) promotes adhesion and transmigration of circulating leukocytes across the blood-brain barrier (BBB). Traumatic brain injury (TBI) causes transmigrated immunocompetent cells to release mediators [function-associated antigen (LFA)-1 and macrophage-1 antigen (Mac-1)] that stimulate glial and endothelial cells to express ICAM-1 and release cytokines, sustaining neuroinflammation and neurodegeneration. Although a strong correlation exists between TBI-mediated inflammation and impairment in functional outcome following brain trauma, the role of ICAM-1 in impairing functional outcome by inducing neuroinflammation and neurodegeneration after TBI remains inconclusive. The experimental TBI was induced in vivo by fluid percussion injury (FPI; 10 and 20 psi) in wild-type (WT) and ICAM-1-/- mice and in vitro by stretch injury (3 psi) in brain endothelial cells. We manipulate ICAM-1 pharmacologically and genetically and conducted several biochemical analyses to gain insight into the mechanisms underlying ICAM-1-mediated neuroinflammation and performed rotarod, grid-walk, sucrose preference, and light-dark tests to assess functional outcome. TBI-induced ICAM-1-mediated neuroinflammation and cell death occur via LFA-1 or Mac-1 signaling pathways that rely on oxidative stress, matrix metalloproteinase (MMP), and vascular endothelial growth factor (VEGF) pathways. The deletion or blocking of ICAM-1 resulted in a better outcome in attenuating neuroinflammation and cell death as marked by the markers such as NF-kB, IL-1ß, TNF-α, cleaved-caspase-3 (cl-caspase-3), Annexin V, and by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and Trypan blue staining. ICAM-1 deletion in TBI improves sensorimotor, depression, and anxiety-like behavior with significant upregulation of norepinephrine (NE), dopamine (DA) D1 receptor (DAD1R), serotonin (5-HT)1AR, and neuropeptide Y (NPY). This study could establish the significance of ICAM-1 as a novel therapeutic target against the pathophysiology to establish functional recovery after TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Molécula 1 de Adhesión Intercelular , Estrés Psicológico , Animales , Lesiones Traumáticas del Encéfalo/complicaciones , Células Endoteliales/metabolismo , Hipocampo/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Ratones , Ratones Noqueados , Corteza Prefrontal/metabolismo , Factor A de Crecimiento Endotelial Vascular
7.
J Neuropathol Exp Neurol ; 80(2): 169-181, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33367790

RESUMEN

The long-term disabilities associated with spinal cord injury (SCI) are primarily due to the absence of robust neuronal regeneration and functional plasticity. The inability of the axon to regenerate after SCI is contributed by several intrinsic factors that trigger a cascade of molecular growth program and modulates axonal sprouting. Phosphatase and tensin homolog (PTEN) is one of the intrinsic factors contributing to growth failure after SCI, however, the underlying mechanism is not well known. Here, we developed a novel therapeutic approach for treating SCI by suppressing the action of PTEN in a mouse model of hemisection SCI. We have used a novel peptide, PTEN antagonistic peptide (PAP) to block the critical domains of PTEN to demonstrate its ability to potentially promote axon growth. PAP treatment not only enhanced regeneration of corticospinal axons into the caudal spinal cord but also promoted the regrowth of descending serotonergic axons in SCI mice. Furthermore, expression levels of p-mTOR, p-S6, p-Akt, p-Erk, p-GSK, p-PI3K downstream of PTEN signaling pathway were increased significantly in the spinal cord of SCI mice systemically treated with PAP than control TAT peptide-treated mice. Our novel strategy of administering deliverable compounds postinjury may facilitate translational feasibility for central nervous system injury.


Asunto(s)
Regeneración Nerviosa/efectos de los fármacos , Fosfohidrolasa PTEN/antagonistas & inhibidores , Péptidos/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Médula Espinal/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Modelos Animales de Enfermedad , Ratones , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Péptidos/farmacología , Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/metabolismo
8.
J Mol Med (Berl) ; 97(12): 1627-1641, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31758217

RESUMEN

Recent studies from our group and others have demonstrated that oxidative stress, Ca2+ signaling, and neuroinflammation are major mechanisms contributing to post-traumatic neurodegeneration. The present study investigated the mechanisms of regulation of nuclear factor E2-related factor 2 (Nrf2) and its role in regulating antioxidant genes and oxidative stress-induced neuroinflammation and neurodegeneration following TBI. Nrf2 transcriptional system is the major regulator of endogenous defense mechanisms operating within the cells. Wild-type (Nrf2+/+) and Nrf2-deficient mice (Nrf2-/-) were subjected to 15 psi fluid percussion injury and demonstrated the regulatory role of Nrf2 in the expression antioxidant genes and oxidative stress, neuroinflammation, and cell death. Immunohistochemistry, q-RT-PCR, and western blotting techniques detected downregulation of Nrf2 and antioxidant proteins such as HO-1, GPx1, GSTm1, and NQO1 in mouse brain samples. Further, our study demonstrated that the downregulation of Nrf2 and antioxidant genes in TBI correlated with the induction of free radical-generating enzyme NADPH oxidase 1 and inducible nitric oxide synthase and their corresponding oxidative/nitrosative stress markers 4-hydroxynonenal and 3-nitrotyrosine. The decrease in Nrf2 with subsequent increase in oxidative stress markers led to the activation of MMP3/9, TGF-ß1, and NF-kB that further led to neuroinflammation and apoptosis. The absence of Nrf2 function in mice resulted in exacerbated brain injury as shown by the increased oxidative stress markers, pro-inflammatory cytokines, and apoptosis markers at 24 h after TBI. In conclusion, this study could establish the significance of Nrf2 in transforming into a novel preventive approach against the pathophysiology of TBI. KEY MESSAGES: • Traumatic brain injury impairs Nrf2 signaling in mouse. • Nrf2-mediated activation of antioxidant genes are altered after TBI. • Impairment of Nrf2 signaling leads to oxidative stress. • TBI-induced downregulation of Nrf2 activates MMPs, TGF-ß1, and NF-kB. • Nrf2 regulates neuroinflammation and apoptotic cell death in TB.


Asunto(s)
Antioxidantes/metabolismo , Apoptosis , Lesiones Traumáticas del Encéfalo/metabolismo , Encéfalo/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Animales , Apoptosis/genética , Encéfalo/citología , Encéfalo/fisiopatología , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/fisiopatología , Regulación hacia Abajo/genética , Inflamación/genética , Inflamación/metabolismo , Masculino , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NADPH Oxidasa 1/metabolismo , Factor 2 Relacionado con NF-E2/química , Factor 2 Relacionado con NF-E2/genética , FN-kappa B/metabolismo , Óxido Nítrico Sintasa/metabolismo , Estrés Oxidativo/genética , Fosforilación , Transducción de Señal/genética , Factor de Crecimiento Transformador beta1/metabolismo
9.
Exp Neurol ; 317: 260-270, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30926390

RESUMEN

The blood-brain barrier (BBB) constitutes a neurovascular unit formed by microvascular endothelial cells, pericytes, and astrocytes. Brain pericytes are important regulators of BBB integrity, permeability, and blood flow. Pericyte loss has been implicated in injury; however, how the crosstalk among pericytes, endothelial cells, and astrocytes ultimately leads to BBB dysfunction in traumatic brain injury (TBI) remains elusive. In this study, we demonstrate the importance of pericyte-endothelium interaction in maintaining the BBB function. TBI causes the platelet-derived growth factor-B (PDGF-B)/PDGF receptor-ß signaling impairment that results in loss of interaction with endothelium and leads to neurovascular dysfunction. Using in vivo mild (7 psi) and moderate (15 psi) fluid percussion injury (FPI) in mice, we demonstrate the expression of various pericyte markers including PDGFR-ß, NG2 and CD13 that were significantly reduced with a subsequent reduction in the expression of various integrins; adherent junction protein, N-cadherin; gap junction protein, connexin-43; and tight junction proteins such as occludin, claudin-5, ZO-1, and JAM-a. Impairment of pericyte-endothelium interaction increases the BBB permeability to water that is marked by a significant increase in aquaporin4 expression in injured animals. Similarly, pericyte-endothelium integrity impairment in FPI animals greatly increases the permeability of small-molecular-weight sodium fluorescein and high-molecular-weight-tracer Evans blue across the BBB. In addition, the injury-inflicted animals show significantly higher levels of S100ß and NSE in the blood samples compared with controls. In conclusion, our data provide an insight that brain trauma causes an early impairment of pericyte-endothelium integrity and results in BBB dysregulation that initiates pathological consequences associated with TBI.


Asunto(s)
Barrera Hematoencefálica/patología , Lesiones Traumáticas del Encéfalo/patología , Endotelio Vascular/patología , Pericitos/patología , Animales , Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Permeabilidad Capilar/fisiología , Endotelio Vascular/metabolismo , Ratones , Pericitos/metabolismo , Receptor Cross-Talk/fisiología
10.
Mol Neurobiol ; 56(2): 1124-1136, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29873042

RESUMEN

Mitogen-activated protein (MAP) kinases are serine/threonine protein kinases that play a critical role in signal transduction and are activated by phosphorylation in response to a variety of pathophysiology stimuli. While MAP kinase signaling has a significant role in the pathophysiology of several neurodegenerative diseases, the precise function of activation of MAP kinase in traumatic brain injury (TBI) is unknown. Therefore, it is important to study the role of MAP kinase signaling in TBI-associated neurological ailments. In this study, using an in vitro stretch injury model in rat embryo neuronal cultures and the in vivo fluid percussion injury (FPI) model in rats, we explored the role of MAP kinase signaling in the mechanisms of cell death in TBI. Our study demonstrated that the stretch injury in vitro and FPI in vivo upregulated the phosphorylation of MAP kinase proteins ERK1/2 and JNK, but not p38. Using ERK1/2 inhibitor U0126, JNK inhibitor SP600125, and p38 inhibitor SB203580, we validated the role of MAP kinase proteins in the activation of NF-kB and caspase-3. By immunofluorescence and western blotting, further, we demonstrated the role of ERK1/2 and JNK phosphorylation in neurodegeneration by analyzing cell death proteins annexin V and Poly-ADP-Ribose-Polymerase p85. Interestingly, combined use of ERK1/2 and JNK inhibitors further attenuated the cell death in stretch-injured neurons. In conclusion, this study could establish the significance of MAP kinase signaling in the pathophysiology of TBI and may have significant implications for developing therapeutic strategies using ERK1/2 and JNK inhibitors for TBI-associated neurological complications.


Asunto(s)
Lesiones Traumáticas del Encéfalo/metabolismo , Encéfalo/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Neuronas/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis/fisiología , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/patología , Muerte Celular/fisiología , Neuronas/patología , Fosforilación , Ratas , Ratas Sprague-Dawley
11.
Brain Sci ; 8(1)2018 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-29316623

RESUMEN

Traumatic brain injury (TBI) can result in persistent sensorimotor and cognitive deficits, which occur through a cascade of deleterious pathophysiological events over time. In this study, we investigated the hypothesis that neurodegeneration caused by TBI leads to impairments in sensorimotor function. TBI induces the activation of the caspase-3 enzyme, which triggers cell apoptosis in an in vivo model of fluid percussion injury (FPI). We analyzed caspase-3 mediated apoptosis by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining and poly (ADP-ribose) polymerase (PARP) and annexin V western blotting. We correlated the neurodegeneration with sensorimotor deficits by conducting the animal behavioral tests including grid walk, balance beam, the inverted screen test, and the climb test. Our study demonstrated that the excess cell death or neurodegeneration correlated with the neuronal dysfunction and sensorimotor impairments associated with TBI.

12.
Mol Neurobiol ; 55(7): 5937-5950, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29128903

RESUMEN

Wernicke's encephalopathy, a common neurological disease, is caused by thiamine (vitamin B1) deficiency. Neuropathy resulting from thiamine deficiency is a hallmark of Wernicke-Korsakoff syndrome in chronic alcohol users. The underlying mechanisms of this deficiency and progression of neuropathy remain to be understood. To uncover the unknown mechanisms of thiamine deficiency in alcohol abuse, we used chronic alcohol consumption or thiamine deficiency diet ingestion in animal models. Observations from animal models were validated in primary human neuronal culture for neurodegenerative process. We employed radio-labeled bio-distribution of thiamine, qualitative and quantitative analyses of the various biomarkers and neurodegenerative process. In the present studies, we established that disruption of thiamine transport across the intestinal gut blood-brain barrier axis as the cause of thiamine deficiency in the brain for neurodegeneration. We found that reduction in thiamine transport across these interfaces was the cause of reduction in the synthesis of thiamine pyrophosphate (TPP), an active cofactor for pyruvate dehydrogenase E1α (PDHE1α). Our findings revealed that decrease in the levels of PDHE1α cofactors switched on the activation of PD kinase (PDK) in the brain, thereby triggering the neuronal phosphorylation of PDHE1α (p-PDHE1α). Dysfunctional phosphorylated PDHE1α causes the reduction of mitochondrial aerobic respiration that led to neurodegeneration. We concluded that impairment of thiamine transport across the gut-BBB-axis that led to insufficient TPP synthesis was critical to Wernicke-neuropathy, which could be effectively prevented by stabilizing the thiamine transporters.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Tracto Gastrointestinal/metabolismo , Tiamina/metabolismo , Encefalopatía de Wernicke/metabolismo , Encefalopatía de Wernicke/patología , Animales , Transporte Biológico , Supervivencia Celular , Dieta , Regulación hacia Abajo , Etanol , Humanos , Masculino , Proteínas de Transporte de Membrana/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Neuronas/metabolismo , Neuronas/patología , Fosforilación , Piruvato Deshidrogenasa (Lipoamida)/metabolismo , Distribución Tisular
13.
Mol Neurobiol ; 55(7): 5901-5912, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29119534

RESUMEN

Angiotensin II (Ang II) is a mediator of oxidative stress via activation/induction of reactive oxygen and nitrogen species-generating enzymes, NADPH oxidase (NOX) and inducible nitric oxide synthase (iNOS). We investigated the hypothesis that overproduction of Ang II during traumatic brain injury (TBI) induces the activation of the oxidative stress, which triggers neuroinflammation and cell apoptosis in a cell culture model of neuronal stretch injury. We first established that stretch injury causes a rapid increase in the level of Ang II, which causes the release of pro-inflammatory cytokines, IL-1ß and TNF-α, via the induction of oxidative stress. Since angiotensin-converting enzyme (ACE) mediates the production of Ang II via the conversion of Ang I into Ang II, we analyzed the expression of ACE by western blotting. Further, we analyzed caspase-3-mediated apoptosis by TUNEL staining and annexin V western blotting. Angiotensin type I (AT1) receptor antagonist losartan attenuated Ang II-induced oxidative stress and associated neuroinflammation and cell death in cultured neurons. Remarkably, we noticed that the expression of Ang II type 1 receptor (AngT1R) upregulated in neuronal stretch injury; losartan mitigates this upregulation. Findings from this study significantly extend our understanding of the pathophysiology of TBI and may have significant implications for developing therapeutic strategies for TBI-associated brain dysfunctions.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Losartán/farmacología , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Receptor de Angiotensina Tipo 1/metabolismo , Estrés Mecánico , Angiotensina II , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Citocinas/metabolismo , Activación Enzimática/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Peptidil-Dipeptidasa A/metabolismo , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
Brain Behav Immun ; 64: 244-258, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28433746

RESUMEN

Mild traumatic brain injury (mTBI) is a low-level injury, which often remains undiagnosed, and in most cases it leads to death and disability as it advances as secondary injury. Therefore, it is important to study the underlying signaling mechanisms of mTBI-associated neurological ailments. While transforming growth factor-beta1 (TGF-ß1) has a significant role in inflammation and apoptosis in myriads of other pathophysiological conditions, the precise function of increased TGF-ß1 after mTBI is unknown. In this study, our objective is to study the physiological relevance and associated mechanisms of TGF-ß1-mediated inflammation and apoptosis in mTBI. Using an in vitro stretch-injury model in rat neuronal cultures and the in vivo fluid percussion injury (FPI) model in rats, we explored the significance of TGF-ß1 activation in mTBI. Our study demonstrated that the activation of TGF-ß1 in mTBI correlated with the induction of free radical generating enzyme NADPH oxidase 1 (NOX1). Further, using TGF-ß type I receptor (TGF-ßRI) inhibitor SB431542 and transfection of TGF-ß1 siRNA and TGF-ß antagonist Smad7, we established the neuroinflammatory and apoptotic role of TGF-ß1 in mTBI. Inhibition of TGF-ßRI or TGF-ß1 diminished TGF-ß1-induced inflammation and apoptosis. Further, the enhanced TGF-ß1 activation increased the phosphorylation of R-Smads including Smad2 and Smad3 proteins. By immunofluorescence, western blotting, ELISA and TUNEL experiments, we demonstrated the up-regulation of pro-inflammatory cytokines IL-1ß and TNF-α and apoptotic cell death in neurons. In conclusion, this study could establish the significance of TGF-ß1 in transforming the pathophysiology of mTBI into secondary injury.


Asunto(s)
Apoptosis , Conmoción Encefálica/metabolismo , Encefalitis/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Conmoción Encefálica/complicaciones , Células Cultivadas , Encefalitis/complicaciones , Mediadores de Inflamación/metabolismo , Masculino , NADPH Oxidasa 1/metabolismo , Estrés Oxidativo , Ratas Sprague-Dawley , Transducción de Señal
15.
Brain Behav Immun ; 62: 291-305, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28232172

RESUMEN

Epidemiological studies showed a strong association between alcoholism and incidence of stroke, for which the underlying causative mechanisms remain to be understood. Here we found that infiltration of immune cells and deposition of cholesterol at the site of brain artery/capillary injury induced atherosclerosis in chronic alcohol (ethanol) consumption in the presence or absence of high-fat diet. Conversion of cholesterol into sharp edges of cholesterol crystals (CCs) in alcohol intake was key to activation of NLRP3 inflammasome, induction of cerebral atherosclerosis, and development of neuropathy around the atherosclerotic lesions. The presence of alcohol was critical for the formation of CCs and development of the neuropathology. Thus, we observed that alcohol consumption elevated the level of plasma cholesterol, deposition and crystallization of cholesterol, as well as activation of NLRP3 inflammasome. This led to arteriole or capillary walls thickening and increase intracranial blood pressure. Distinct neuropathy around the atherosclerotic lesions indicated vascular inflammation as an initial cause of neuronal degeneration. We demonstrated the molecular mechanisms of NLRP3 activation and downstream signaling cascade event in primary culture of human brain arterial/capillary endothelial cells in the setting of dose-/time-dependent effects of alcohol/CCs using NLRP3 gene silencing technique. We also detected CCs in blood samples from alcohol users, which validated the clinical importance of the findings. Finally, combined therapy of acetyl-l-carnitine and Lipitor® prevented deposition of cholesterol, formation of CCs, activation of NLRP3, thickening of vessel walls, and elevation of intracranial blood pressure. We conclude that alcohol-induced accumulation and crystallization of cholesterol activates NLRP3/caspase-1 in the cerebral vessel that leads to early development of atherosclerosis.


Asunto(s)
Consumo de Bebidas Alcohólicas/metabolismo , Aterosclerosis/metabolismo , Encéfalo/metabolismo , Colesterol/metabolismo , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Presión Sanguínea/fisiología , Dieta Alta en Grasa , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
16.
Mol Neurobiol ; 54(6): 3964-3975, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27289225

RESUMEN

We investigated the hypothesis that high Ca2+ influx during traumatic brain injury induces the activation of the caspase-1 enzyme, which triggers neuroinflammation and cell apoptosis in a cell culture model of neuronal stretch injury and an in vivo model of fluid percussion injury (FPI). We first established that stretch injury causes a rapid increase in the intracellular Ca2+ level, which activates interleukin-converting enzyme caspase-1. The increase in the intracellular Ca2+ level and subsequent caspase-1 activation culminates into neuroinflammation via the maturation of IL-1ß. Further, we analyzed caspase-1-mediated apoptosis by TUNEL staining and PARP western blotting. The voltage-gated sodium channel blocker, tetrodotoxin, mitigated the stretch injury-induced neuroinflammation and subsequent apoptosis by blocking Ca2+ influx during the injury. The effect of tetrodotoxin was similar to the caspase-1 inhibitor, zYVAD-fmk, in neuronal culture. To validate the in vitro results, we demonstrated an increase in caspase-1 activity, neuroinflammation and neurodegeneration in fluid percussion-injured animals. Our data suggest that neuronal injury/traumatic brain injury (TBI) can induce a high influx of Ca2+ to the cells that cause neuroinflammation and cell death by activating caspase-1, IL-1ß, and intrinsic apoptotic pathways. We conclude that excess IL-1ß production and cell death may contribute to neuronal dysfunction and cognitive impairment associated with TBI.


Asunto(s)
Apoptosis , Lesiones Traumáticas del Encéfalo/enzimología , Lesiones Traumáticas del Encéfalo/patología , Calcio/metabolismo , Caspasa 1/metabolismo , Inflamación/enzimología , Inflamación/patología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Interleucina-1beta/metabolismo , Modelos Biológicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas Sprague-Dawley , Tetrodotoxina/toxicidad
17.
Brain Behav Immun ; 59: 190-199, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27614125

RESUMEN

Traumatic brain injury (TBI), even at mild levels, can activate matrix metalloproteinases (MMPs) and the induction of neuroinflammation that can result in blood brain barrier breakdown and neurodegeneration. MMP2 has a significant role in neuroinflammation and neurodegeneration by modulating the chemokine CXCL12α (stromal cell derived factor SDF-1α) signaling pathway and the induction of apoptosis. SDF-1α is responsible for cell proliferation and differentiation throughout the nervous system and is also implicated in various neurodegenerative illnesses. We hypothesized that TBI leads to MMP2 activation and cleavage of the N-terminal 4 amino acid residues of CXCL12α with generation of the highly neurotoxic fragment SDF-1(5-67). Using an in vitro stretch-injury model of rat neuronal cultures and the in vivo fluid percussion injury (FPI) model in rats, we found that oxidative stress has a significant role in the activation of MMP2. This is initiated by the induction of free radical generating enzyme NADPH oxidase 1 (NOX1). Induction of NOX1 correlated well with the signatures of oxidative stress marker, 4HNE in the injured neuronal cultures and cerebral cortex of rats. Further, using MMP2 siRNA and pharmacological MMP2 inhibitor, ARP100, we established the neurodegenerative role of MMP2 in cleaving SDF-1α to a neurotoxic fragment SDF-1(5-67). By immunofluorescence, western blotting and TUNEL experiments, we show the cleaved form of SDF leads to apoptotic cell death in neurons. This work identifies a new potential therapeutic target to reduce the complications of brain damage in TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/enzimología , Quimiocina CXCL12/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Degeneración Nerviosa/enzimología , Degeneración Nerviosa/genética , Animales , Apoptosis/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/genética , Caspasa 3/biosíntesis , Caspasa 3/genética , Supervivencia Celular/genética , Células Cultivadas , Quimiocina CXCL12/genética , Activación Enzimática , Técnicas de Silenciamiento del Gen , Metaloproteinasa 2 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , NADPH Oxidasa 1/biosíntesis , NADPH Oxidasa 1/genética , Neuronas/efectos de los fármacos , Estrés Oxidativo , ARN Interferente Pequeño/farmacología , Ratas , Ratas Sprague-Dawley
18.
Sci Rep ; 6: 37152, 2016 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-27849007

RESUMEN

Receptor protein tyrosine phosphatase σ (PTPσ) and its subfamily member LAR act as transmembrane receptors that mediate growth inhibition of chondroitin sulfate proteoglycans (CSPGs). Inhibition of either receptor increases axon growth into and beyond scar tissues after CNS injury. However, it is unclear why neurons express two similar CSPG receptors, nor whether they use the same or different intracellular pathways. We have now studied the signaling pathways of these two receptors using N2A cells and primary neurons derived from knockout mice. We demonstrate that both receptors share certain signaling pathways (RhoA, Akt and Erk), but also use distinct signals to mediate CSPG actions. Activation of PTPσ by CSPGs selectively inactivated CRMP2, APC, S6 kinase and CREB. By contrast LAR activation inactivated PKCζ, cofilin and LKB1. For the first time, we propose a model of the signaling pathways downstream of these two CSPG receptors. We also demonstrate that deleting both receptors exhibits additive enhancement of axon growth in adult neuronal cultures in vitro. Our findings elucidate the novel downstream pathways of CSPGs and suggest potential synergy of blocking their two PTP receptors.


Asunto(s)
Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Neuronas/metabolismo , Proteínas Quinasas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Línea Celular , Proteoglicanos Tipo Condroitín Sulfato/genética , Ratones , Neuronas/citología , Proteínas Quinasas/genética , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/genética
20.
Mol Neurobiol ; 53(9): 6106-6123, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26541883

RESUMEN

Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Studies revealed that the pathogenesis of TBI involves upregulation of MMPs. MMPs form a large family of closely related zinc-dependent endopeptidases, which are primarily responsible for the dynamic remodulation of the extracellular matrix (ECM). Thus, they are involved in several normal physiological processes like growth, development, and wound healing. During pathophysiological conditions, MMPs proteolytically degrade various components of ECM and tight junction (TJ) proteins of BBB and cause BBB disruption. Impairment of BBB causes leakiness of the blood from circulation to brain parenchyma that leads to microhemorrhage and edema. Further, MMPs dysregulate various normal physiological processes like angiogenesis and neurogenesis, and also they participate in the inflammatory and apoptotic cascades by inducing or regulating the specific mediators and their receptors. In this review, we explore the roles of MMPs in various physiological/pathophysiological processes associated with neurological complications, with special emphasis on TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo/enzimología , Lesiones Traumáticas del Encéfalo/etiología , Metaloproteinasas de la Matriz/metabolismo , Animales , Barrera Hematoencefálica/patología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Humanos , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Metaloproteinasas de la Matriz/clasificación , Terapia Molecular Dirigida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...