Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
Sci Prog ; 107(3): 368504241259299, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39196597

RESUMEN

BACKGROUND: Inflammatory processes are key factors in pathological events associated with severe traumatic brain injury (STBI). The aim of this trial was to determine the effect of probiotics on anthropometric measures, disease severity, inflammatory markers, and T cells in patients with STBI. METHODS: Forty adult patients with STBI were enrolled in this parallel randomized, double-blind, placebo-controlled trial. Energy and protein status, Acute Physiology and Chronic Health Evaluation (APACHE II) score, Sequential Organ Failure Assessment (SOFA), interleukin 10 (IL-10), interleukin 1ß (IL-1ß), tumor necrosis factor-alpha (TNF-α), transforming growth factor beta (TGF-ß), T-helper 17 (Th17), and T- Regulator (T-reg) cells were assessed at baseline (day 1), and week 2 (day 14) for each patient. RESULTS: Probiotic supplementation led to a substantial reduction in the serum levels of TNF-α (from 10.15 ± 6.52 to 5.05 ± 3.27) (P = 0.034), IL-1ß (from 11.84 ± 7.74 to 5.87 ± 3.77) (P < 0.001), and Th17 cells (from 5.19 ± 1.69 to 2.67 ± 1.89) (P < 0.001) and a substantial increase in the serum levels of IL-10 (from 3.35 ± 1.45 to 6.17 ± 2.04) (P = 0.038), TGF-ß (from 30.5 ± 15.27 to 46.25 ± 21.05) (P < 0.001), and T-reg cells (from 2.83 ± 1.43 to 4.29 ± 1.89) (P < 0.001) compared with the placebo group. Furthermore, no notable changes were observed in energy and protein intake and also, terms of SOFA and APACHE II scores following probiotic treatment compared with the placebo. CONCLUSIONS: Probiotics could reduce inflammation and improve cellular immunity and may be considered as an adjunctive therapy in STBI patients.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Probióticos , Humanos , Probióticos/uso terapéutico , Probióticos/administración & dosificación , Lesiones Traumáticas del Encéfalo/terapia , Lesiones Traumáticas del Encéfalo/dietoterapia , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/sangre , Método Doble Ciego , Masculino , Femenino , Adulto , Biomarcadores/sangre , Persona de Mediana Edad , Inflamación , Linfocitos T/inmunología , Citocinas/sangre , Citocinas/metabolismo
2.
J Neuroinflammation ; 21(1): 172, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39014496

RESUMEN

Post-traumatic epilepsy (PTE) is one of the most debilitating consequences of traumatic brain injury (TBI) and is one of the most drug-resistant forms of epilepsy. Novel therapeutic treatment options are an urgent unmet clinical need. The current focus in healthcare has been shifting to disease prevention, rather than treatment, though, not much progress has been made due to a limited understanding of the disease pathogenesis. Neuroinflammation has been implicated in the pathophysiology of traumatic brain injury and may impact neurological sequelae following TBI including functional behavior and post-traumatic epilepsy development. Inflammasome signaling is one of the major components of the neuroinflammatory response, which is increasingly being explored for its contribution to the epileptogenic mechanisms and a novel therapeutic target against epilepsy. This review discusses the role of inflammasomes as a possible connecting link between TBI and PTE with a particular focus on clinical and preclinical evidence of therapeutic inflammasome targeting and its downstream effector molecules for their contribution to epileptogenesis. Finally, we also discuss emerging evidence indicating the potential of evaluating inflammasome proteins in biofluids and the brain by non-invasive neuroimaging, as potential biomarkers for predicting PTE development.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Epilepsia Postraumática , Inflamasomas , Humanos , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/inmunología , Inflamasomas/metabolismo , Animales , Epilepsia Postraumática/metabolismo , Epilepsia Postraumática/etiología
4.
Int Immunopharmacol ; 139: 112774, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39067398

RESUMEN

Repetitive traumatic brain injury (RTBI) is acknowledged as a silent overlooked public health crisis, with an incomplete understanding of its pathomechanistic signaling pathways. Mounting evidence suggests the involvement of thrombin and its receptor, the protease-activated receptor (PAR)1, in the development of secondary injury in TBI; however, the consequences of PAR1 modulation and its impact on ferroptosis-redox signaling, and NLRP3 inflammasome activation in RTBI, remain unclear. Further, the utilitarian function of PAR1 as a therapeutic target in RTBI has not been elucidated. To study this crosstalk, RTBI was induced in Wistar rats by daily weight drops on the right frontal region for five days. Three groups were included: normal control, untreated RTBI, and RTBI+SCH79797 (a PAR1 inhibitor administered post-trauma at 25 µg/kg/day). The concomitant treatment of PAR1 antagonism improved altered behavior function, cortical histoarchitecture, and neuronal cell survival. Moreover, the receptor blockade downregulated mRNA expression of PAR1 but upregulatedthat of the neuroprotective receptor PPAR-γ. The anti-inflammatory impact of SCH79797 was signified by the low immune expression/levels of NF-κB p65,TNF-α, IL-1ß, and IL-18. Consequently, the PAR1 blocker hindered the formation of inflammasome components NLRP3, ASC, and activated caspase-1. Ultimately, SCH79797 treatment abated ferroptosis-dependent iron redox signaling through the activation of the antioxidant Nrf2/HO-1 axis and its subsequent antioxidant machinery (GPX4, SOD) to limit lipid peroxidation, iron accumulation, and transferrin serum increment. Collectively, SCH79797 offered putative preventive mechanisms against secondary RTBI consequences in rats by impeding ferroptosis and NLRP3 inflammasome through activating the PPAR-γ/Nrf2 antioxidant cue.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Ferroptosis , Inflamasomas , Factor 2 Relacionado con NF-E2 , Proteína con Dominio Pirina 3 de la Familia NLR , PPAR gamma , Ratas Wistar , Receptor PAR-1 , Transducción de Señal , Animales , Masculino , Ratas , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/patología , Modelos Animales de Enfermedad , Ferroptosis/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Inflamasomas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , PPAR gamma/metabolismo , Receptor PAR-1/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
5.
Medicine (Baltimore) ; 103(29): e39007, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-39029062

RESUMEN

Traumatic brain injury (TBI) is a significant health problem with a high mortality rate. Inflammatory markers can predict the prognosis of TBI where neuroinflammation is essential. In this study, the prognostic value of the systemic immune-inflammation index (SII), neutrophil-lymphocyte ratio (NLR), and platelet-lymphocyte ratio (PLR) at admission in patients with critical TBI was investigated. Patients with moderately severe TBI in the intensive care unit (ICU) of a tertiary center between June 2020 and June 2022 were retrospectively reviewed. Patients were classified into survivor and mortality groups. The predictive performance of SII, PLR, and NLR levels calculated from blood results at admission and 28-day mortality and patient outcomes were analyzed. One hundred sixty-one patients were included in this study. The median age of the entire population was 41 (18-90) years, and 80.7% (n = 130) of the patients were male. Falls (42.2%) and traffic accidents (40.4%) were the most common causes of TBI. The most common primary diagnoses in patients with TBI were acute subdural hematoma (30.4%) and subarachnoid hemorrhage (26.1%). The SII and NLR levels were significantly higher in the mortality group, and PLR levels were significantly lower (P = .004, P < .001, P < .001, respectively). In multivariate regression analysis, SII and PLR were independent predictors of mortality (P = .031 and P < .001, respectively). In the receiver operating characteristics (ROC) curve analysis, the cutoff value for SII was ≥ 2951, and the area under the curve (AUC) was 0.662 (95% CI, 0.540-0.784). The cutoff value for NLR was ≥ 9.85, AUC was 0.717 (95% CI, 0.600-0.834), and the cutoff value for PLR was ≤ 130.4, AUC was 0.871 (95% CI, 0.796-0.947). 28-day mortality was 21.1%. Neuroinflammation is essential in patients with critical TBI, and inflammatory markers SII, NLR, and PLR have prognostic importance. SII and PLR are independent predictors of mortality. Early detection of those with a poor prognosis in critically ill TBI patients and planning aggressive treatments may contribute to reducing mortality.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Enfermedad Crítica , Linfocitos , Neutrófilos , Humanos , Masculino , Lesiones Traumáticas del Encéfalo/sangre , Lesiones Traumáticas del Encéfalo/mortalidad , Lesiones Traumáticas del Encéfalo/inmunología , Femenino , Persona de Mediana Edad , Adulto , Pronóstico , Estudios Retrospectivos , Anciano , Adolescente , Anciano de 80 o más Años , Plaquetas , Adulto Joven , Recuento de Plaquetas , Inflamación/sangre , Curva ROC , Unidades de Cuidados Intensivos/estadística & datos numéricos , Recuento de Linfocitos , Valor Predictivo de las Pruebas
6.
CNS Neurosci Ther ; 30(6): e14781, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38887195

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) and spinal cord injury (SCI) are acquired injuries to the central nervous system (CNS) caused by external forces that cause temporary or permanent sensory and motor impairments and the potential for long-term disability or even death. These conditions currently lack effective treatments and impose substantial physical, social, and economic burdens on millions of people and families worldwide. TBI and SCI involve intricate pathological mechanisms, and the inflammatory response contributes significantly to secondary injury in TBI and SCI. It plays a crucial role in prolonging the post-CNS trauma period and becomes a focal point for a potential therapeutic intervention. Previous research on the inflammatory response has traditionally concentrated on glial cells, such as astrocytes and microglia. However, increasing evidence highlights the crucial involvement of lymphocytes in the inflammatory response to CNS injury, particularly CD8+ T cells and NK cells, along with their downstream XCL1-XCR1 axis. OBJECTIVE: This review aims to provide an overview of the role of the XCL1-XCR1 axis and the T-cell response in inflammation caused by TBI and SCI and identify potential targets for therapy. METHODS: We conducted a comprehensive search of PubMed and Web of Science using relevant keywords related to the XCL1-XCR1 axis, T-cell response, TBI, and SCI. RESULTS: This study examines the upstream and downstream pathways involved in inflammation caused by TBI and SCI, including interleukin-15 (IL-15), interleukin-12 (IL-12), CD8+ T cells, CD4+ T cells, NK cells, XCL1, XCR1+ dendritic cells, interferon-gamma (IFN-γ), helper T0 cells (Th0 cells), helper T1 cells (Th1 cells), and helper T17 cells (Th17 cells). We describe their proinflammatory effect in TBI and SCI. CONCLUSIONS: The findings suggest that the XCL1-XCR1 axis and the T-cell response have great potential for preclinical investigations and treatments for TBI and SCI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Quimiocinas C , Traumatismos de la Médula Espinal , Humanos , Traumatismos de la Médula Espinal/inmunología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/patología , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/patología , Animales , Quimiocinas C/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Enfermedades Neuroinflamatorias/inmunología
7.
J Neuroinflammation ; 21(1): 135, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802931

RESUMEN

Traumatic brain injury (TBI) is a major cause of disability and mortality worldwide, particularly among the elderly, yet our mechanistic understanding of what renders the post-traumatic brain vulnerable to poor outcomes, and susceptible to neurological disease, is incomplete. It is well established that dysregulated and sustained immune responses elicit negative consequences after TBI; however, our understanding of the neuroimmune interface that facilitates crosstalk between central and peripheral immune reservoirs is in its infancy. The meninges serve as the interface between the brain and the immune system, facilitating important bi-directional roles in both healthy and disease settings. It has been previously shown that disruption of this system exacerbates neuroinflammation in age-related neurodegenerative disorders such as Alzheimer's disease; however, we have an incomplete understanding of how the meningeal compartment influences immune responses after TBI. In this manuscript, we will offer a detailed overview of the holistic nature of neuroinflammatory responses in TBI, including hallmark features observed across clinical and animal models. We will highlight the structure and function of the meningeal lymphatic system, including its role in immuno-surveillance and immune responses within the meninges and the brain. We will provide a comprehensive update on our current knowledge of meningeal-derived responses across the spectrum of TBI, and identify new avenues for neuroimmune modulation within the neurotrauma field.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Meninges , Enfermedades Neuroinflamatorias , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/patología , Humanos , Animales , Meninges/inmunología , Meninges/patología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/patología , Neuroinmunomodulación/fisiología , Neuroinmunomodulación/inmunología
8.
Neuroreport ; 35(9): 549-557, 2024 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-38739900

RESUMEN

Neuroinflammation after traumatic brain injury (TBI) exhibits a strong correlation with neurological impairment, which is a crucial target for improving the prognosis of TBI patients. The involvement of CXCL5/CXCR2 signaling in the regulation of neuroinflammation in brain injury models has been documented. Therefore, the effects of CXCL5 on post-TBI neuroinflammation and its potential mechanisms need to be explored. Following TBI, C57BL/6 mice were administered intraperitoneal injections of a CXCL5 neutralizing antibody (Nab-CXCL5) (5 mg/kg, 2 times/day). Subsequently, the effects on neuroinflammation, nerve injury, and neurological function were assessed. Nab-CXCL5 significantly reduced the release of inflammatory factors, inhibited the formation of inflammatory microglia and astrocytes, and reduced the infiltration of peripheral immune cells in TBI mice. Additionally, this intervention led to a reduction in neuronal impairment and facilitated the restoration of sensorimotor abilities, as well as improvements in learning and memory functions. Peripheral administration of the Nab-CXCL5 to TBI mice could suppress neuroinflammation, reduce neurological damage, and improve neurological function. Our data suggest that neutralizing antibodies against CXCL5 (Nab-CXCL5) may be a promising agent for treating TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Quimiocina CXCL5 , Enfermedades Neuroinflamatorias , Animales , Masculino , Ratones , Anticuerpos Neutralizantes/farmacología , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Quimiocina CXCL5/metabolismo , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Recuperación de la Función/efectos de los fármacos
9.
J Neurotrauma ; 41(13-14): e1708-e1718, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38623766

RESUMEN

Traumatic brain injury (TBI) is a common cause of morbidity and mortality in children. We have previously shown that TBI with a concurrent extracranial injury reliably leads to post-injury suppression of the innate and adaptive immune systems. In patients with post-injury immune suppression, if immune function could be preserved, this might represent a therapeutic opportunity. As such, we examined, in an animal injury model, whether systemic administration of granulocyte macrophage colony-stimulating factor (GM-CSF) could reverse post-injury immune suppression and whether treatment was associated with neuroinflammation or functional deficit. Prepubescent male rats were injured using a controlled cortical impact model and then subjected to removal of 25% blood volume (TBI/H). Sham animals underwent surgery without injury induction, and the treatment groups were sham and injured animals treated with either saline vehicle or 50 µg/kg GM-CSF. GM-CSF was administered following injury and then daily until sacrifice at post-injury day (PID) 7. Immune function was measured by assessing tumor necrosis factor-α (TNF-α) levels in whole blood and spleen following ex vivo stimulation with pokeweed mitogen (PWM). Brain samples were assessed by multiplex enzyme-linked immunosorbent assay (ELISA) for cytokine levels and by immunohistochemistry for microglia and astrocyte proliferation. Neuronal cell count was examined using cresyl violet staining. Motor coordination was evaluated using the Rotarod performance test. Treatment with GM-CSF was associated with a significantly increased response to PWM in both whole blood and spleen. GM-CSF in injured animals did not lead to increases in levels of pro-inflammatory cytokines in brain samples but was associated with significant increases in counted astrocytes. Finally, while injured animals treated with saline showed a significant impairment on behavioral testing, injured animals treated with GM-CSF performed similarly to uninjured animals. GM-CSF treatment in animals with combined injury led to increased systemic immune cell response in whole blood and spleen in the acute phase following injury. Improved immune response was not associated with elevated pro-inflammatory cytokine levels in the brain or functional impairment.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Traumatismo Múltiple , Ratas Sprague-Dawley , Animales , Masculino , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/inmunología , Ratas , Traumatismo Múltiple/inmunología , Traumatismo Múltiple/complicaciones , Terapia de Inmunosupresión/métodos
10.
Cytotherapy ; 26(7): 714-718, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38506768

RESUMEN

BACKGROUND AIMS: In this paper, we present a review of several selected talks presented at the CTTACC conference (Cellular Therapies in Trauma and Critical Care) held in Scottsdale, AZ in May 2023. This conference review highlights the potential for cellular therapies to "reset" the dysregulated immune response and restore physiologic functions to normal. Improvements in medical care systems and technology have increasingly saved lives after major traumatic events. However, many of these patients have complicated post-traumatic sequelae, ranging from short-term multi-organ failure to chronic critical illness. METHODS/RESULTS: Patients with chronic critical illness have been found to have dysregulated immune responses. These abnormal and harmful immune responses persist for years after the initial insult and can potentially be mitigated by treatment with cellular therapies. CONCLUSIONS: The sessions emphasized the need for more research and clinical trials with cellular therapies for the treatment of a multitude of chronic illnesses: post-trauma, radiation injury, COVID-19, burns, traumatic brain injury (TBI) and other chronic infections.


Asunto(s)
Quemaduras , COVID-19 , Tratamiento Basado en Trasplante de Células y Tejidos , Humanos , Lesiones Traumáticas del Encéfalo/terapia , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/complicaciones , Quemaduras/terapia , Quemaduras/inmunología , Quemaduras/complicaciones , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedad Crónica , COVID-19/inmunología , COVID-19/terapia , Enfermedad Crítica , Sistema Inmunológico , Infecciones/terapia , Infecciones/inmunología , Infecciones/etiología , SARS-CoV-2 , Heridas y Lesiones/terapia , Heridas y Lesiones/inmunología , Heridas y Lesiones/complicaciones , Congresos como Asunto
11.
Front Immunol ; 14: 1074207, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36761764

RESUMEN

Introduction: Clinical and preclinical data suggest accelerated bone fracture healing in subjects with an additional traumatic brain injury (TBI). Mechanistically, altered metabolism and neuro-endocrine regulations have been shown to influence bone formation after combined fracture and TBI, thereby increasing the bone content in the fracture callus. However, the early inflammatory response towards fracture and TBI has not been investigated in detail so far. This is of great importance, since the early inflammatory phase of fracture healing is known to be essential for the initiation of downstream regenerative processes for adequate fracture repair. Methods: Therefore, we analyzed systemic and local inflammatory mediators and immune cells in mice which were exposed to fracture only or fracture + TBI 6h and 24h after injury. Results: We found a dysregulated systemic immune response and significantly fewer neutrophils and mast cells locally in the fracture hematoma. Further, local CXCL10 expression was significantly decreased in the animals with combined trauma, which correlated significantly with the reduced mast cell numbers. Discussion: Since mast cells and mast cell-derived CXCL10 have been shown to increase osteoclastogenesis, the reduced mast cell numbers might contribute to higher bone content in the fracture callus of fracture + TBI mice due to decreased callus remodeling.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Fracturas Óseas , Animales , Ratones , Callo Óseo/metabolismo , Lesiones Traumáticas del Encéfalo/inmunología , Curación de Fractura , Fracturas Óseas/inmunología , Osteogénesis/fisiología
12.
J Neuroinflammation ; 19(1): 289, 2022 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-36463233

RESUMEN

BACKGROUND: Neuroinflammation is one of the most important processes in secondary injury after traumatic brain injury (TBI). Triggering receptor expressed on myeloid cells 2 (TREM2) has been proven to exert neuroprotective effects in neurodegenerative diseases and stroke by modulating neuroinflammation, and promoting phagocytosis and cell survival. However, the role of TREM2 in TBI has not yet been elucidated. In this study, we are the first to use COG1410, an agonist of TREM2, to assess the effects of TREM2 activation in a murine TBI model. METHODS: Adult male wild-type (WT) C57BL/6 mice and adult male TREM2 KO mice were subjected to different treatments. TBI was established by the controlled cortical impact (CCI) method. COG1410 was delivered 1 h after CCI via tail vein injection. Western blot analysis, immunofluorescence, laser speckle contrast imaging (LSCI), neurological behaviour tests, brain electrophysiological monitoring, Evans blue assays, magnetic resonance imaging (MRI), and brain water content measurement were performed in this study. RESULTS: The expression of endogenous TREM2 peaked at 3 d after CCI, and it was mainly expressed on microglia and neurons. We found that COG1410 improved neurological functions within 3 d, as well as neurological functions and brain electrophysiological activity at 2 weeks after CCI. COG1410 exerted neuroprotective effects by inhibiting neutrophil infiltration and microglial activation, and suppressing neuroinflammation after CCI. In addition, COG1410 treatment alleviated blood brain barrier (BBB) disruption and brain oedema; furthermore, COG1410 promoted cerebral blood flow (CBF) recovery at traumatic injury sites after CCI. In addition, COG1410 suppressed neural apoptosis at 3 d after CCI. TREM2 activation upregulated p-Akt, p-CREB, BDNF, and Bcl-2 and suppressed TNF-α, IL-1ß, Bax, and cleaved caspase-3 at 3 d after CCI. Moreover, TREM2 knockout abolished the effects of COG1410 on vascular phenotypes and microglial states. Finally, the neuroprotective effects of COG1410 were suppressed by TREM2 depletion. CONCLUSIONS: Altogether, we are the first to demonstrate that TREM2 activation by COG1410 alleviated neural damage through activation of Akt/CREB/BDNF signalling axis in microglia after CCI. Finally, COG1410 treatment improved neurological behaviour and brain electrophysiological activity after CCI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Animales , Masculino , Ratones , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/inmunología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/inmunología , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores Inmunológicos/agonistas , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Modelos Animales de Enfermedad , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/genética , Enfermedades Neuroinflamatorias/inmunología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/inmunología , Sistema Nervioso/efectos de los fármacos , Sistema Nervioso/inmunología
13.
Neuroimmunomodulation ; 29(4): 439-449, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35609523

RESUMEN

BACKGROUND: Neuroinflammation subsequent to traumatic brain injury (TBI) is important for the recovery of patients and is associated with neurodegenerative changes post-TBI. The tripartite motif containing 44 (TRIM44) protein is an E3 ligase involved in the regulation of immune function with no previously known link to TBI. This study explores the connection between TRIM44 and TBI. METHODS: After induction of TBI in rats by control cortex injury, TRIM44 expressions were determined with quantitative real-time reverse transcription polymerase chain reaction and Western blot, and Toll-like receptor 4 (TLR4)-NF-κB signaling was examined by the expression of TLR4, p65 phosphorylation, and the specific NF-κB transcription activity. The effects of TRIM44 knockdown on inflammation, neurological function, and TLR4-NF-κB signaling in TBI rats were revealed by the detection of proinflammatory cytokines and TLR4-NF-κB signaling molecules, modified neurological severity score, brain water content, and Evans blue permeability. RESULTS: We found that TRIM44 expression was significantly increased following TBI induction along with TLR4-NF-κB activation. Silencing of TRIM44 suppressed proinflammatory cytokine production, improved neurological outcomes, alleviated brain edema, and inhibited TLR4-NF-κB signaling in TBI rats. CONCLUSION: Our findings suggest that suppressing TRIM44 or modulation of relevant pathways may be a therapeutic strategy for TBI.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Inflamación , Proteínas de Motivos Tripartitos , Animales , Ratas , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/inmunología , Inflamación/genética , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , FN-kappa B/genética , FN-kappa B/inmunología , Ratas Sprague-Dawley , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/inmunología
14.
Brain Res Bull ; 181: 46-54, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35077842

RESUMEN

The aim of the current study was to determine the effects of cerebral contusion injury with purinergic adenosine triphosphate Y1 (P2Y1) receptor blockers on postinjury inflammatory responses. Adenosine triphosphate (ATP) is released into the extracellular space in several in vivo models, including traumatic brain injury. Released ATP triggers neuroinflammation via activation of microglial cells. P2Y1 receptor blockers were reported to suppress extracellular ATP elevation in several disease models through inhibition of cellular ATP release. In addition to the beneficial effects of inflammation, excess inflammatory reactions cause secondary damage and aggravate outcomes. Here, we assessed the effect of the selective P2Y1 receptor blocker MRS2179 on its potential to prevent posttraumatic inflammation in a rat cerebral contusion model. Cerebral contusion injury was induced in the rat cerebral cortex. Either MRS2179 or artificial cerebral spinal fluid as a control was administered in situ into the center of contused tissue via a subcutaneously implanted osmotic pump. Galectin 3, a marker of microglia and proinflammatory cytokines, was measured 1, 3 and 7 days following injury. Another group of rats was assessed for behavioral performance up to 28 days after injury, including the beam walk test, neurological response test and plus maze test. The Galectin 3 levels in the cortex around the contusion cavity and in the cortex far from the contusion cavity were significantly suppressed by MRS2179 administration on postinjury Days 1 and 3 (p < 0.05). However, administration of MRS2179 failed to improve behavioral outcome. Administration of MRS2179 successfully suppressed microglial activation in a traumatic brain injury model, which will be a potent treatment option in the future. Further study is required to conclude its therapeutic effects.


Asunto(s)
Adenosina Difosfato/análogos & derivados , Antiinflamatorios/farmacología , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/lesiones , Galectina 3/efectos de los fármacos , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Antagonistas del Receptor Purinérgico P2Y/farmacología , Adenosina Difosfato/farmacología , Animales , Conducta Animal/efectos de los fármacos , Lesiones Traumáticas del Encéfalo/complicaciones , Lesiones Traumáticas del Encéfalo/inmunología , Corteza Cerebral/inmunología , Modelos Animales de Enfermedad , Microglía/efectos de los fármacos , Enfermedades Neuroinflamatorias/etiología , Enfermedades Neuroinflamatorias/inmunología , Ratas
15.
PLoS Biol ; 20(1): e3001456, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35081110

RESUMEN

In traumatic brain injury (TBI), the initial injury phase is followed by a secondary phase that contributes to neurodegeneration, yet the mechanisms leading to neuropathology in vivo remain to be elucidated. To address this question, we developed a Drosophila head-specific model for TBI termed Drosophila Closed Head Injury (dCHI), where well-controlled, nonpenetrating strikes are delivered to the head of unanesthetized flies. This assay recapitulates many TBI phenotypes, including increased mortality, impaired motor control, fragmented sleep, and increased neuronal cell death. TBI results in significant changes in the transcriptome, including up-regulation of genes encoding antimicrobial peptides (AMPs). To test the in vivo functional role of these changes, we examined TBI-dependent behavior and lethality in mutants of the master immune regulator NF-κB, important for AMP induction, and found that while sleep and motor function effects were reduced, lethality effects were enhanced. Similarly, loss of most AMP classes also renders flies susceptible to lethal TBI effects. These studies validate a new Drosophila TBI model and identify immune pathways as in vivo mediators of TBI effects.


Asunto(s)
Lesiones Traumáticas del Encéfalo/patología , Drosophila melanogaster , Neuroglía/inmunología , Animales , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/mortalidad , Modelos Animales de Enfermedad , Inmunidad Innata , Locomoción , Masculino , Mutación , FN-kappa B/genética , FN-kappa B/metabolismo , Trastornos del Sueño-Vigilia , Transcriptoma
16.
Int Immunopharmacol ; 103: 108219, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34953447

RESUMEN

Neuronal death and neuroinflammation play critical roles in regulating the progression of traumatic brain injury (TBI). However, associated pathogenesis has not been fully understood. Tumor necrosis factor receptor-associated factor 7 (TRAF7), as the unique noncanonical member of the TRAF family, mediates various essential biological processes. Nevertheless, the effects of TRAF7 on TBI are still unclear. In this study, we showed that TRAF7 expression was markedly up-regulated in cortex and hippocampus of mice after TBI. Brain-specific TRAF7 deletion markedly ameliorated neuronal death in cortical and hippocampal samples of TBI mice, accompanied with cognitive impairments and motor dysfunction. Moreover, the aberrant activation of astrocyte and microglia in cortex and hippocampus of TBI mice was significantly restrained by TRAF7 conditional knockout in brain, as indicated by the increased expression of GFAP and Iba1. In addition, the releases of pro-inflammatory factors caused by TBI were also considerably diminished by brain-specific TRAF7 knockout, which were largely through the blockage of nuclear factor-κB (NF-κB) and mitogen-activated protein kinases (MAPKs) signaling pathways. Importantly, mitogen-activated protein kinase kinase kinase 3 (MEKK3) expression levels were greatly enhanced in cortex and hippocampus of mice with TBI, while being dramatically ameliorated by TRAF7 knockout in brain. Mechanistically, we showed that TRAF7 directly interacted with MEKK3. Of note, MEKK3 over-expression almost abrogated the capacity of TRAF7 knockout to mitigate neuronal death and neuroinflammation in the isolated primary cortical neurons and glial cells upon oxygen-glucose-deprivation/reperfusion (OGD/R) stimulation. Collectively, TRAF7 may be an important molecular switch that leads to TBI in a MEKK3-dependent manner, and can be served as a therapeutic target for TBI treatment.


Asunto(s)
Lesiones Traumáticas del Encéfalo/inmunología , Encéfalo/fisiología , Neuroglía/fisiología , Neuronas/fisiología , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Apoptosis , Células Cultivadas , Humanos , Terapia de Inmunosupresión , MAP Quinasa Quinasa Quinasa 3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Inflamación Neurogénica , Especificidad de Órganos , Eliminación de Secuencia , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética
17.
Exp Neurol ; 348: 113943, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34863998

RESUMEN

Astrocytes control many processes of the nervous system in health and disease, and respond to injury quickly. Astrocytes produce neuroprotective factors in the injured brain to clear cellular debris and to orchestrate neurorestorative processes that are beneficial for neurological recovery after traumatic brain injury (TBI). However, astrocytes also become dysregulated and produce cytotoxic mediators that hinder CNS repair by induction of neuronal dysfunction and cell death. Hence, we discuss the potential role of astrocytes in neuropathological processes such as neuroinflammation, neurogenesis, synaptogenesis and blood-brain barrier repair after TBI. Thus, an improved understanding of the dual role of astrocytes may advance our knowledge of post-brain injury recovery, and provide opportunities for the development of novel therapeutic strategies for TBI.


Asunto(s)
Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Encéfalo/metabolismo , Mediadores de Inflamación/metabolismo , Animales , Astrocitos/inmunología , Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/inmunología , Muerte Celular/fisiología , Humanos , Mediadores de Inflamación/inmunología , Neurogénesis/fisiología , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/metabolismo
18.
Int J Med Sci ; 18(16): 3644-3651, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790036

RESUMEN

Emerging evidence suggests that immune-inflammatory processes are key elements in the physiopathological events associated with traumatic brain injury (TBI). TBI is followed by T-cell-specific immunological changes involving several subsets of T-helper cells and the cytokines they produce; these processes can have opposite effects depending on the disease course and cytokine concentrations. Efforts are underway to identify the T-helper cells and cytokine profiles associated with prognosis. These predictors may eventually serve as effective treatment targets to decrease morbidity and mortality and to improve the management of TBI patients. Here, we review the immunological response to TBI, the possible molecular mechanisms of this response, and therapeutic strategies to address it.


Asunto(s)
Lesiones Traumáticas del Encéfalo/inmunología , Sistema Inmunológico/fisiología , Linfocitos T Colaboradores-Inductores/fisiología , Lesiones Traumáticas del Encéfalo/diagnóstico , Lesiones Traumáticas del Encéfalo/patología , Humanos , Inflamación/inmunología , Inflamación/patología
19.
J Neuroimmunol ; 361: 577723, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34619426

RESUMEN

BACKGROUND: Traumatic brain injury (TBI) is a common cause of morbidity and mortality. We have previously shown that TBI with a concurrent extra-cranial injury reliably leads to post-injury suppression of the innate immune system, but the impact of this injury on the adaptive immune system is unknown. We present data showing that combined injury reduced immune response as assayed in both blood and spleen samples and that these changes parallel apoptosis in the spleen. To assess the clinical relevance of these changes, we examined lungs for spontaneous bacterial colonization. METHODS: For these studies, prepubescent (28 day old) rats were injured using a controlled cortical impact model and then 25% blood volume removal by arteriotomy, and injured animals were compared with sham injured animals. Blood and spleen samples at post-injury day 1 were incubated with or without immunostimulant and examined for IFN-γ production using an Eli-Spot assay. Spleen samples were also examined for apoptosis using Annexin V staining, and lungs were harvested and plated on blood agar to examine for spontaneous bacterial colonization. RESULTS: Stimulations of whole blood and spleen samples with phorbol 12-myristate 13-acetate/ionomycin (PMA/I) at post-injury day 1 were associated with significant decreases in IFN-γ-positive cells/million in injured animals. Stimulation of whole blood with either PMA/I or pokeweed mitogen led to reduced tumor necrosis factor alpha production. Spleen from injured animals showed a marked increase in apoptosis. Lung samples showed a 300% increase in colonies per plate in injured animals. CONCLUSIONS: These data suggest that the combined injury can lead to adaptive immunosuppression, and our findings further suggest a potential role for the spleen in altering leukocyte function following injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/inmunología , Hemorragia Cerebral/inmunología , Tolerancia Inmunológica , Traumatismo Múltiple/inmunología , Bazo/inmunología , Inmunidad Adaptativa , Factores de Edad , Animales , Apoptosis , Carga Bacteriana , Lesiones Traumáticas del Encéfalo/complicaciones , Hemorragia Cerebral/etiología , Modelos Animales de Enfermedad , Ensayos de Liberación de Interferón gamma , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Pulmón/microbiología , Masculino , Mitógenos de Phytolacca americana/farmacología , Ratas , Método Simple Ciego , Bazo/patología , Acetato de Tetradecanoilforbol/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis
20.
Nat Immunol ; 22(10): 1280-1293, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34556874

RESUMEN

Traumatic brain injury (TBI) and cerebrovascular injury are leading causes of disability and mortality worldwide. Systemic infections often accompany these disorders and can worsen outcomes. Recovery after brain injury depends on innate immunity, but the effect of infections on this process is not well understood. Here, we demonstrate that systemically introduced microorganisms and microbial products interfered with meningeal vascular repair after TBI in a type I interferon (IFN-I)-dependent manner, with sequential infections promoting chronic disrepair. Mechanistically, we discovered that MDA5-dependent detection of an arenavirus encountered after TBI disrupted pro-angiogenic myeloid cell programming via induction of IFN-I signaling. Systemic viral infection similarly blocked restorative angiogenesis in the brain parenchyma after intracranial hemorrhage, leading to chronic IFN-I signaling, blood-brain barrier leakage and a failure to restore cognitive-motor function. Our findings reveal a common immunological mechanism by which systemic infections deviate reparative programming after central nervous system injury and offer a new therapeutic target to improve recovery.


Asunto(s)
Antiinfecciosos/inmunología , Lesiones Traumáticas del Encéfalo/inmunología , Sistema Nervioso Central/inmunología , Inmunidad Innata/inmunología , Animales , Barrera Hematoencefálica/inmunología , Encéfalo/inmunología , Modelos Animales de Enfermedad , Femenino , Interferón Tipo I/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA