Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros













Intervalo de año de publicación
1.
Life Sci ; 349: 122721, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38754813

RESUMEN

AIMS: Infection is a complication after stroke and outcomes vary by sex. Thus, we investigated if sepsis affects brain from ischemic stroke and sex involvement. MAIN METHODS: Male and female Wistar rats, were submitted to middle cerebral artery occlusion (MCAO) and after 7 days sepsis to cecal ligation and perforation (CLP). Infarct size, neuroinflammation, oxidative stress, and mitochondrial activity were quantified 24 h after CLP in the prefrontal cortex and hippocampus. Survival and neurological score were assessed up to 15 days after MCAO or 8 days after CLP (starting at 2 h after MCAO) and memory at the end. KEY FINDINGS: CLP decreased survival, increased neurological impairments in MCAO females. Early, in male sepsis following MCAO led to increased glial activation in the brain structures, and increased TNF-α and IL-1ß in the hippocampus. All groups had higher IL-6 in both tissues, but the hippocampus had lower IL-10. CLP potentiated myeloperoxidase (MPO) in the prefrontal cortex of MCAO male and female. In MCAO+CLP, only male increased MPO and nitrite/nitrate in hippocampus. Males in all groups had protein oxidation in the prefrontal cortex, but only MCAO+CLP in the hippocampus. Catalase decreased in the prefrontal cortex and hippocampus of all males and females, and MCAO+CLP only increased this activity in males. Female MCAO+CLP had higher prefrontal cortex complex activity than males. In MCAO+CLP-induced long-term memory impairment only in females. SIGNIFICANCE: The parameters evaluated for early sepsis after ischemic stroke show a worse outcome for males, while females are affected during long-term follow-up.


Asunto(s)
Accidente Cerebrovascular Isquémico , Ratas Wistar , Sepsis , Caracteres Sexuales , Animales , Masculino , Femenino , Sepsis/complicaciones , Sepsis/metabolismo , Ratas , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/complicaciones , Accidente Cerebrovascular Isquémico/patología , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Estrés Oxidativo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Recuperación de la Función , Factores Sexuales , Isquemia Encefálica/metabolismo , Isquemia Encefálica/complicaciones , Peroxidasa/metabolismo
2.
Chem Biodivers ; 21(5): e202301260, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38513005

RESUMEN

Microglia are key immune cells in the brain that maintain homeostasis and defend against immune threats. Targeting the dysfunctional microglia is one of the most promising approaches to inhibit neuroinflammation. In the current study, a diverse series of molecular hybrids were designed and screened through molecular docking against two neuroinflammatory targets, namely HMGB1 (2LY4) and HMGB1 Box A (4QR9) proteins. Based on the outcomes of docking scores fifteen compounds; ten furanyl-pyrazolyl acetamides 11(a-j), and five 2,4-thiazolidinyl-furan-3-carboxamide 15(a-e) derivatives were selected for further synthesis, followed by biological evaluation. The selected compounds, 11(a-j) and 15(a-e) were successfully synthesized with moderate to good yields, and structures were confirmed by IR, NMR, and mass spectra. The in-vitro cytotoxicity was evaluated on microglial cells namely BV-2, N-9, HMO6, leukemic HAP1, and human fibroblast cells. Further western-blot analysis revealed that 11h, 11f, 11c, 11j, 15d, 15c, 15e, and 15b compounds significantly suppressed anti-inflammatory markers such as TNF-α, IL-1, IL-6, and Bcl-2. All derivatives were moderate in potency compared to reference doxorubicin and could potentially act as novel anti-neuroinflammatory agents. This study can act as a beacon for further research in the application of furan-pyrazole and furan-2,4-thiazolidinediones as lead moieties for anti-neuroinflammatory and related diseases.


Asunto(s)
Acetamidas , Furanos , Simulación del Acoplamiento Molecular , Humanos , Furanos/química , Furanos/farmacología , Furanos/síntesis química , Acetamidas/farmacología , Acetamidas/química , Acetamidas/síntesis química , Relación Estructura-Actividad , Microglía/efectos de los fármacos , Microglía/metabolismo , Pirazoles/química , Pirazoles/farmacología , Pirazoles/síntesis química , Estructura Molecular , Animales , Ratones , Supervivencia Celular/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/síntesis química , Fármacos Neuroprotectores/química , Línea Celular , Relación Dosis-Respuesta a Droga , Antiinflamatorios/farmacología , Antiinflamatorios/síntesis química , Antiinflamatorios/química
3.
Mol Neurobiol ; 61(1): 239-251, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37603152

RESUMEN

Bacterial meningitis is considered a life-threatening condition with high mortality rates. In response to the infection, signaling cascades, producing pro-inflammatory mediators trigger an exacerbated host immune response. Another inflammatory pathway occurs through the activation of inflammasomes. Studies highlight the role of the NLR family pyrin domain containing 3 (NLRP3) in central nervous system disorders commonly involved in neuroinflammation. We aimed to investigate the role of NLRP3 and its inhibitor MCC950 on neurochemical, immunological, and behavioral parameters in the early and late stages of experimental pneumococcal meningitis. For this, adult male Wistar rats received an intracisternal injection of Streptococcus pneumoniae or artificial cerebrospinal fluid as a placebo. The animals were divided into control/saline, control/MCC950, meningitis/saline, and meningitis/MCC950. Immediately after the meningitis induction, the animals received 140 ng/kg MCC950 via intracisternal injection. For the acute protocol, 24 h after induction, brain structures were collected to evaluate cytokines, NLRP3, and microglia. In the long-term group, the animals were submitted to open field and recognition of new objects tests at ten days after the meningitis induction. After the behavioral tests, the same markers were evaluated. The animals in the meningitis group at 24 h showed increased levels of cytokines, NLRP3, and IBA-1 expression, and the use of the MCC950 significantly reduced those levels. Although free from infection, ten days after meningitis induction, the animals in the meningitis group had elevated cytokine levels and demonstrated behavioral deficits; however, the single dose of NLRP3 inhibitor rescued the behavior deficits and decreased the brain inflammatory profile.


Asunto(s)
Meningitis Neumocócica , Animales , Masculino , Ratas , Citocinas/metabolismo , Inflamasomas/metabolismo , Trastornos de la Memoria , Meningitis Neumocócica/complicaciones , Meningitis Neumocócica/tratamiento farmacológico , Modelos Teóricos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ratas Wistar , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico
4.
Mol Psychiatry ; 28(10): 4463-4473, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37452088

RESUMEN

While our understanding of the molecular biology of Alzheimer's disease (AD) has grown, the etiology of the disease, especially the involvement of peripheral infection, remains a challenge. In this study, we hypothesize that peripheral infection represents a risk factor for AD pathology. To test our hypothesis, APP/PS1 mice underwent cecal ligation and puncture (CLP) surgery to develop a polymicrobial infection or non-CLP surgery. Mice were euthanized at 3, 30, and 120 days after surgery to evaluate the inflammatory mediators, glial cell markers, amyloid burden, gut microbiome, gut morphology, and short-chain fatty acids (SCFAs) levels. The novel object recognition (NOR) task was performed 30 and 120 days after the surgery, and sepsis accelerated the cognitive decline in APP/PS1 mice at both time points. At 120 days, the insoluble Aß increased in the sepsis group, and sepsis modulated the cytokines/chemokines, decreasing the cytokines associated with brain homeostasis IL-10 and IL-13 and increasing the eotaxin known to influence cognitive function. At 120 days, we found an increased density of IBA-1-positive microglia in the vicinity of Aß dense-core plaques, compared with the control group confirming the predictable clustering of reactive glia around dense-core plaques within 15 µm near Aß deposits in the brain. In the gut, sepsis negatively modulated the α- and ß-diversity indices evaluated by 16S rRNA sequencing, decreased the levels of SCFAs, and significantly affected ileum and colon morphology in CLP mice. Our data suggest that sepsis-induced peripheral infection accelerates cognitive decline and AD pathology in the AD mouse model.


Asunto(s)
Enfermedad de Alzheimer , Microbioma Gastrointestinal , Sepsis , Ratones , Animales , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Enfermedades Neuroinflamatorias , ARN Ribosómico 16S , Ratones Transgénicos , Amiloide , Citocinas , Placa Amiloide , Sepsis/complicaciones , Péptidos beta-Amiloides , Modelos Animales de Enfermedad
5.
Transl Neurodegener ; 12(1): 37, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37496074

RESUMEN

A wealth of pre-clinical reports and data derived from human subjects and brain autopsies suggest that microbial infections are relevant to Alzheimer's disease (AD). This has inspired the hypothesis that microbial infections increase the risk or even trigger the onset of AD. Multiple models have been developed to explain the increase in pathogenic microbes in AD patients. Although this hypothesis is well accepted in the field, it is not yet clear whether microbial neuroinvasion is a cause of AD or a consequence of the pathological changes experienced by the demented brain. Along the same line, the gut microbiome has also been proposed as a modulator of AD. In this review, we focus on human-based evidence demonstrating the elevated abundance of microbes and microbe-derived molecules in AD hosts as well as their interactions with AD hallmarks. Further, the direct-purpose and potential off-target effects underpinning the efficacy of anti-microbial treatments in AD are also addressed.


Asunto(s)
Enfermedad de Alzheimer , Humanos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Encéfalo/patología
6.
Clin Sci (Lond) ; 137(6): 401-414, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36942500

RESUMEN

Sepsis is a life-threatening organ dysfunction triggered by a dysregulated host immune response to eliminate an infection. After the host immune response is activated, a complex, dynamic, and time-dependent process is triggered. This process promotes the production of inflammatory mediators, including acute-phase proteins, complement system proteins, cytokines, chemokines, and antimicrobial peptides, which are required to initiate an inflammatory environment for eliminating the invading pathogen. The physiological response of this sepsis-induced systemic inflammation can affect blood-brain barrier (BBB) function; subsequently, endothelial cells produce inflammatory mediators, including cytokines, chemokines, and matrix metalloproteinases (MMPs) that degrade tight junction (TJ) proteins and decrease BBB function. The resulting BBB permeability allows peripheral immune cells from the bloodstream to enter the brain, which then release a range of inflammatory mediators and activate glial cells. The activated microglia and astrocytes release reactive oxygen species (ROS), cytokines, chemokines, and neurochemicals, initiate mitochondrial dysfunction and neuronal damage, and exacerbate the inflammatory milieu in the brain. These changes trigger sepsis-associated encephalopathy (SAE), which has the potential to increase cognitive deterioration and susceptibility to cognitive decline later in life.


Asunto(s)
Células Endoteliales , Sepsis , Humanos , Células Endoteliales/metabolismo , Encéfalo/metabolismo , Barrera Hematoencefálica/metabolismo , Sepsis/complicaciones , Sepsis/metabolismo , Citocinas/metabolismo , Quimiocinas/metabolismo , Mediadores de Inflamación/metabolismo
8.
Drug Discov Today ; 27(11): 103334, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35998800

RESUMEN

In the past three decades, research on the gut microbiome and its metabolites, such as trimethylamines (TMA), trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), branched-chain amino acids (BCAAs), bile acids, tryptophan and indole derivatives, has attracted the attention of many scientists and industrialists. Among these metabolites, TMAO is produced from dietary choline, phosphatidylcholine, carnitine,andbetaine. TMAO and other gut metabolites, such as TMA and SCFAs, reach the brain by crossing the blood-brain barrier (BBB) and are involved in brain development, neurogenesis, and behavior. Gut-microbiota composition is influenced by diet, lifestyle, antibiotics, and age. Several studies have confirmed that altered TMAO levels contribute to metabolic, vascular, psychiatric, and neurodegenerative disorders. This review focuses on how altered TMAO levels impact oxidative stress, microglial activation, and the apoptosis of neurons, and may lead to neuroinflammation, which can subsequently result in the development of psychiatric, cognitive, and behavioral disorders.

10.
Neurosci Res ; 181: 17-21, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35577241

RESUMEN

Alzheimer's disease (AD) and related dementias (ADRD) are still a serious global public health concern more than a century after the German neuropathologist and psychiatrist Dr. Aloysius Alzheimer described the first case. The World Health Organization (WHO) estimates that over 50 million people worldwide suffer from dementia, with AD accounting for 60-70% of all cases. In addition, the global dementia epidemic is estimated to affect 82 million individuals by 2030 and 152 million by 2050. Along with genetic factors, environmental factors, and aging also increase the risks of developing neurodegenerative disorders. For example, gut microbiota can serve as non-genetic factors that define a threshold for maintaining a homeostatic balance or developing illnesses. The scientific community has explored and identified that patients with AD often present with dysbiosis of the bowel and dysregulated gastrointestinal (GI) tract. Research describes it as a bidirectional relationship by which the brain communicates with the gut's microbiome through the vagus nerve, immune and neuroimmune systems, enteroendocrine system, neurotransmitters, branched-chain amino acids, short-chain fatty acids (SCFAs), agonists of aryl hydrocarbon receptors (AHRs), bile acids, and the hypothalamic-pituitary-adrenal (HPA) axis. In this narrative review, we explore and clarify the involvement of the microbiota-gut-brain axis in AD pathology.


Asunto(s)
Enfermedad de Alzheimer , Microbioma Gastrointestinal , Microbiota , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Eje Cerebro-Intestino , Microbioma Gastrointestinal/fisiología , Humanos
11.
J Neuroinflammation ; 19(1): 114, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35606817

RESUMEN

BACKGROUND: Sepsis is a potentially fatal disease characterized by acute organ failure that affects more than 30 million people worldwide. Inflammation is strongly associated with sepsis, and patients can experience impairments in memory, concentration, verbal fluency, and executive functioning after being discharged from the hospital. We hypothesize that sepsis disrupts the microbiota-gut-brain axis homeostasis triggering cognitive impairment. This immune activation persists during treatment, causing neurological dysfunction in sepsis survivors. METHODS: To test our hypothesis, adult Wistar rats were subjected to cecal-ligation and perforation (CLP) or sham (non-CLP) surgeries. The animals were subjected to the [11C]PBR28 positron emission tomography (PET)/computed tomography (CT) imaging at 24 h and 10 days after CLP and non-CLP surgeries. At 24 h and 10 days after surgery, we evaluated the gut microbiome, bacterial metabolites, cytokines, microglia, and astrocyte markers. Ten days after sepsis induction, the animals were subjected to the novel object recognition (NOR) and the Morris water maze (MWM) test to assess their learning and memory. RESULTS: Compared to the control group, the 24-h and 10-day CLP groups showed increased [11C]PBR28 uptake, glial cells count, and cytokine levels in the brain. Results show that sepsis modulates the gut villus length and crypt depth, alpha and beta microbial diversities, and fecal short-chain fatty acids (SCFAs). In addition, sepsis surviving animals showed a significant cognitive decline compared with the control group. CONCLUSIONS: Since several pharmacological studies have failed to prevent cognitive impairment in sepsis survivors, a better understanding of the function of glial cells and gut microbiota can provide new avenues for treating sepsis patients.


Asunto(s)
Eje Cerebro-Intestino , Disfunción Cognitiva , Sepsis , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Disfunción Cognitiva/diagnóstico por imagen , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Citocinas/metabolismo , Microbioma Gastrointestinal , Humanos , Ratas , Ratas Wistar , Sepsis/complicaciones , Sepsis/tratamiento farmacológico
12.
Crit Care Med ; 50(3): e241-e252, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34402457

RESUMEN

OBJECTIVES: Sepsis is a life-threatening organ dysfunction caused by a host's unregulated immune response to eliminate the infection. After hospitalization, sepsis survivors often suffer from long-term impairments in memory, attention, verbal fluency, and executive functioning. To understand the effects of sepsis and the exacerbated peripheral inflammatory response in the brain, we asked the question: What are the findings and inflammatory markers in the brains of deceased sepsis patients? To answer this question, we conducted this systematic review by the recommendations of Preferred Reporting Items for Systematic Reviews and Meta-Analyses. DATA SOURCES: Relevant studies were identified by searching the PubMed/National Library of Medicine, PsycINFO, EMBASE, Bibliographical Index in Spanish in Health Sciences, Latin American and Caribbean Health Sciences Literature, and Web of Science databases for peer-reviewed journal articles published on April 05, 2021. STUDY SELECTION: A total of 3,745 articles were included in the primary screening; after omitting duplicate articles, animal models, and reviews, 2,896 articles were selected for the study. These studies were selected based on the title and abstract, and 2,772 articles were still omitted based on the exclusion criteria. DATA EXTRACTION: The complete texts of the remaining 124 articles were obtained and thoroughly evaluated for the final screening, and 104 articles were included. DATA SYNTHESIS: The postmortem brain had edema, abscess, hemorrhagic and ischemic injuries, infarction, hypoxia, atrophy, hypoplasia, neuronal loss, axonal injuries, demyelination, and necrosis. CONCLUSIONS: The mechanisms by which sepsis induces brain dysfunction are likely to include vascular and neuronal lesions, followed by the activation of glial cells and the presence of peripheral immune cells in the brain.


Asunto(s)
Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Inflamación/diagnóstico por imagen , Inflamación/metabolismo , Sepsis/metabolismo , Sepsis/patología , Atrofia/patología , Autopsia , Biomarcadores , Encéfalo/patología , Humanos , Inflamación/patología , Imagen por Resonancia Magnética , Sepsis/diagnóstico por imagen
14.
Int J Dev Neurosci ; 81(5): 407-415, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33788296

RESUMEN

Maternal deprivation (MD) is known to be related to long-term changes that could influence the onset of psychiatric disorders. Studies have demonstrated that early life stress makes the cells in the brain more susceptible to subsequent stressors. To test it, we used an animal model of MD conducted from postnatal day (PND) 1 to 10. Deprived and non-deprived rats (control) were randomized to receive or not lipopolysaccharide (LPS) at 5 mg/kg on PND 50. The behavior and glial cells activation were evaluated in all groups from 51 to 53 PND. There was an increase in the immobility time in the MD and MD+LPS groups. The spontaneous locomotor activity was not changed between groups. We found elevated ionized calcium-binding adapter molecule 1 (Iba-1)-positive cells levels in the control+LPS and MD+LPS groups. In the MD+LPS group, it was found an increase in Iba-positive cells compared to the MD+sal group. The glial fibrillary acidic protein (GFAP)-positive cells were also increased in the MD+LPS, compared to control+sal, control+LPS, and MD+sal groups. Immune challenge by LPS in late adolescence, which was subjected to MD, did not influence the depressive-like behavior but exerted a pronounced effect in the microglial activation and astrocyte atrophy.


Asunto(s)
Conducta Animal , Inmunidad , Privación Materna , Neuroglía , Estrés Psicológico , Animales , Femenino , Ratas , Astrocitos/patología , Proteínas de Unión al Calcio/metabolismo , Depresión , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/biosíntesis , Inmunidad/fisiología , Lipopolisacáridos , Activación de Macrófagos , Proteínas de Microfilamentos/metabolismo , Actividad Motora , Neuroglía/inmunología , Ratas Wistar , Estrés Psicológico/inmunología , Natación/psicología
16.
Neurotox Res ; 39(2): 119-132, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33025358

RESUMEN

Sepsis-associated encephalopathy is a serious consequence of sepsis, triggered by the host response against an infectious agent, that can lead to brain damage and cognitive impairment. Several mechanisms have been proposed in this bidirectional communication between the immune system and the brain after sepsis as neuroinflammation, oxidative stress, and mitochondrial dysfunction. Stanniocalcin-1 (STC-1), an endogen neuroprotective protein, acts as an anti-inflammatory and suppresses superoxide generation through induction of uncoupling proteins (UCPs) in the mitochondria. Here, we demonstrated a protective role of STC-1 on inflammatory responses in vitro, in activated microglia stimulated with LPS, and on neuroinflammation, oxidative stress, and mitochondrial function in the hippocampus of rats subjected to an animal model of sepsis by cecal ligation and puncture (CLP), as well the consequences on long-term memory. Recombinant human STC-1 (rhSTC1) suppressed the pro-inflammatory cytokine production in LPS-stimulated microglia without changing the UCP-2 expression. Besides, rhSTC1 injected into the cisterna magna decreased acute hippocampal inflammation and oxidative stress and increased the activity of complex I and II activity of mitochondrial respiratory chain and creatine kinase at 24 h after sepsis. rhSTC1 was effective in preventing long-term cognitive impairment after CLP. In conclusion, rhSTC1 confers significant neuroprotection by inhibiting the inflammatory response in microglia and protecting against sepsis-associated encephalopathy in rats.


Asunto(s)
Encefalitis/prevención & control , Glicoproteínas/administración & dosificación , Microglía/efectos de los fármacos , Microglía/metabolismo , Fármacos Neuroprotectores/administración & dosificación , Encefalopatía Asociada a la Sepsis/prevención & control , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas Wistar
17.
Braz J Psychiatry ; 43(3): 293-305, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32667590

RESUMEN

The microbiota-gut-brain axis is a bidirectional signaling mechanism between the gastrointestinal tract and the central nervous system. The complexity of the intestinal ecosystem is extraordinary; it comprises more than 100 trillion microbial cells that inhabit the small and large intestine, and this interaction between microbiota and intestinal epithelium can cause physiological changes in the brain and influence mood and behavior. Currently, there has been an emphasis on how such interactions affect mental health. Evidence indicates that intestinal microbiota are involved in neurological and psychiatric disorders. This review covers evidence for the influence of gut microbiota on the brain and behavior in Alzheimer disease, dementia, anxiety, autism spectrum disorder, bipolar disorder, major depressive disorder, Parkinson's disease, and schizophrenia. The primary focus is on the pathways involved in intestinal metabolites of microbial origin, including short-chain fatty acids, tryptophan metabolites, and bacterial components that can activate the host's immune system. We also list clinical evidence regarding prebiotics, probiotics, and fecal microbiota transplantation as adjuvant therapies for neuropsychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Depresivo Mayor , Microbioma Gastrointestinal , Encéfalo , Ecosistema , Humanos
18.
Neurotherapeutics ; 18(1): 640-653, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32886341

RESUMEN

Pneumococcal meningitis is a life-threatening infection of the central nervous system (CNS), and half of the survivors of meningitis suffer from neurological sequelae. We hypothesized that pneumococcal meningitis causes CNS inflammation via the disruption of the blood-brain barrier (BBB) and by increasing the receptor for advanced glycation end product (RAGE) expression in the brain, which causes glial cell activation, leading to cognitive impairment. To test our hypothesis, 60-day-old Wistar rats were subjected to meningitis by receiving an intracisternal injection of Streptococcus pneumoniae or artificial cerebrospinal fluid as a control group and were treated with a RAGE-specific inhibitor (FPS-ZM1) in saline. The rats also received ceftriaxone 100 mg/kg intraperitoneally, bid, and fluid replacements. Experimental pneumococcal meningitis triggered BBB disruption after meningitis induction, and FPS-ZM1 treatment significantly suppressed BBB disruption. Ten days after meningitis induction, surviving animals were free from infection, but they presented increased levels of TNF-α and IL-1ß in the prefrontal cortex (PFC); high expression levels of RAGE, amyloid-ß (Aß1-42), and microglial cell activation in the PFC and hippocampus; and memory impairment, as evaluated by the open-field, novel object recognition task and Morris water maze behavioral tasks. Targeted RAGE inhibition was able to reduce cytokine levels, decrease the expression of RAGE and Aß1-42, inhibit microglial cell activation, and improve cognitive deficits in meningitis survivor rats. The sequence of events generated by pneumococcal meningitis can persist long after recovery, triggering neurocognitive decline; however, RAGE blocker attenuated the development of brain inflammation and cognitive impairment in experimental meningitis.


Asunto(s)
Disfunción Cognitiva/etiología , Meningitis Neumocócica/complicaciones , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Animales , Benzamidas/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Western Blotting , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Masculino , Meningitis Neumocócica/tratamiento farmacológico , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Prueba de Campo Abierto/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas , Ratas Wistar , Receptor para Productos Finales de Glicación Avanzada/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
19.
Pharmacol Res ; 164: 105314, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33246175

RESUMEN

The gut microbiota is a complex ecosystem that comprises of more than 100 trillion symbiotic microbial cells. The microbiota, the gut, and the brain form an association, 'the microbiota-gut-brain axis,' and synchronize the gut with the central nervous system and modify the behavior and brain immune homeostasis. The bidirectional communication between gut and brain occurs via the immune system, the vagus nerve, the enteric nervous system, and microbial metabolites, including short-chain fatty acids (SCFAs), proteins, and tryptophan metabolites. Recent studies have implicated the gut microbiota in many neurodegenerative diseases, including Alzheimer's disease (AD). In this review, we present an overview of gut microbiota, including Firmicutes, Bacteroidetes, SCFA, tryptophan, bacterial composition, besides age-related changes in gut microbiota composition, the microbiota-gut-brain axis pathways, the role of gut metabolites in amyloid-beta clearance, and gut microbiota modulation from experimental and clinical AD models. Understanding the role of the microbiota may provide new targets for treatment to delay the onset, progression, or reverse AD, and may help in reducing the prevalence of AD.


Asunto(s)
Enfermedad de Alzheimer/microbiología , Encéfalo , Microbioma Gastrointestinal , Animales , Humanos
20.
Curr Top Behav Neurosci ; 48: 215-237, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33040314

RESUMEN

Bipolar disorder (BD) is a severe, debilitating psychiatric condition with onset in adolescence or young adulthood and often follows a relapsing and remitting course throughout life. The concept of neuroprogression in BD refers to the progressive path with an identifiable trajectory that takes place with recurrent mood episodes, which eventually leads to cognitive, functional, and clinical deterioration in the course of BD. Understanding the biological basis of neuroprogression helps to explain the subset of BD patients who experience worsening of their disorder over time. Additionally, the study of the neurobiological mechanisms underpinning neuroprogression will help BD staging based on systems biology. Replicated epidemiological studies have suggested inflammatory mechanisms as primary contributors to the neuroprogression of mood disorders. It is known that dysregulated inflammatory/immune pathways are often associated with BD pathophysiology. Hence, in this chapter, we focus on the evidence for the involvement of inflammation and immune regulated pathways in the neurobiological consequences of BD neuroprogression. Herein we put forth the evidence of immune markers from autoimmune disorders, chronic infections, and gut-brain axis that lead to BD neuroprogression. Further, we highlighted the peripheral and central inflammatory components measured along with BD progression.


Asunto(s)
Trastorno Bipolar , Adolescente , Adulto , Biomarcadores , Encéfalo , Progresión de la Enfermedad , Humanos , Inflamación , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA