Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.077
Filtrar
1.
Front Immunol ; 15: 1402349, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38938572

RESUMEN

Objective: Immunoregulation is a complex and critical process in the pathological process of spinal cord injury (SCI), which is regulated by various factors and plays an important role in the functional repair of SCI. This study aimed to explore the research hotspots and trends of glial cell immunoregulation after SCI from a bibliometric perspective. Methods: Data on publications related to glial cell immunoregulation after SCI, published from 2004 to 2023, were obtained from the Web of Science Core Collection. Countries, institutions, authors, journals, and keywords in the topic were quantitatively analyzed using the R package "bibliometrix", VOSviewer, Citespace, and the Bibliometrics Online Analysis Platform. Results: A total of 613 papers were included, with an average annual growth rate of 9.39%. The papers came from 36 countries, with the United States having the highest output, initiating collaborations with 27 countries. Nantong University was the most influential institution. We identified 3,177 authors, of whom Schwartz, m, of the Weizmann Institute of Science, was ranked first regarding both field-specific H-index (18) and average number of citations per document (151.44). Glia ranked first among journals with 2,574 total citations. The keywords "microglia," "activation," "macrophages," "astrocytes," and "neuroinflammation" represented recent hot topics and are expected to remain a focus of future research. Conclusion: These findings strongly suggest that the immunomodulatory effects of microglia, astrocytes, and glial cell interactions may be critical in promoting nerve regeneration and repair after SCI. Research on the immunoregulation of glial cells after SCI is emerging, and there should be greater cooperation and communication between countries and institutions to promote the development of this field and benefit more SCI patients.


Asunto(s)
Bibliometría , Neuroglía , Traumatismos de la Médula Espinal , Traumatismos de la Médula Espinal/inmunología , Humanos , Neuroglía/inmunología , Animales , Astrocitos/inmunología
2.
Brain Behav Immun ; 119: 878-897, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38710338

RESUMEN

Metabolites and compounds derived from gut-associated bacteria can modulate numerous physiological processes in the host, including immunity and behavior. Using a model of oral bacterial infection, we previously demonstrated that gut-derived peptidoglycan (PGN), an essential constituent of the bacterial cell envelope, influences female fruit fly egg-laying behavior by activating the NF-κB cascade in a subset of brain neurons. These findings underscore PGN as a potential mediator of communication between gut bacteria and the brain in Drosophila, prompting further investigation into its impact on all brain cells. Through high-resolution mass spectrometry, we now show that PGN fragments produced by gut bacteria can rapidly reach the central nervous system. In Addition, by employing a combination of whole-genome transcriptome analyses, comprehensive genetic assays, and reporter gene systems, we reveal that gut bacterial infection triggers a PGN dose-dependent NF-κB immune response in perineurial glia, forming the continuous outer cell layer of the blood-brain barrier. Furthermore, we demonstrate that persistent PGN-dependent NF-κB activation in perineurial glial cells correlates with a reduction in lifespan and early neurological decline. Overall, our findings establish gut-derived PGN as a critical mediator of the gut-immune-brain axis in Drosophila.


Asunto(s)
Eje Cerebro-Intestino , Encéfalo , Microbioma Gastrointestinal , FN-kappa B , Peptidoglicano , Animales , Peptidoglicano/metabolismo , FN-kappa B/metabolismo , Encéfalo/metabolismo , Encéfalo/inmunología , Microbioma Gastrointestinal/fisiología , Eje Cerebro-Intestino/fisiología , Femenino , Drosophila , Neuroglía/metabolismo , Neuroglía/inmunología , Drosophila melanogaster/metabolismo , Neuronas/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/inmunología , Proteínas de Drosophila/metabolismo
3.
J Autoimmun ; 147: 103256, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38788538

RESUMEN

Parkinson's disease (PD) is a progressive neurodegenerative disorder associated with the loss of dopaminergic neurons and neuroinflammation. Recent studies have identified a role of T cells in the pathogenesis of PD. Additionally, these studies suggested that α-synuclein (α-Syn) is related to abnormal T-cell responses and may act as an epitope and trigger autoimmune T-cell responses. However, it is unclear whether the α-Syn-mediated autoimmune response occurs and whether it is related to neuronal cell death and glial cell activation. In this study, we investigated the autoimmune T-cell response induced by α-Syn peptides and evaluated the neurotoxic effect of the α-Syn peptide-mediated autoimmune response. The immunization of mice with α-Syn peptides resulted in enhanced autoimmune responses, such as the peptide recall response, polarization toward Th1/Th17 cells, and regulatory T cell imbalance. Furthermore, the α-Syn autoimmune response led to the death of primary neurons cocultured with splenocytes. Treatment with conditioned media from α-Syn peptide-immunized splenocytes induced microglia and toxic A1-type astrocyte activation. Taken together, our results provide evidence of the potential role of the α-Syn-initiated autoimmune response and its contribution to neuronal cell death and glial cell activation.


Asunto(s)
Autoinmunidad , Muerte Celular , Neuronas , alfa-Sinucleína , Animales , alfa-Sinucleína/inmunología , alfa-Sinucleína/metabolismo , Ratones , Muerte Celular/efectos de los fármacos , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/patología , Neuroglía/inmunología , Neuroglía/metabolismo , Neuroglía/efectos de los fármacos , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/metabolismo , Ratones Endogámicos C57BL , Humanos , Activación de Linfocitos/inmunología , Activación de Linfocitos/efectos de los fármacos , Péptidos/inmunología , Células Cultivadas , Femenino , Linfocitos T Reguladores/inmunología
4.
J Neuroimmunol ; 390: 578346, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38648696

RESUMEN

The frequency of corticospinal tract (CST) T2/FLAIR hyperintensity in disorders with neuroglial antibodies is unclear. Herein, we retrospectively reviewed brain MRIs of 101 LGI1-antibody encephalitis patients, and observed CST hyperintensity in 30/101 (30%). It was mostly bilateral (93%), not associated with upper motor neuron signs/symptoms (7%), and frequently decreased over time (39%). In a systematic review including patients with other neuroglial antibodies, CST hyperintensity was reported in 110 with neuromyelitis optica (94%), myelin oligodendrocyte glycoprotein-associated disease (2%), Ma2-antibody (3%) and GAD65-antibody paraneoplastic neurological syndrome (1%). CST hyperintensity is not an infrequent finding in LGI1-Ab encephalitis and other disorders with neuroglial antibodies.


Asunto(s)
Autoanticuerpos , Encefalitis , Péptidos y Proteínas de Señalización Intracelular , Tractos Piramidales , Humanos , Autoanticuerpos/inmunología , Autoanticuerpos/sangre , Femenino , Persona de Mediana Edad , Masculino , Estudios Retrospectivos , Anciano , Adulto , Encefalitis/inmunología , Encefalitis/diagnóstico por imagen , Tractos Piramidales/diagnóstico por imagen , Tractos Piramidales/patología , Tractos Piramidales/inmunología , Péptidos y Proteínas de Señalización Intracelular/inmunología , Imagen por Resonancia Magnética , Adulto Joven , Neuroglía/patología , Neuroglía/inmunología , Adolescente , Anciano de 80 o más Años , Enfermedades del Sistema Nervioso Central/inmunología , Enfermedades del Sistema Nervioso Central/diagnóstico por imagen
5.
Int Immunopharmacol ; 132: 111942, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38565045

RESUMEN

Endometriosis (EM) is a gynecological inflammatory disease often accompanied by stress, chronic pelvic pain (CPP), anxiety, and depression, leading to a diminished quality of life. This review aims to discuss the relationship between systemic and local inflammatory responses in the central nervous system (CNS), focusing on glial dysfunctions (astrocytes and microglia) as in critical brain regions involved in emotion, cognition, pain processing, anxiety, and depression. The review presents that EM is connected to increased levels of pro-inflammatory cytokines in the circulation. Additionally, chronic stress and CPP as stressors may contribute to the dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, depleting the production of inflammatory mediators in the circulatory system and the brain. The systemic cytokines cause blood-brain barrier (BBB) breakdown, activate microglia in the brain, and lead to neuroinflammation. Furthermore, CPP may induce neuronal morphological alterations in critical regions through central sensitization and the activation of glial cells. The activation of glial cells, particularly the polarization of microglia, leads to the activation of the NLRP3 inflammasome and the overproduction of inflammatory cytokines. These inflammatory cytokines interact with the signaling pathways involved in neural plasticity. Additionally, persistent inflammatory conditions in the brain lead to neuronal death, which is correlated with a reduced volume of key brain regions such as the hippocampus. This review highlights the involvement of glial cells in the pathogenesis of the mental comorbidities of EM (i.e., pain, anxiety, and depression) and to discuss potential therapeutic approaches for targeting the inflammation and activation of microglia in key brain regions.


Asunto(s)
Ansiedad , Depresión , Endometriosis , Neuroglía , Humanos , Femenino , Endometriosis/inmunología , Endometriosis/patología , Depresión/inmunología , Depresión/etiología , Depresión/metabolismo , Ansiedad/inmunología , Animales , Neuroglía/inmunología , Inflamación/inmunología , Estrés Psicológico/inmunología , Citocinas/metabolismo , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/metabolismo
6.
Curr Hypertens Rep ; 26(7): 339-347, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38613621

RESUMEN

PURPOSE OF REVIEW: Cardiovascular disease (CVD) is a leading cause of death and chronic disability worldwide. Yet, despite extensive intervention strategies the number of persons affected by CVD continues to rise. Thus, there is great interest in unveiling novel mechanisms that may lead to new treatments. Considering this dilemma, recent focus has turned to the neuroimmune mechanisms involved in CVD pathology leading to a deeper understanding of the brain's involvement in disease pathology. This review provides an overview of new and salient findings regarding the neuroimmune mechanisms that contribute to CVD. RECENT FINDINGS: The brain contains neuroimmune niches comprised of glia in the parenchyma and immune cells at the brain's borders, and there is strong evidence that these neuroimmune niches are important in both health and disease. Mechanistic studies suggest that the activation of glia and immune cells in these niches modulates CVD progression in hypertension and heart failure and contributes to the inevitable end-organ damage to the brain. This review provides evidence supporting the role of neuroimmune niches in CVD progression. However, additional research is needed to understand the effects of prolonged neuroimmune activation on brain function.


Asunto(s)
Encéfalo , Enfermedades Cardiovasculares , Neuroinmunomodulación , Humanos , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/fisiopatología , Neuroinmunomodulación/fisiología , Encéfalo/inmunología , Encéfalo/fisiopatología , Encéfalo/patología , Neuroglía/inmunología , Animales
8.
Cell Mol Gastroenterol Hepatol ; 18(1): 89-104, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38556049

RESUMEN

BACKGROUND & AIMS: Mounting evidence suggests the gastrointestinal microbiome is a determinant of peripheral immunity and central neurodegeneration, but the local disease mechanisms remain unknown. Given its potential relevance for early diagnosis and therapeutic intervention, we set out to map the pathogenic changes induced by bacterial amyloids in the gastrointestinal tract and its enteric nervous system. METHODS: To examine the early response, we challenged primary murine myenteric networks with curli, the prototypical bacterial amyloid, and performed shotgun RNA sequencing and multiplex enzyme-linked immunosorbent assay. Using enteric neurosphere-derived glial and neuronal cell cultures, as well as in vivo curli injections into the colon wall, we further scrutinized curli-induced pathogenic pathways. RESULTS: Curli induced a proinflammatory response, with strong up-regulation of Saa3 and the secretion of several cytokines. This proinflammatory state was induced primarily in enteric glia, was accompanied by increased levels of DNA damage and replication, and triggered the influx of immune cells in vivo. The addition of recombinant Serum Amyloid A3 (SAA3) was sufficient to recapitulate this specific proinflammatory phenotype while Saa3 knock-out attenuated curli-induced DNA damage and replication. Similar to curli, recombinant SAA3 caused a strong up-regulation of Saa3 transcripts, illustrating its self-amplifying potential . Since colonization of curli-producing Salmonella and dextran sulfate sodium-induced colitis triggered a significant increase in Saa3 transcripts as well, we assume SAA3plays a central role in enteric dysfunction. Inhibition of dual leucine zipper kinase, an upstream regulator of the c-Jun N-terminal kinase pathway responsible for SAA3 production, attenuated curli- and recombinant SAA3-induced Saa3 up-regulation, DNA damage, and replication in enteric glia. CONCLUSIONS: Our results position SAA3 as an important mediator of gastrointestinal vulnerability to bacterial-derived amyloids and demonstrate the potential of dual leucine zipper kinase inhibition to dampen enteric pathology.


Asunto(s)
Sistema Nervioso Entérico , Proteína Amiloide A Sérica , Animales , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/patología , Sistema Nervioso Entérico/inmunología , Proteína Amiloide A Sérica/metabolismo , Proteína Amiloide A Sérica/genética , Ratones , Proteínas Bacterianas/metabolismo , Inflamación/inmunología , Inflamación/patología , Inflamación/metabolismo , Neuroglía/metabolismo , Neuroglía/inmunología , Neuroglía/patología , Ratones Endogámicos C57BL , Citocinas/metabolismo , Microbioma Gastrointestinal/inmunología , Ratones Noqueados , Colitis/inmunología , Colitis/microbiología , Colitis/patología , Neuronas/metabolismo , Neuronas/patología
9.
Cell Mol Gastroenterol Hepatol ; 18(1): 133-153, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38428588

RESUMEN

BACKGROUND & AIMS: The presence of myenteric plexitis in the proximal resection margins is a predictive factor of early postoperative recurrence in Crohn's disease. To decipher the mechanisms leading to their formation, T-cell interactions with enteric neural cells were studied in vitro and in vivo. METHODS: T cells close to myenteric neural cells were retrospectively quantified in ileocolonic resections from 9 control subjects with cancer and 20 patients with Crohn's disease. The mechanisms involved in T-cell adhesion were then investigated in co-cultures of T lymphocytes with enteric glial cells (glia). Finally, the implication of adhesion molecules in the development of plexitis and colitis was studied in vitro but also in vivo in Winnie mice. RESULTS: The mean number of T cells close to glia, but not neurons, was significantly higher in the myenteric ganglia of relapsing patients with Crohn's disease (2.42 ± 0.5) as compared with controls (0.36 ± 0.08, P = .0007). Co-culture experiments showed that exposure to proinflammatory cytokines enhanced T-cell adhesion to glia and increased intercellular adhesion molecule-1 (ICAM-1) expression in glia. We next demonstrated that T-cell adhesion to glia was inhibited by an anti-ICAM-1 antibody. Finally, using the Winnie mouse model of colitis, we showed that the blockage of ICAM-1/lymphocyte function-associated antigen-1 (LFA-1) with lifitegrast reduced colitis severity and decreased T-cell infiltration in the myenteric plexus. CONCLUSIONS: Our present work argues for a role of glia-T-cell interaction in the development of myenteric plexitis through the adhesion molecules ICAM-1/LFA-1 and suggests that deciphering the functional consequences of glia-T-cell interaction is important to understand the mechanisms implicated in the development and recurrence of Crohn's disease.


Asunto(s)
Adhesión Celular , Técnicas de Cocultivo , Enfermedad de Crohn , Molécula 1 de Adhesión Intercelular , Plexo Mientérico , Neuroglía , Linfocitos T , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Enfermedad de Crohn/patología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Plexo Mientérico/patología , Plexo Mientérico/metabolismo , Plexo Mientérico/inmunología , Neuroglía/metabolismo , Neuroglía/patología , Neuroglía/inmunología , Estudios Retrospectivos , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Vis Neurosci ; 39: E005, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-36164752

RESUMEN

To study the macroglia and microglia and the immune role in long-time light exposure in rat eyes, we performed glial cell characterization along the time-course of retinal degeneration induced by chronic exposure to low-intensity light. Animals were exposed to light for periods of 2, 4, 6, or 8 days, and the retinal glial response was evaluated by immunohistochemistry, western blot and real-time reverse transcription polymerase chain reaction. Retinal cells presented an increased expression of the macroglia marker GFAP, as well as increased mRNA levels of microglia markers Iba1 and CD68 after 6 days. Also, at this time-point, we found a higher number of Iba1-positive cells in the outer nuclear layer area; moreover, these cells showed the characteristic activated-microglia morphology. The expression levels of immune mediators TNF, IL-6, and chemokines CX3CR1 and CCL2 were also significantly increased after 6 days. All the events of glial activation occurred after 5-6 days of constant light exposure, when the number of photoreceptor cells has already decreased significantly. Herein, we demonstrated that glial and immune activation are secondary to neurodegeneration; in this scenario, our results suggest that photoreceptor death is an early event that occurs independently of glial-derived immune responses.


Asunto(s)
Interleucina-6 , Neuroglía , Traumatismos por Radiación , Retina , Degeneración Retiniana , Animales , Quimiocinas/genética , Quimiocinas/metabolismo , Interleucina-6/metabolismo , Luz , Neuroglía/inmunología , ARN Mensajero/genética , Traumatismos por Radiación/etiología , Traumatismos por Radiación/inmunología , Ratas , Retina/inmunología , Retina/efectos de la radiación , Degeneración Retiniana/etiología , Degeneración Retiniana/inmunología
11.
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
12.
Sci Rep ; 11(1): 23569, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34876649

RESUMEN

Microvascular compression of the trigeminal root entry zone (TREZ) is the main cause of most primary trigeminal neuralgia (TN), change of glial plasticity was previously studied in the TREZ of TN rat model induced by chronic compression. To better understand the role of astrocytes and immune cells in the TREZ, different cell markers including glial fibrillary acidic protein (GFAP), complement C3, S100A10, CD45, CD11b, glutamate-aspartate transporter (GLAST), Iba-1 and TMEM119 were used in the TN rat model by immunohistochemistry and flow cytometry. On the post operation day 28, GFAP/C3-positive A1 astrocytes and GFAP/S100A10-positive A2 astrocytes were activated in the TREZ after compression injury, there were no statistical differences in the ratios of A1/A2 astrocytes between the sham and TN groups. There was no significant difference in Iba-1-positive cells between the two groups. The ratios of infiltrating lymphocytes (CD45+CD11b-) (p = 0.0075) and infiltrating macrophages (CD45highCD11b+) (p = 0.0388) were significantly higher than those of the sham group. In conclusion, different subtypes A1/A2 astrocytes in the TREZ were activated after compression injury, infiltrating macrophages and lymphocytes increased, these neuroimmune cells in the TREZ may participate in the pathogenesis of TN rat model.


Asunto(s)
Neuralgia del Trigémino/inmunología , Neuralgia del Trigémino/patología , Animales , Anexina A2/metabolismo , Astrocitos/inmunología , Astrocitos/metabolismo , Astrocitos/patología , Complemento C3/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Proteína Ácida Fibrilar de la Glía/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Linfocitos/patología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Neuroglía/inmunología , Neuroglía/metabolismo , Neuroglía/patología , Ratas , Ratas Sprague-Dawley , Proteínas S100/metabolismo , Nervio Trigémino/inmunología , Nervio Trigémino/metabolismo , Nervio Trigémino/patología
13.
Int J Mol Sci ; 22(24)2021 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-34948035

RESUMEN

Epilepsy is one of the most common neurological conditions. Yearly, five million people are diagnosed with epileptic-related disorders. The neuroprotective and therapeutic effect of (endo)cannabinoid compounds has been extensively investigated in several models of epilepsy. Therefore, the study of specific cell-type-dependent mechanisms underlying cannabinoid effects is crucial to understanding epileptic disorders. It is estimated that about 100 billion neurons and a roughly equal number of glial cells co-exist in the human brain. The glial population is in charge of neuronal viability, and therefore, their participation in brain pathophysiology is crucial. Furthermore, glial malfunctioning occurs in a wide range of neurological disorders. However, little is known about the impact of the endocannabinoid system (ECS) regulation over glial cells, even less in pathological conditions such as epilepsy. In this review, we aim to compile the existing knowledge on the role of the ECS in different cell types, with a particular emphasis on glial cells and their impact on epilepsy. Thus, we propose that glial cells could be a novel target for cannabinoid agents for treating the etiology of epilepsy and managing seizure-like disorders.


Asunto(s)
Endocannabinoides/metabolismo , Epilepsia/metabolismo , Neuroglía/metabolismo , Animales , Modelos Animales de Enfermedad , Epilepsia/tratamiento farmacológico , Epilepsia/inmunología , Regulación de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Neuroglía/inmunología
14.
Viruses ; 13(12)2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34960633

RESUMEN

The environment of the central nervous system (CNS) represents a double-edged sword in the context of viral infections. On the one hand, the infectious route for viral pathogens is restricted via neuroprotective barriers; on the other hand, viruses benefit from the immunologically quiescent neural environment after CNS entry. Both the herpes simplex virus (HSV) and the rabies virus (RABV) bypass the neuroprotective blood-brain barrier (BBB) and successfully enter the CNS parenchyma via nerve endings. Despite the differences in the molecular nature of both viruses, each virus uses retrograde transport along peripheral nerves to reach the human CNS. Once inside the CNS parenchyma, HSV infection results in severe acute inflammation, necrosis, and hemorrhaging, while RABV preserves the intact neuronal network by inhibiting apoptosis and limiting inflammation. During RABV neuroinvasion, surveilling glial cells fail to generate a sufficient type I interferon (IFN) response, enabling RABV to replicate undetected, ultimately leading to its fatal outcome. To date, we do not fully understand the molecular mechanisms underlying the activation or suppression of the host inflammatory responses of surveilling glial cells, which present important pathways shaping viral pathogenesis and clinical outcome in viral encephalitis. Here, we compare the innate immune responses of glial cells in RABV- and HSV-infected CNS, highlighting different viral strategies of neuroprotection or Neuroinflamm. in the context of viral encephalitis.


Asunto(s)
Encefalitis Viral/inmunología , Herpes Simple/inmunología , Inmunidad Innata , Inflamación , Virus de la Rabia/inmunología , Rabia/inmunología , Simplexvirus/inmunología , Animales , Astrocitos/inmunología , Astrocitos/virología , Barrera Hematoencefálica/virología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Encefalitis Viral/virología , Herpes Simple/virología , Humanos , Microglía/inmunología , Microglía/virología , Neuroglía/inmunología , Neuroglía/virología , Rabia/virología , Transducción de Señal
15.
Front Immunol ; 12: 783725, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804074

RESUMEN

Interferons (IFNs) are cytokines that possess antiviral, antiproliferative, and immunomodulatory actions. IFN-α and IFN-ß are two major family members of type-I IFNs and are used to treat diseases, including hepatitis and multiple sclerosis. Emerging evidence suggests that type-I IFN receptors (IFNARs) are also expressed by microglia, astrocytes, and neurons in the central and peripheral nervous systems. Apart from canonical transcriptional regulations, IFN-α and IFN-ß can rapidly suppress neuronal activity and synaptic transmission via non-genomic regulation, leading to potent analgesia. IFN-γ is the only member of the type-II IFN family and induces central sensitization and microglia activation in persistent pain. We discuss how type-I and type-II IFNs regulate pain and infection via neuro-immune modulations, with special focus on neuroinflammation and neuro-glial interactions. We also highlight distinct roles of type-I IFNs in the peripheral and central nervous system. Insights into IFN signaling in nociceptors and their distinct actions in physiological vs. pathological and acute vs. chronic conditions will improve our treatments of pain after surgeries, traumas, and infections.


Asunto(s)
Dolor Agudo/inmunología , Dolor Crónico/inmunología , Interferón Tipo I/metabolismo , Interferón gamma/metabolismo , Enfermedades Neuroinflamatorias/inmunología , Dolor Agudo/patología , Animales , Dolor Crónico/patología , Modelos Animales de Enfermedad , Humanos , Neuroglía/citología , Neuroglía/inmunología , Neuroglía/patología , Enfermedades Neuroinflamatorias/patología , Nociceptores/inmunología , Nociceptores/metabolismo , Receptores de Interferón/metabolismo , Transducción de Señal/inmunología , Médula Espinal/citología , Médula Espinal/inmunología , Médula Espinal/patología
16.
Nature ; 599(7883): 125-130, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34671159

RESUMEN

Tissue maintenance and repair depend on the integrated activity of multiple cell types1. Whereas the contributions of epithelial2,3, immune4,5 and stromal cells6,7 in intestinal tissue integrity are well understood, the role of intrinsic neuroglia networks remains largely unknown. Here we uncover important roles of enteric glial cells (EGCs) in intestinal homeostasis, immunity and tissue repair. We demonstrate that infection of mice with Heligmosomoides polygyrus leads to enteric gliosis and the upregulation of an interferon gamma (IFNγ) gene signature. IFNγ-dependent gene modules were also induced in EGCs from patients with inflammatory bowel disease8. Single-cell transcriptomics analysis of the tunica muscularis showed that glia-specific abrogation of IFNγ signalling leads to tissue-wide activation of pro-inflammatory transcriptional programs. Furthermore, disruption of the IFNγ-EGC signalling axis enhanced the inflammatory and granulomatous response of the tunica muscularis to helminths. Mechanistically, we show that the upregulation of Cxcl10 is an early immediate response of EGCs to IFNγ signalling and provide evidence that this chemokine and the downstream amplification of IFNγ signalling in the tunica muscularis are required for a measured inflammatory response to helminths and resolution of the granulomatous pathology. Our study demonstrates that IFNγ signalling in enteric glia is central to intestinal homeostasis and reveals critical roles of the IFNγ-EGC-CXCL10 axis in immune response and tissue repair after infectious challenge.


Asunto(s)
Homeostasis , Intestinos/inmunología , Intestinos/fisiología , Neuroglía/inmunología , Neuroglía/fisiología , Regeneración , Adventicia/inmunología , Adventicia/parasitología , Animales , Quimiocina CXCL10/inmunología , Duodeno/inmunología , Duodeno/parasitología , Duodeno/patología , Duodeno/fisiología , Femenino , Gliosis , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Interferón gamma/inmunología , Intestinos/parasitología , Intestinos/patología , Masculino , Ratones , Nematospiroides dubius/inmunología , Nematospiroides dubius/patogenicidad , Transducción de Señal/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología
17.
Cells ; 10(9)2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34571930

RESUMEN

Localisation of mast cells (MCs) at the abluminal side of blood vessels in the brain favours their interaction with glial cells, neurons, and endothelial cells, resulting in the activation of these cells and the release of pro-inflammatory mediators. In turn, stimulation of glial cells, such as microglia, astrocytes, and oligodendrocytes may result in the modulation of MC activities. MCs, microglia, astrocytes, and oligodendrocytes all express P2X receptors (P2XRs) family members that are selectively engaged by ATP. As increased concentrations of extracellular adenosine 5'-triphosphate (ATP) are present in the brain in neuropathological conditions, P2XR activation in MCs and glial cells contributes to the control of their communication and amplification of the inflammatory response. In this review we discuss P2XR-mediated MC activation, its bi-directional effect on microglia, astrocytes and oligodendrocytes and role in neuroinflammation.


Asunto(s)
Inflamación/patología , Mastocitos/inmunología , Neuroglía/inmunología , Neuronas/inmunología , Receptores Purinérgicos P2X/metabolismo , Animales , Humanos , Inflamación/inmunología , Inflamación/metabolismo
18.
J Neuropathol Exp Neurol ; 80(10): 933-943, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34498065

RESUMEN

Resected lesions from the pineal region are rare specimens encountered by surgical pathologists, and their heterogeneity can pose significant diagnostic challenges. Here, we reviewed 221 pineal region lesions resected at New York-Presbyterian Hospital/Columbia University Irving Medical Center from 1994 to 2019 and found the most common entities to be pineal parenchymal tumors (25.3%), glial neoplasms (18.6%), and germ cell tumors (17.6%) in this predominantly adult cohort of patients. Six cases of a rare midline entity usually found exclusively in the fourth ventricle, the rosette-forming glioneuronal tumor, were identified. These tumors exhibit biphasic morphology, with a component resembling pilocytic astrocytoma admixed with variable numbers of small cells forming compact rosettes and perivascular pseudorosettes. Targeted sequencing revealed a 100% co-occurrence of novel and previously described genetic alterations in the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling pathways, suggesting a synergistic role in tumor formation. The most common recurrent mutation, PIK3CA H1047R, was identified in tumor cells forming rosettes and perivascular pseudorosettes. A review of the literature revealed 16 additional cases of rosette-forming glioneuronal tumors in the pineal region. Although rare, this distinctive low-grade tumor warrants consideration in the differential diagnosis of pineal region lesions.


Asunto(s)
Neoplasias Encefálicas/patología , Neuroglía/patología , Glándula Pineal/patología , Pinealoma/patología , Formación de Roseta , Adolescente , Adulto , Neoplasias Encefálicas/inmunología , Niño , Femenino , Humanos , Masculino , Neuroglía/inmunología , Glándula Pineal/inmunología , Pinealoma/inmunología , Estudios Retrospectivos , Adulto Joven
19.
Brain Res Bull ; 175: 196-204, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34339780

RESUMEN

Reactive gliosis is a key feature and an important pathophysiological mechanism underlying chronic neurodegeneration following traumatic brain injury (TBI). In this study, we have explored the effects of intramuscular IGF-1 gene therapy on reactive gliosis and functional outcome after an injury of the cerebral cortex. Young adult male rats were intramuscularly injected with a recombinant adenoviral construct harboring the cDNA of human IGF-1 (RAd-IGF1), with a control vector expressing green fluorescent protein (RAd-GFP) or PBS as control. Three weeks after the intramuscular injections of adenoviral vectors, animals were subjected to a unilateral penetrating brain injury. The data revealed that RAd-IGF1 gene therapy significantly increased serum IGF1 levels and improved working memory performance after one week of TBI as compared to PBS or RAd-GFP lesioned animals. At the same time, when we analyzed the effects of therapy on glial scar formation, the treatment with RAd-IGF1 did not modify the number of glial fibrillary acidic protein (GFAP) positive cells, but we observed a decrease in vimentin immunoreactive astrocytes at 7 days post-lesion in the injured hemisphere compared to RAd-GFP group. Moreover, IGF-1 gene therapy reduced the number of Iba1+ cells with reactive phenotype and the number of MHCII + cells in the injured hemisphere. These results suggest that intramuscular IGF-1 gene therapy may represent a new approach to prevent traumatic brain injury outcomes in rats.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Terapia Genética/métodos , Gliosis/genética , Gliosis/terapia , Factor I del Crecimiento Similar a la Insulina/genética , Microglía , Animales , Lesiones Traumáticas del Encéfalo/psicología , Proteínas de Unión al Calcio/metabolismo , Vectores Genéticos/administración & dosificación , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inyecciones Intramusculares , Masculino , Memoria a Corto Plazo , Proteínas de Microfilamentos/metabolismo , Neuroglía/inmunología , Neuroprotección , Desempeño Psicomotor , Ratas , Resultado del Tratamiento , Vimentina/metabolismo
20.
Front Immunol ; 12: 639008, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34394070

RESUMEN

Background: Previous reports identified proteins associated with 'apoptosis' following cross-linking PrPC with motif-specific anti-PrP antibodies in vivo and in vitro. The molecular mechanisms underlying this IgG-mediated neurotoxicity and the role of the activated proteins in the apoptotic pathways leading to neuronal death has not been properly defined. Previous reports implicated a number of proteins, including apolipoprotein E, cytoplasmic phospholipase A2, prostaglandin and calpain with anti-PrP antibody-mediated 'apoptosis', however, these proteins are also known to play an important role in allergy. In this study, we investigated whether cross-linking PrPC with anti-PrP antibodies stimulates a neuronal allergenic response. Methods: Initially, we predicted the allergenicity of the epitope sequences associated with 'neurotoxic' anti-PrP antibodies using allergenicity prediction servers. We then investigated whether anti-PrP antibody treatment of mouse primary neurons (MPN), neuroblastoma cells (N2a) and microglia (N11) cell lines lead to a neuronal allergenic response. Results: In-Silico studies showed that both tail- and globular-epitopes were allergenic. Specifically, binding regions that contain epitopes for previously reported 'neurotoxic' antibodies such as ICSM18 (146-159), ICSM35 (91-110), POM 1 (138-147) and POM 3 (95-100) lead to activation of allergenic related proteins. Following direct application of anti-PrPC antibodies on N2a cells, we identified 4 neuronal allergenic-related proteins when compared with untreated cells. Furthermore, we identified 8 neuronal allergenic-related proteins following treatment of N11 cells with anti-PrPC antibodies prior to co-culture with N2a cells when compared with untreated cells. Antibody treatment of MPN or MPN co-cultured with antibody-treated N11 led to identifying 10 and 7 allergenic-related proteins when compared with untreated cells. However, comparison with 3F4 antibody treatment revealed 5 and 4 allergenic-related proteins respectively. Of importance, we showed that the allergenic effects triggered by the anti-PrP antibodies were more potent when antibody-treated microglia were co-cultured with the neuroblastoma cell line. Finally, co-culture of N2a or MPN with N11-treated with anti-PrP antibodies resulted in significant accumulation of NO and IL6 but not TNF-α in the cell culture media supernatant. Conclusions: This study showed for the first time that anti-PrP antibody binding to PrPC triggers a neuronal hypersensitivity response and highlights the important role of microglia in triggering an IgG-mediated neuronal hypersensitivity response. Moreover, this study provides an important impetus for including allergenic assessment of therapeutic antibodies for neurodegenerative disorders to derive safe and targeted biotherapeutics.


Asunto(s)
Anticuerpos/inmunología , Hipersensibilidad/inmunología , Neuronas/inmunología , Proteínas PrPC/inmunología , Proteínas PrPC/metabolismo , Animales , Epítopos de Linfocito B/inmunología , Humanos , Ratones , Neuroglía/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA