Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 995
Filtrar
1.
Int Immunopharmacol ; 132: 111982, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38569430

RESUMEN

RTS,S is the first malaria vaccine recommended for implementation among young children at risk. However, vaccine efficacy is modest and short-lived. To mitigate the risk of cerebral malaria (CM) among children under the age of 5, it is imperative to develop new vaccines. EVs are potential vaccine candidates as they obtain the ability of brain-targeted delivery and transfer plasmodium antigens and immunomodulators during infections. This study extracted EVs from BALB/c mice infected with Plasmodium yoelii 17XNL (P.y17XNL). C57BL/6J mice were intravenously immunized with EVs (EV-I.V. + CM group) or subcutaneously vaccinated with the combination of EVs and CpG ODN-1826 (EV + CPG ODN-S.C. + CM group) on days 0 and 20, followed by infection with Plasmodium berghei ANKA (P.bANKA) on day 20 post-second immunization. We monitored Parasitemia and survival rate. The integrity of the Blood-brain barrier (BBB) was examined using Evans blue staining.The levels of cytokines and adhesion molecules were evaluated using Luminex, RT-qPCR, and WB. Brain pathology was evaluated by hematoxylin and eosin and immunohistochemical staining. The serum levels of IgG, IgG1, and IgG2a were analyzed by enzyme-linked immunosorbent assay. Compared with those in the P.bANKA-infected group, parasitemia increased slowly, death was delayed (day 10 post-infection), and the survival rate reached 75 %-83.3 % in the EV-I.V. + ECM and EV + CPG ODN-S.C. + ECM groups. Meanwhile, compared with the EV + CPG ODN-S.C. + ECM group, although parasitemia was almost the same, the survival rate increased in the EV-I.V. + ECM group.Additionally, EVs immunization markedly downregulated inflammatory responses in the spleen and brain and ameliorated brain pathological changes, including BBB disruption and infected red blood cell (iRBC) sequestration. Furthermore, the EVs immunization group exhibited enhanced antibody responses (upregulation of IgG1 and IgG2a production) compared to the normal control group. EV immunization exerted protective effects, improving the integrity of the BBB, downregulating inflammation response of brain tissue, result in reduces the incidence of CM. The protective effects were determined by immunological pathways and brain targets elicited by EVs. Intravenous immunization exhibited better performance than subcutaneous immunization, which perhaps correlated with EVs, which can naturally cross BBB to play a better role in brain protection.


Asunto(s)
Barrera Hematoencefálica , Eritrocitos , Vesículas Extracelulares , Malaria Cerebral , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos , Plasmodium berghei , Animales , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Malaria Cerebral/prevención & control , Plasmodium berghei/inmunología , Vesículas Extracelulares/inmunología , Eritrocitos/parasitología , Eritrocitos/inmunología , Barrera Hematoencefálica/inmunología , Ratones , Oligodesoxirribonucleótidos/administración & dosificación , Vacunas contra la Malaria/inmunología , Vacunas contra la Malaria/administración & dosificación , Femenino , Encéfalo/parasitología , Encéfalo/inmunología , Encéfalo/patología , Citocinas/metabolismo , Citocinas/sangre , Plasmodium yoelii/inmunología , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Parasitemia/inmunología , Modelos Animales de Enfermedad , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología
2.
Pharmacol Res ; 203: 107168, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38583689

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative disease characterized by progressive loss of dopaminergic neurons in the substantia nigra and the aggregation of alpha-synuclein (α-syn). The central nervous system (CNS) has previously been considered as an immune-privileged area. However, studies have shown that the immune responses are involved in PD. The major histocompatibility complex (MHC) presents antigens from antigen-presenting cells (APCs) to T lymphocytes, immune responses will be induced. MHCs are expressed in microglia, astrocytes, and dopaminergic neurons. Single nucleotide polymorphisms in MHC are related to the risk of PD. The aggregated α-syn triggers the expression of MHCs by activating glia cells. CD4+ and CD8+ T lymphocytes responses and microglia activation are detected in brains of PD patients. In addiction immune responses further increase blood-brain barrier (BBB) permeability and T cell infiltration in PD. Thus, MHCs are involved in PD through participating in immune and inflammatory responses.


Asunto(s)
Complejo Mayor de Histocompatibilidad , Enfermedad de Parkinson , Humanos , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/genética , Animales , Complejo Mayor de Histocompatibilidad/inmunología , alfa-Sinucleína/inmunología , alfa-Sinucleína/metabolismo , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Microglía/inmunología , Microglía/metabolismo
3.
J Integr Neurosci ; 21(1): 9, 2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-35164445

RESUMEN

Epilepsy is one of the most common neurology diseases. It is characterized by recurrent, spontaneous seizures and accompanied by various comorbidities which can significantly affect a person's life. Accumulating evidence indicates an essential pathophysiological role for neuroinflammation in epilepsy, which involves activation of microglia and astrocytes, recruitment of peripheral leukocytes into the central nervous system, and release of some inflammatory mediators, including pro-inflammatory factors and anti-inflammatory cytokines. There is complex crosstalk between the central nervous system and peripheral immune responses associated with the progression of epilepsy. This review provides an update of current knowledge about the contribution of this crosstalk associated with epilepsy. Additionally, how gut microbiota is involved in epilepsy and its possible influence on crosstalk is also discussed. Such recent advances in understanding suggest innovative methods for targeting the molecules correlated with the crosstalk and may provide a better prognosis for patients diagnosed with epilepsy.


Asunto(s)
Barrera Hematoencefálica/inmunología , Epilepsia/inmunología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/inmunología , Enfermedades Neuroinflamatorias/inmunología , Animales , Humanos
4.
Eur J Immunol ; 52(1): 161-177, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34524684

RESUMEN

The migration of CD4+ effector/memory T cells across the blood-brain barrier (BBB) is a critical step in MS or its animal model, EAE. T-cell diapedesis across the BBB can occur paracellular, via the complex BBB tight junctions or transcellular via a pore through the brain endothelial cell body. Making use of primary mouse brain microvascular endothelial cells (pMBMECs) as in vitro model of the BBB, we here directly compared the transcriptome profile of pMBMECs favoring transcellular or paracellular T-cell diapedesis by RNA sequencing (RNA-seq). We identified the atypical chemokine receptor 1 (Ackr1) as one of the main candidate genes upregulated in pMBMECs favoring transcellular T-cell diapedesis. We confirmed upregulation of ACKR1 protein in pMBMECs promoting transcellular T-cell diapedesis and in venular endothelial cells in the CNS during EAE. Lack of endothelial ACKR1 reduced transcellular T-cell diapedesis across pMBMECs under physiological flow in vitro. Combining our previous observation that endothelial ACKR1 contributes to EAE pathogenesis by shuttling chemokines across the BBB, the present data support that ACKR1 mediated chemokine shuttling enhances transcellular T-cell diapedesis across the BBB during autoimmune neuroinflammation.


Asunto(s)
Barrera Hematoencefálica , Linfocitos T CD4-Positivos , Sistema del Grupo Sanguíneo Duffy , Encefalomielitis Autoinmune Experimental , Células T de Memoria , Esclerosis Múltiple , Receptores de Superficie Celular , Migración Transendotelial y Transepitelial , Animales , Ratones , Barrera Hematoencefálica/inmunología , Linfocitos T CD4-Positivos/inmunología , Sistema del Grupo Sanguíneo Duffy/genética , Sistema del Grupo Sanguíneo Duffy/inmunología , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/genética , Inflamación/inmunología , Células T de Memoria/inmunología , Ratones Noqueados , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Migración Transendotelial y Transepitelial/genética , Migración Transendotelial y Transepitelial/inmunología
5.
FASEB J ; 36(1): e22107, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34939700

RESUMEN

Mounting evidence has linked the metabolic disease to neurovascular disorders and cognitive decline. Using a murine model of a high-fat high-sugar diet mimicking obesity-induced type 2 diabetes mellitus (T2DM) in humans, we show that pro-inflammatory mediators and altered immune responses damage the blood-brain barrier (BBB) structure, triggering a proinflammatory metabolic phenotype. We find that disruption to tight junctions and basal lamina due to loss of control in the production of matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) causes BBB impairment. Together the disruption to the structural and functional integrity of the BBB results in enhanced transmigration of leukocytes across the BBB that could contribute to an initiation of a neuroinflammatory response through activation of microglia. Using a humanized in vitro model of the BBB and T2DM patient post-mortem brains, we show the translatable applicability of our results. We find a leaky BBB phenotype in T2DM patients can be attributed to a loss of junctional proteins through changes in inflammatory mediators and MMP/TIMP levels, resulting in increased leukocyte extravasation into the brain parenchyma. We further investigated therapeutic avenues to reduce and restore the BBB damage caused by HFHS-feeding. Pharmacological treatment with recombinant annexin A1 (hrANXA1) or reversion from a high-fat high-sugar diet to a control chow diet (dietary intervention), attenuated T2DM development, reduced inflammation, and restored BBB integrity in the animals. Given the rising incidence of diabetes worldwide, understanding metabolic-disease-associated brain microvessel damage is vital and the proposed therapeutic avenues could help alleviate the burden of these diseases.


Asunto(s)
Barrera Hematoencefálica/inmunología , Colagenasas/inmunología , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Tipo 2/inmunología , Inhibidores Tisulares de Metaloproteinasas/inmunología , Animales , Anexina A1/farmacología , Barrera Hematoencefálica/patología , Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Humanos , Masculino , Ratones , Proteínas Recombinantes/farmacología
6.
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
7.
Fluids Barriers CNS ; 18(1): 53, 2021 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-34863201

RESUMEN

BACKGROUND: The Gram-negative bacterium Neisseria meningitidis (Nm) can cause meningitis in humans, but the host signalling pathways manipulated by Nm during central nervous system (CNS) entry are not completely understood. METHODS: We investigate the role of the mitogen-activated protein kinases (MAPK) Erk1/2 and p38 in an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB) based on human epithelial choroid plexus (CP) papilloma (HIBCPP) cells during infection with Nm serogroup B (NmB) and serogroup C (NmC) strains. A transcriptome analysis of HIBCPP cells following infection with Nm by massive analysis of cDNA ends (MACE) was done to further characterize the cellular response to infection of the barrier. RESULTS: Interestingly, whereas NmB and NmC wild type strains required active Erk1/2 and p38 pathways for infection, invasion by capsule-deficient mutants was independent of Erk1/2 and, in case of the NmB strain, of p38 activity. The transcriptome analysis of HIBCPP cells following infection with Nm demonstrated specific regulation of genes involved in the immune response dependent on Erk1/2 signalling. Gene ontology (GO) analysis confirmed loss of MAPK signalling after Erk1/2 inhibition and revealed an additional reduction of cellular responses including NFκB and JAK-STAT signalling. Interestingly, GO terms related to TNF signalling and production of IL6 were lost specifically following Erk1/2 inhibition during infection with wild type Nm, which correlated with the reduced infection rates by the wild type in absence of Erk1/2 signalling. CONCLUSION: Our data point towards a role of MAPK signalling during infection of the CP epithelium by Nm, which is strongly influenced by capsule expression, and affects infection rates as well as the host cell response.


Asunto(s)
Barrera Hematoencefálica , Líquido Cefalorraquídeo , Plexo Coroideo , Células Epiteliales , Interacciones Huésped-Patógeno/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Neisseria meningitidis/patogenicidad , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/microbiología , Línea Celular Tumoral , Líquido Cefalorraquídeo/inmunología , Líquido Cefalorraquídeo/metabolismo , Líquido Cefalorraquídeo/microbiología , Plexo Coroideo/inmunología , Plexo Coroideo/metabolismo , Plexo Coroideo/microbiología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Humanos
8.
Front Immunol ; 12: 783882, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34868063

RESUMEN

It has been reported that several immune cells can release chromatin and granular proteins into extracellular space in response to the stimulation, forming extracellular traps (ETs). The cells involved in the extracellular trap formation are recognized including neutropils, macrophages, basophils, eosinophils, and mast cells. With the development of research related to central nervous system, the role of ETs has been valued in neuroinflammation, blood-brain barrier, and other fields. Meanwhile, it has been found that microglial cells as the resident immune cells of the central nervous system can also release ETs, updating the original understanding. This review aims to clarify the role of the ETs in the central nervous system, especially in neuroinflammation and blood-brain barrier.


Asunto(s)
Barrera Hematoencefálica/citología , Trampas Extracelulares/inmunología , Microglía/citología , Enfermedades Neuroinflamatorias/inmunología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Modelos Animales de Enfermedad , Trampas Extracelulares/metabolismo , Humanos , Microglía/inmunología , Microglía/metabolismo , Microglía/patología , Enfermedades Neuroinflamatorias/patología
9.
PLoS Negl Trop Dis ; 15(11): e0009892, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34762691

RESUMEN

Human African trypanosomiasis (HAT), also known as sleeping sickness, is a major cause of mortality and morbidity in sub-Saharan Africa. We hypothesised that recent findings of neurological features and parasite brain infiltration occurring at much earlier stages in HAT than previously thought could be explained by early activation of host genetic programmes controlling CNS disease. Accordingly, a transcriptomal analysis was performed on brain tissue at 0, 7, 14, 21 and 28dpi from the HAT CD1/GVR35 mouse model. Up to 21dpi, most parasites are restricted to the blood and lymphatic system. Thereafter the trypanosomes enter the brain initiating the encephalitic stage. Analysis of ten different time point Comparison pairings, revealed a dynamic transcriptome comprising four message populations. All 7dpi Comparisons had by far more differentially expressed genes compared to all others. Prior to invasion of the parenchyma, by 7dpi, ~2,000 genes were up-regulated, denoted [7dpi↑] in contrast to a down regulated population [7dpi↓] also numbering ~2,000. However, by 14dpi both patterns had returned to around the pre-infected levels. The third, [28dpi↑] featured over three hundred transcripts which had increased modestly up to14dpi, thereafter were significantly up-regulated and peaked at 28dpi. The fourth, a minor population, [7dpi↑-28dpi↑], had similar elevated levels at 7dpi and 28dpi. KEGG and GO enrichment analysis predicted a diverse phenotype by 7dpi with changes to innate and adaptive immunity, a Type I interferon response, neurotransmission, synaptic plasticity, pleiotropic signalling, circadian activity and vascular permeability without disruption of the blood brain barrier. This key observation is consistent with recent rodent model neuroinvasion studies and clinical reports of Stage 1 HAT patients exhibiting CNS symptoms. Together, these findings challenge the strict Stage1/Stage2 phenotypic demarcation in HAT and show that that significant neurological, and immune changes can be detected prior to the onset of CNS disease.


Asunto(s)
Encéfalo/parasitología , Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/inmunología , Trypanosoma brucei brucei/fisiología , Tripanosomiasis Africana/genética , Tripanosomiasis Africana/inmunología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/parasitología , Encéfalo/inmunología , Enfermedades del Sistema Nervioso Central/parasitología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Fenotipo , Análisis por Matrices de Proteínas , Transcripción Genética , Tripanosomiasis Africana/parasitología
10.
Cell Mol Immunol ; 18(11): 2489-2501, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34594000

RESUMEN

The vascular blood-brain barrier is a highly regulated interface between the blood and brain. Its primary function is to protect central neurons while signaling the presence of systemic inflammation and infection to the brain to enable a protective sickness behavior response. With increasing degrees and duration of systemic inflammation, the vascular blood-brain barrier becomes more permeable to solutes, undergoes an increase in lymphocyte trafficking, and is infiltrated by innate immune cells; endothelial cell damage may occasionally occur. Perturbation of neuronal function results in the clinical features of encephalopathy. Here, the molecular and cellular anatomy of the vascular blood-brain barrier is reviewed, first in a healthy context and second in a systemic inflammatory context. Distinct from the molecular and cellular mediators of the blood-brain barrier's response to inflammation, several moderators influence the direction and magnitude at genetic, system, cellular and molecular levels. These include sex, genetic background, age, pre-existing brain pathology, systemic comorbidity, and gut dysbiosis. Further progress is required to define and measure mediators and moderators of the blood-brain barrier's response to systemic inflammation in order to explain the heterogeneity observed in animal and human studies.


Asunto(s)
Barrera Hematoencefálica/inmunología , Encefalopatías/inmunología , Disbiosis/inmunología , Infecciones/inmunología , Inflamación/inmunología , Neuronas/inmunología , Animales , Interacción Gen-Ambiente , Humanos , Transducción de Señal
11.
J Gen Virol ; 102(10)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34704923

RESUMEN

The highly pathogenic Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a severe respiratory virus. Recent reports indicate additional central nervous system (CNS) involvement. In this study, human DPP4 transgenic mice were infected with MERS-CoV, and viral antigens were first detected in the midbrain-hindbrain 4 days post-infection, suggesting the virus may enter the brainstem via peripheral nerves. Neurons and astrocytes throughout the brain were infected, followed by damage of the blood brain barrier (BBB), as well as microglial activation and inflammatory cell infiltration, which may be caused by complement activation based on the observation of deposition of complement activation product C3 and high expression of C3a receptor (C3aR) and C5a receptor (C5aR1) in neurons and glial cells. It may be concluded that these effects were mediated by complement activation in the brain, because of their reduction resulted from the treatment with mouse C5aR1-specific mAb. Such mAb significantly reduced nucleoprotein expression, suppressed microglial activation and decreased activation of caspase-3 in neurons and p38 phosphorylation in the brain. Collectively, these results suggest that MERS-CoV infection of CNS triggers complement activation, leading to inflammation-mediated damage of brain tissue, and regulating of complement activation could be a promising intervention and adjunctive treatment for CNS injury by MERS-CoV and other coronaviruses.


Asunto(s)
Encéfalo/patología , Proteínas del Sistema Complemento/inmunología , Infecciones por Coronavirus/patología , Dipeptidil Peptidasa 4/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Encéfalo/virología , Activación de Complemento/efectos de los fármacos , Inactivadores del Complemento/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Humanos , Inflamación , Ratones , Ratones Transgénicos , Microglía/inmunología , Microglía/patología
12.
Immunol Res ; 69(6): 471-486, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34554405

RESUMEN

With recent advances, chimeric antigen receptor (CAR) immunotherapy has become a promising modality for patients with refractory cancer diseases. The successful results of CAR T cell therapy in relapsed and refractory B-cell malignancies shifted the paradigm of cancer immunotherapy by awakening the scientific, clinical, and commercial interest in translating this technology for the treatment of solid cancers. This review elaborates on fundamental principles of CAR T cell therapy (development of CAR construct, challenges of CAR T cell therapy) and its application on solid tumors as well as CAR T cell therapy potential in the field of neuro-oncology. Glioblastoma (GBM) is identified as one of the most challenging solid tumors with a permissive immunological milieu and dismal prognosis. Standard multimodal treatment using maximal safe resection, radiochemotherapy, and maintenance chemotherapy extends the overall survival beyond a year. Recurrence is, however, inevitable. GBM holds several unique features including its vast intratumoral heterogeneity, immunosuppressive environment, and a partially permissive anatomic blood-brain barrier, which offers a unique opportunity to investigate new treatment approaches. Tremendous efforts have been made in recent years to investigate novel CAR targets and target combinations with standard modalities for solid tumors and GBM to improve treatment efficacy. In this review, we outline the history of CAR immunotherapy development, relevant CAR target antigens validated with CAR T cells as well as preclinical approaches in combination with adjunct approaches via checkpoint inhibition, bispecific antibodies, and second-line systemic therapies that enhance anticancer efficacy of the CAR-based cancer immunotherapy.


Asunto(s)
Barrera Hematoencefálica/inmunología , Neoplasias Encefálicas , Glioblastoma , Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos/uso terapéutico , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Glioblastoma/inmunología , Glioblastoma/terapia , Humanos , Receptores Quiméricos de Antígenos/inmunología
13.
Cells ; 10(9)2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34572077

RESUMEN

Ischemic stroke is still among the leading causes of mortality and morbidity worldwide. Despite intensive advancements in medical sciences, the clinical options to treat ischemic stroke are limited to thrombectomy and thrombolysis using tissue plasminogen activator within a narrow time window after stroke. Current state of the art knowledge reveals the critical role of local and systemic inflammation after stroke that can be triggered by interactions taking place at the brain and immune system interface. Here, we discuss different cellular and molecular mechanisms through which brain-immune interactions can take place. Moreover, we discuss the evidence how the brain influence immune system through the release of brain derived antigens, damage-associated molecular patterns (DAMPs), cytokines, chemokines, upregulated adhesion molecules, through infiltration, activation and polarization of immune cells in the CNS. Furthermore, the emerging concept of stemness-induced cellular immunity in the context of neurodevelopment and brain disease, focusing on ischemic implications, is discussed. Finally, we discuss current evidence on brain-immune system interaction through the autonomic nervous system after ischemic stroke. All of these mechanisms represent potential pharmacological targets and promising future research directions for clinically relevant discoveries.


Asunto(s)
Isquemia Encefálica/inmunología , Encéfalo/inmunología , Neuroinmunomodulación , Accidente Cerebrovascular/inmunología , Alarminas/metabolismo , Barrera Hematoencefálica/inmunología , Lesiones Encefálicas/inmunología , Isquemia Encefálica/terapia , Quimiocinas/metabolismo , Citocinas/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Inflamación/inmunología , Sistema Nervioso/inmunología , Células Madre/inmunología , Accidente Cerebrovascular/terapia
14.
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
15.
Exp Neurol ; 346: 113865, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34547288

RESUMEN

Leukocyte infiltration and blood-brain barrier breakdown contribute to secondary brain damage after traumatic brain injury (TBI). TBI induces neuroimmune responses triggering pathogenic complement activation through different pathways, including the lectin pathway. We investigated mechanisms underlying mannose-binding lectin (MBL)-mediated brain damage focusing on neutrophil infiltration and blood-brain barrier breakdown in a TBI mouse model. Wild type mice and MBL-/- null mice were subjected to controlled cortical impact. We studied neutrophil infiltration and regional localization by confocal microscopy 1, 4 and 15 days post-trauma, and investigated neutrophil extracellular trap (NET) formation. By immunofluorescence and/or Western blotting in various brain regions we studied the presence of fibrin(ogen), pentraxin-3, albumin and immunoglobulin G. Finally, we studied neurofilament proteins, synaptophysin, and αII-spectrin, and assessed white matter content in the injured tissue. TBI triggered an acute wave of neutrophil infiltration at day 1 followed by a more discrete persistence of neutrophils in the injured tissue at least until day 15. We detected the presence of NETs and pentraxin-3 in the injured tissue, as well as accumulation of fibrin(ogen), increased blood-brain barrier permeability, and neurofilament, synaptophysin and white matter loss, and calpain-mediated αII spectrin breakdown. MBL-/- mice showed reduced number of Ly6G+ neutrophils 4 days after TBI, lower accumulation of pentraxin-3 and fibrin(ogen) in the injured tissue, reduced global plasma protein extravasation, and better preservation of axonal and white matter integrity. These results show that MBL participates in secondary neutrophil accumulation and blood-brain barrier breakdown, and promotes axonal and white matter damage after TBI in mice.


Asunto(s)
Axones/metabolismo , Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Encéfalo/metabolismo , Lectina de Unión a Manosa/deficiencia , Animales , Axones/inmunología , Axones/patología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Encéfalo/inmunología , Encéfalo/patología , Lesiones Traumáticas del Encéfalo/inmunología , Lesiones Traumáticas del Encéfalo/patología , Masculino , Lectina de Unión a Manosa/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
PLoS One ; 16(9): e0256972, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34473764

RESUMEN

High titers of anti-NMDAR1 autoantibodies in brain cause anti-NMDAR1 encephalitis that displays psychiatric symptoms of schizophrenia and/or other psychiatric disorders in addition to neurological symptoms. Low titers of anti-NMDAR1 autoantibodies are reported in the blood of a subset of the general human population and psychiatric patients. Since ~0.1-0.2% of blood circulating antibodies cross the blood-brain barriers and antibodies can persist for months and years in human blood, it is important to investigate whether chronic presence of these blood circulating anti-NMDAR1 autoantibodies may impair human cognitive functions and contribute to the development of psychiatric symptoms. Here, we generated mice carrying low titers of anti-NMDAR1 autoantibodies in blood against a single antigenic epitope of mouse NMDAR1. Mice carrying the anti-NMDAR1 autoantibodies are healthy and display no differences in locomotion, sensorimotor gating, and contextual memory compared to controls. Chronic presence of the blood circulating anti-NMDAR1 autoantibodies, however, is sufficient to impair T-maze spontaneous alternation in the integrity of blood-brain barriers across all 3 independent mouse cohorts, indicating a robust cognitive deficit in spatial working memory and/or novelty detection. Our studies implicate that chronic presence of low titers of blood circulating anti-NMDAR1 autoantibodies may impair cognitive functions in both the general healthy human population and psychiatric patients.


Asunto(s)
Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Cognición , Disfunción Cognitiva/sangre , Disfunción Cognitiva/inmunología , Proteínas del Tejido Nervioso/inmunología , Receptores de N-Metil-D-Aspartato/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Conducta Animal , Barrera Hematoencefálica/inmunología , Adyuvante de Freund/administración & dosificación , Locomoción/inmunología , Masculino , Memoria a Corto Plazo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Mycobacterium tuberculosis/inmunología , Proteínas del Tejido Nervioso/química , Péptidos/administración & dosificación , Péptidos/inmunología , Receptores de N-Metil-D-Aspartato/química , Memoria Espacial , Vacunación/métodos
17.
Biomolecules ; 11(8)2021 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-34439797

RESUMEN

Single-domain antibodies derive from the heavy-chain-only antibodies of Camelidae (camel, dromedary, llama, alpaca, vicuñas, and guananos; i.e., nanobodies) and cartilaginous fishes (i.e., VNARs). Their small size, antigen specificity, plasticity, and potential to recognize unique conformational epitopes represent a diagnostic and therapeutic opportunity for many central nervous system (CNS) pathologies. However, the blood-brain barrier (BBB) poses a challenge for their delivery into the brain parenchyma. Nevertheless, numerous neurological diseases and brain pathologies, including cancer, result in BBB leakiness favoring single-domain antibodies uptake into the CNS. Some single-domain antibodies have been reported to naturally cross the BBB. In addition, different strategies and methods to deliver both nanobodies and VNARs into the brain parenchyma can be exploited when the BBB is intact. These include device-based and physicochemical disruption of the BBB, receptor and adsorptive-mediated transcytosis, somatic gene transfer, and the use of carriers/shuttles such as cell-penetrating peptides, liposomes, extracellular vesicles, and nanoparticles. Approaches based on single-domain antibodies are reaching the clinic for other diseases. Several tailoring methods can be followed to favor the transport of nanobodies and VNARs to the CNS, avoiding the limitations imposed by the BBB to fulfill their therapeutic, diagnostic, and theragnostic promises for the benefit of patients suffering from CNS pathologies.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Portadores de Fármacos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Anticuerpos de Dominio Único/uso terapéutico , Transcitosis , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Encéfalo/patología , Camelidae , Péptidos de Penetración Celular/farmacocinética , Portadores de Fármacos/química , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Humanos , Liposomas/farmacocinética , Modelos Moleculares , Nanopartículas/administración & dosificación , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/inmunología , Enfermedades Neurodegenerativas/patología , Permeabilidad , Conformación Proteica , Anticuerpos de Dominio Único/metabolismo
18.
Brain ; 144(12): 3576-3588, 2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-34398188

RESUMEN

Coronavirus disease 2019 (COVID-19) is a global pandemic caused by SARS-CoV-2 infection and is associated with both acute and chronic disorders affecting the nervous system. Acute neurological disorders affecting patients with COVID-19 range widely from anosmia, stroke, encephalopathy/encephalitis, and seizures to Guillain-Barré syndrome. Chronic neurological sequelae are less well defined although exercise intolerance, dysautonomia, pain, as well as neurocognitive and psychiatric dysfunctions are commonly reported. Molecular analyses of CSF and neuropathological studies highlight both vascular and immunologic perturbations. Low levels of viral RNA have been detected in the brains of few acutely ill individuals. Potential pathogenic mechanisms in the acute phase include coagulopathies with associated cerebral hypoxic-ischaemic injury, blood-brain barrier abnormalities with endotheliopathy and possibly viral neuroinvasion accompanied by neuro-immune responses. Established diagnostic tools are limited by a lack of clearly defined COVID-19 specific neurological syndromes. Future interventions will require delineation of specific neurological syndromes, diagnostic algorithm development and uncovering the underlying disease mechanisms that will guide effective therapies.


Asunto(s)
Encéfalo/inmunología , COVID-19/epidemiología , COVID-19/inmunología , Enfermedades del Sistema Nervioso/epidemiología , Enfermedades del Sistema Nervioso/inmunología , Neuroinmunomodulación/fisiología , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/patología , Encéfalo/patología , COVID-19/complicaciones , COVID-19/diagnóstico por imagen , Humanos , Enfermedades del Sistema Nervioso/diagnóstico por imagen , Síndrome Post Agudo de COVID-19
19.
Nat Immunol ; 22(9): 1083-1092, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34429552

RESUMEN

For decades, it was commonly accepted that the brain is secluded from peripheral immune activity and is self-sufficient for its maintenance and repair. This simplistic perception was based on the presence of resident immune cells, the microglia, and barrier systems within the brain, and the assumption that the central nervous system (CNS) lacks lymphatic drainage. This view was revised with the discoveries that higher functions of the CNS, homeostasis and repair are supported by peripheral innate and adaptive immune cells. The findings of bone marrow-derived immune cells in specialized niches, and the renewed observation that a lymphatic drainage system exists within the brain, further contributed to this revised model. In this Review, we describe the immune niches within the brain, the contribution of professional immune cells to brain functions, the bidirectional relationships between the CNS and the immune system and the relevance of immune components to brain aging and neurodegenerative diseases.


Asunto(s)
Encéfalo/inmunología , Inmunidad/fisiología , Microglía/inmunología , Enfermedades Neurodegenerativas/inmunología , Envejecimiento/inmunología , Barrera Hematoencefálica/inmunología , Células de la Médula Ósea/inmunología , Líquido Cefalorraquídeo/citología , Líquido Cefalorraquídeo/inmunología , Humanos , Subgrupos Linfocitarios/inmunología , Macrófagos/inmunología
20.
Cells ; 10(8)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34440810

RESUMEN

Multiple sclerosis (MS), an immune-mediated demyelinating disease of the central nervous system (CNS), initially presents with a relapsing-remitting disease course. During this early stage of the disease, leukocytes cross the blood-brain barrier to drive the formation of focal demyelinating plaques. Disease-modifying agents that modulate or suppress the peripheral immune system provide a therapeutic benefit during relapsing-remitting MS (RRMS). The majority of individuals with RRMS ultimately enter a secondary progressive disease stage with a progressive accumulation of neurologic deficits. The cellular and molecular basis for this transition is unclear and the role of inflammation during the secondary progressive disease stage is a subject of intense and controversial debate. In this review article, we discuss the following main hypothesis: during both disease stages, peripheral immune cells are triggered by CNS-intrinsic stimuli to invade the brain parenchyma. Furthermore, we outline the different neuroanatomical routes by which peripheral immune cells might migrate from the periphery into the CNS.


Asunto(s)
Sistema Nervioso Central/inmunología , Leucocitos Mononucleares/inmunología , Animales , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/metabolismo , Comunicación Celular , Sistema Nervioso Central/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Humanos , Leucocitos Mononucleares/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...