Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.730
Filtrar
1.
Mult Scler Relat Disord ; 59: 103682, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35158189

RESUMEN

OBJECTIVES: To determine anti-SARS-Cov2 antibodies and T-cell immunity in convalescent people with multiple sclerosis (pwMS) and/or pwMS vaccinated against Covid-19, depending on the disease modifying therapy, and in comparison to healthy controls (HC). METHODS: 75 participants were enrolled: Group 1-29 (38.7%) COVID-19 convalescent participants; Group 2-34 (45.3%) COVID-19 vaccinated; Group 3-12 (16.0%) COVID-19 convalescent participants who were later vaccinated against COVID-19. Cellular immunity was evaluated by determination of number of CD4+ and CD8+ cells secreting TNFα, IFNγ, and IL2 after stimulation with SARS-CoV-2 peptides. RESULTS: pwMS treated with ocrelizumab were less likely to develop humoral immunity after COVID-19 recovery or vaccination. No difference was observed in the cellular immunity in all studied parameters between pwMS treated with ocrelizumab compared to HC or pwMS who were treatment naïve or on first line therapies. These findings were consistent in convalescent, vaccinated, and convalescent+vaccinated participants. COVID-19 vaccinated convalescent pwMS on ocrelizumab compared to COVID-19 convalescent HC who were vaccinated did not show statistically difference in the rate of seroconversion nor titers of SARS-CoV-2 antibodies. CONCLUSION: Presence of cellular immunity in pwMS on B-cell depleting therapies is reassuring, as at least partial protection from more severe COVID-19 outcomes can be expected.


Asunto(s)
COVID-19 , Esclerosis Múltiple , Anticuerpos Antivirales , COVID-19/prevención & control , Humanos , Inmunidad Celular/fisiología , Inmunidad Humoral/inmunología , Esclerosis Múltiple/tratamiento farmacológico , SARS-CoV-2/química , SARS-CoV-2/inmunología
2.
Acta Neuropathol Commun ; 10(1): 14, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35105380

RESUMEN

Coronavirus disease 2019 (COVID-19) is associated with an increased risk of thrombotic events. Ischemic stroke in COVID-19 patients entails high severity and mortality rates. Here we aimed to analyze cerebral thrombi of COVID-19 patients with large vessel occlusion (LVO) acute ischemic stroke to expose molecular evidence for SARS-CoV-2 in the thrombus and to unravel any peculiar immune-thrombotic features. We conducted a systematic pathological analysis of cerebral thrombi retrieved by endovascular thrombectomy in patients with LVO stroke infected with COVID-19 (n = 7 patients) and non-covid LVO controls (n = 23). In thrombi of COVID-19 patients, the SARS-CoV-2 docking receptor ACE2 was mainly expressed in monocytes/macrophages and showed higher expression levels compared to controls. Using polymerase chain reaction and sequencing, we detected SARS-CoV-2 Clade20A, in the thrombus of one COVID-19 patient. Comparing thrombus composition of COVID-19 and control patients, we noted no overt differences in terms of red blood cells, fibrin, neutrophil extracellular traps (NETs), von Willebrand Factor (vWF), platelets and complement complex C5b-9. However, thrombi of COVID-19 patients showed increased neutrophil density (MPO+ cells) and a three-fold higher Neutrophil-to-Lymphocyte Ratio (tNLR). In the ROC analysis both neutrophils and tNLR had a good discriminative ability to differentiate thrombi of COVID-19 patients from controls. In summary, cerebral thrombi of COVID-19 patients can harbor SARS-CoV2 and are characterized by an increased neutrophil number and tNLR and higher ACE2 expression. These findings suggest neutrophils as the possible culprit in COVID-19-related thrombosis.


Asunto(s)
Isquemia Encefálica/inmunología , COVID-19/inmunología , Inmunidad Celular/fisiología , Trombosis Intracraneal/inmunología , Neutrófilos/inmunología , Accidente Cerebrovascular/inmunología , Anciano , Anciano de 80 o más Años , Enzima Convertidora de Angiotensina 2/sangre , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/inmunología , Isquemia Encefálica/sangre , Isquemia Encefálica/genética , COVID-19/sangre , COVID-19/genética , Femenino , Humanos , Trombosis Intracraneal/sangre , Trombosis Intracraneal/genética , Masculino , Trombolisis Mecánica/métodos , Persona de Mediana Edad , Neutrófilos/metabolismo , Estudios Prospectivos , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/metabolismo , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/genética
3.
Biomed Pharmacother ; 146: 112249, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34972632

RESUMEN

The emergence of many new viruses in recent times has resulted in a significant scientific challenge for discovering drugs and vaccines that effectively treat and prevent viral diseases. Nanotechnology has opened doors to prevent the spread of several diseases, including those caused by viruses. Polymer-hybrid nanodevices are a class of nanotechnology platforms for biomedical applications that present synergistic properties among their components, with improved performance compared to conventional forms of therapy. Considering the growing interest in this emerging field and the promising technological advantages of polymer-hybrid nanodevices, this work presents the current status of these systems in the context of prevention and treatment of viral diseases. A brief description of the different types of polymer-hybrid nanodevices highlighting some peculiar characteristics such as their composition, biodistribution, delivery of antigens, and overall immune responses in systemic tissues are discussed. Finally, the work presents the future trends for new nanotechnological hybrid materials based on polymers and perspectives for clinical use.


Asunto(s)
Antivirales/administración & dosificación , Nanopartículas/administración & dosificación , Nanotecnología/tendencias , Polímeros/administración & dosificación , Virosis/prevención & control , Animales , Antivirales/metabolismo , Sistemas de Liberación de Medicamentos , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/fisiología , Nanopartículas/metabolismo , Polímeros/metabolismo , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Virosis/metabolismo
4.
ACS Appl Mater Interfaces ; 14(5): 6331-6342, 2022 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-35084819

RESUMEN

The intranasal (i.n.) route is an ideal vaccination approach for infectious respiratory diseases like influenza. Polycationic polyethylenimine (PEI) could form nanoscale complexes with negatively charged viral glycoproteins. Here we fabricated PEI-hemagglutinin (HA) and PEI-HA/CpG nanoparticles and investigated their immune responses and protective efficacies with an i.n. vaccination regimen in mice. Our results revealed that the nanoparticles significantly enhanced HA immunogenicity, providing heterologous cross-protection. The conserved HA stalk region induced substantial antibodies in the nanoparticle immunization groups. In contrast to the Th2-biased, IgG1-dominant antibody response generated by PEI-HA nanoparticles, PEI-HA/CpG nanoparticles generated more robust and balanced IgG1/IgG2a antibody responses with augmented neutralization activity and Fc-mediated antibody-dependent cellular cytotoxicity (ADCC). PEI-HA/CpG nanoparticles also induced enhanced local and systemic cellular immune responses. These immune responses did not decay over six months of observation postimmunization. PEI and CpG synergized these comprehensive immune responses. Thus, the PEI-HA/CpG nanoparticle is a potential cross-protective influenza vaccine candidate. Polycationic PEI nanoplatforms merit future development into mucosal vaccine systems.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Inmunidad Celular/fisiología , Inmunidad Humoral/fisiología , Nanopartículas/química , Polietileneimina/química , Adyuvantes Inmunológicos/química , Administración Intranasal , Animales , Formación de Anticuerpos , Islas de CpG , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Inmunoglobulina G/sangre , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Células Th2/inmunología , Células Th2/metabolismo , Vacunación
5.
Anesthesiology ; 136(2): 293-313, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34965287

RESUMEN

BACKGROUND: Mechanical ventilation for pneumonia may contribute to lung injury due to factors that include mitochondrial dysfunction, and mesenchymal stem cells may attenuate injury. This study hypothesized that mechanical ventilation induces immune and mitochondrial dysfunction, with or without pneumococcal pneumonia, that could be mitigated by mesenchymal stem cells alone or combined with antibiotics. METHODS: Male rabbits underwent protective mechanical ventilation (8 ml/kg tidal volume, 5 cm H2O end-expiratory pressure) or adverse mechanical ventilation (20 ml/kg tidal-volume, zero end-expiratory pressure) or were allowed to breathe spontaneously. The same settings were then repeated during pneumococcal pneumonia. Finally, infected animals during adverse mechanical ventilation received human umbilical cord-derived mesenchymal stem cells (3 × 106/kg, intravenous) and/or ceftaroline (20 mg/kg, intramuscular) or sodium chloride, 4 h after pneumococcal challenge. Twenty-four-hour survival (primary outcome), lung injury, bacterial burden, immune and mitochondrial dysfunction, and lung transcriptomes (secondary outcomes) were assessed. RESULTS: High-pressure adverse mechanical ventilation reduced the survival of infected animals (0%; 0 of 7) compared with spontaneous breathing (100%; 7 of 7) and protective mechanical ventilation (86%; 6 of 7; both P < 0.001), with higher lung pathology scores (median [interquartile ranges], 5.5 [4.5 to 7.0] vs. 12.6 [12.0 to 14.0]; P = 0.046), interleukin-8 lung concentrations (106 [54 to 316] vs. 804 [753 to 868] pg/g of lung; P = 0.012), and alveolar mitochondrial DNA release (0.33 [0.28 to 0.36] vs. 0.98 [0.76 to 1.21] ng/µl; P < 0.001) compared with infected spontaneously breathing animals. Survival (0%; 0 of 7; control group) was improved by mesenchymal stem cells (57%; 4 of 7; P = 0.001) or ceftaroline alone (57%; 4 of 7; P < 0.001) and improved even more with a combination treatment (86%; 6 of 7; P < 0.001). Mesenchymal stem cells reduced lung pathology score (8.5 [7.0 to 10.5] vs. 12.6 [12.0 to 14.0]; P = 0.043) and alveolar mitochondrial DNA release (0.39 (0.34 to 0.65) vs. 0.98 (0.76 to 1.21) ng/µl; P = 0.025). Mesenchymal stem cells combined with ceftaroline reduced interleukin-8 lung concentrations (665 [595 to 795] vs. 804 [753 to 868] pg/g of lung; P = 0.007) compared to ceftaroline alone. CONCLUSIONS: In this preclinical study, mesenchymal stem cells improved the outcome of rabbits with pneumonia and high-pressure mechanical ventilation by correcting immune and mitochondrial dysfunction and when combined with the antibiotic ceftaroline was synergistic in mitigating lung inflammation.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Inmunidad Celular/fisiología , Mitocondrias/inmunología , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/terapia , Respiración Artificial/efectos adversos , Animales , Masculino , Células Madre Mesenquimatosas/fisiología , Mitocondrias/metabolismo , Neumonía Neumocócica/metabolismo , Estudios Prospectivos , Conejos , Distribución Aleatoria
6.
J Gerontol A Biol Sci Med Sci ; 77(1): 33-40, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34252180

RESUMEN

Understanding how older people respond to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is critical if we are to confront the coronavirus disease 2019 (COVID-19) pandemic and establish effective vaccination strategies. Immunosenescence reduces the ability to respond to neoantigens and may compromise the life of infected individuals. Here, we analyzed the immunological memory to SARS-CoV-2 in 102 recovered patients aged over 60 years several months after the infection had been resolved. Specific memory T lymphocytes against the virus were measured by interferon-γ (IFN-γ) and granzyme B release by ELISpot; memory B-lymphocyte responses were quantified by detection of anti-S IgG1 producer cells by ELISpot and anti-S and anti-N antibodies were determined by enzyme-linked immunosorbent assay (ELISA). Memory T lymphocytes were found in peripheral blood of most of the studied donors, more than 7 months after the infection in some of them. Fewer patients maintained memory B lymphocytes, but antibodies, mainly anti-S, were highly durable and positively correlated with T responses. More robust humoral responses were found in patients who had more severe symptoms and had been admitted to hospital. We concluded that specific immunity against SARS-CoV-2 is effectively preserved regardless of age, despite the great heterogeneity of their immune responses, and that memory T lymphocytes and anti-S IgG might be more durable than memory B cells and anti-N IgG.


Asunto(s)
Anticuerpos Antivirales/inmunología , COVID-19 , Inmunidad Celular/fisiología , Inmunidad Humoral , Memoria Inmunológica , SARS-CoV-2 , Anciano , Anciano de 80 o más Años , Ensayo de Immunospot Ligado a Enzimas , Femenino , Humanos , Inmunoglobulina G , Masculino , Células B de Memoria , Células T de Memoria , Persona de Mediana Edad
7.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34880136

RESUMEN

Identification of type 1 innate lymphoid cells (ILC1s) has been problematic. The transcription factor Hobit encoded by Zfp683 has been proposed as a major driver of ILC1 programs. Using Zfp683 reporter mice, we showed that correlation of Hobit expression with ILC1s is tissue- and context-dependent. In liver and intestinal mucosa, Zfp683 expression correlated well with ILC1s; in salivary glands, Zfp683 was coexpressed with the natural killer (NK) master transcription factors Eomes and TCF1 in a unique cell population, which we call ILC1-like NK cells; during viral infection, Zfp683 was induced in conventional NK cells of spleen and liver. The impact of Zfp683 deletion on ILC1s and NK cells was also multifaceted, including a marked decrease in granzyme- and interferon-gamma (IFNγ)-producing ILC1s in the liver, slightly fewer ILC1s and more Eomes+ TCF1+ ILC1-like NK cells in salivary glands, and only reduced production of granzyme B by ILC1 in the intestinal mucosa. NK cell-mediated control of viral infection was unaffected. We conclude that Hobit has two major impacts on ILC1s: It sustains liver ILC1 numbers, while promoting ILC1 functional maturation in other tissues by controlling TCF1, Eomes, and granzyme expression.


Asunto(s)
Inmunidad Celular/fisiología , Inmunidad Innata/fisiología , Subgrupos Linfocitarios/clasificación , Subgrupos Linfocitarios/fisiología , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Animales , Antígenos CD , Biomarcadores , Eliminación de Gen , Regulación de la Expresión Génica/fisiología , Granzimas/genética , Granzimas/metabolismo , Interferón gamma/genética , Interferón gamma/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/genética , Ratones , ARN Citoplasmático Pequeño/genética , ARN Citoplasmático Pequeño/metabolismo , RNA-Seq , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética
8.
Life Sci ; 287: 120117, 2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34740577

RESUMEN

Denervated skeletal muscular atrophy is primarily characterized by loss of muscle strength and mass and an unideal functional recovery of the muscle after extended denervation. This review emphasizes the interaction between the immune system and the denervated skeletal muscle. Immune cells such as neutrophils, macrophages and T-cells are activated and migrate to denervated muscle, where they release a high concentration of cytokines and chemokines. The migration of these immune cells, the transformation of different functional immune cell subtypes, and the cytokine network in the immune microenvironment may be involved in the regulatory process of muscle atrophy or repair. However, the exact mechanisms of the interaction between these immune cells and immune molecules in skeletal muscles are unclear. In this paper, the immune microenvironment regulation of muscle atrophy induced by peripheral nerve injury is reviewed.


Asunto(s)
Investigación Biomédica/tendencias , Microambiente Celular/fisiología , Inmunidad Celular/fisiología , Músculo Esquelético/inmunología , Atrofia Muscular/inmunología , Traumatismos de los Nervios Periféricos/inmunología , Animales , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Desnervación Muscular/métodos , Músculo Esquelético/inervación , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo
9.
Front Immunol ; 12: 745332, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671359

RESUMEN

The induction of trained immunity represents an emerging concept defined as the ability of innate immune cells to acquire a memory phenotype, which is a typical hallmark of the adaptive response. Key points modulated during the establishment of trained immunity include epigenetic, metabolic and functional changes in different innate-immune and non-immune cells. Regarding to epigenetic changes, it has been described that long non-coding RNAs (LncRNAs) act as molecular scaffolds to allow the assembly of chromatin-remodeling complexes that catalyze epigenetic changes on chromatin. On the other hand, relevant metabolic changes that occur during this process include increased glycolytic rate and the accumulation of metabolites from the tricarboxylic acid (TCA) cycle, which subsequently regulate the activity of histone-modifying enzymes that ultimately drive epigenetic changes. Functional consequences of established trained immunity include enhanced cytokine production, increased antigen presentation and augmented antimicrobial responses. In this article, we will discuss the current knowledge regarding the ability of different cell subsets to acquire a trained immune phenotype and the molecular mechanisms involved in triggering such a response. This knowledge will be helpful for the development of broad-spectrum therapies against infectious diseases based on the modulation of epigenetic and metabolic cues regulating the development of trained immunity.


Asunto(s)
Interacciones Huésped-Patógeno/inmunología , Inmunidad Celular , Inmunidad Innata/inmunología , Memoria Inmunológica/inmunología , Inmunidad Adaptativa/genética , Inmunidad Adaptativa/inmunología , Inmunidad Adaptativa/fisiología , Animales , Vacuna BCG/inmunología , Bronquios/citología , Bronquios/inmunología , Citocinas/fisiología , Metabolismo Energético , Epigénesis Genética , Células Epiteliales/inmunología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/inmunología , Células Madre Hematopoyéticas/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Humanos , Inmunidad Celular/genética , Inmunidad Celular/fisiología , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Memoria Inmunológica/genética , Memoria Inmunológica/fisiología , Linfocitos/inmunología , Ratones , Células Mieloides/inmunología , NAD/fisiología , Piel/citología , Piel/inmunología
11.
Biomed Pharmacother ; 144: 112230, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34628168

RESUMEN

The COVID-19 pandemic caused by the coronavirus SARS-CoV-2 has become a serious challenge for medicine and science. Analysis of the molecular mechanisms associated with the clinical manifestations and severity of COVID-19 has identified several key points of immune dysregulation observed in SARS-CoV-2 infection. For diabetic patients, factors including higher binding affinity and virus penetration, decreased virus clearance and decreased T cell function, increased susceptibility to hyperinflammation, and cytokine storm may make these patients susceptible to a more severe course of COVID-19 disease. Metabolic changes induced by diabetes, especially hyperglycemia, can directly affect the immunometabolism of lymphocytes in part by affecting the activity of the mTOR protein kinase signaling pathway. High mTOR activity can enhance the progression of diabetes due to the activation of effector proinflammatory subpopulations of lymphocytes and, conversely, low activity promotes the differentiation of T-regulatory cells. Interestingly, metformin, an extensively used antidiabetic drug, inhibits mTOR by affecting the activity of AMPK. Therefore, activation of AMPK and/or inhibition of the mTOR-mediated signaling pathway may be an important new target for drug therapy in COVID-19 cases mostly by reducing the level of pro-inflammatory signaling and cytokine storm. These suggestions have been partially confirmed by several retrospective analyzes of patients with diabetes mellitus hospitalized for severe COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Diabetes Mellitus/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Inmunidad Celular/efectos de los fármacos , Metformina/uso terapéutico , Índice de Severidad de la Enfermedad , COVID-19/epidemiología , COVID-19/inmunología , COVID-19/metabolismo , Diabetes Mellitus/epidemiología , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Humanos , Hipoglucemiantes/farmacología , Inmunidad Celular/fisiología , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/metabolismo , Metformina/farmacología , Mortalidad/tendencias , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/inmunología , Serina-Treonina Quinasas TOR/metabolismo
12.
Acta Neuropathol Commun ; 9(1): 148, 2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34496929

RESUMEN

It is recognized that the tumor microenvironment (TME) plays a critical role in the biology of cancer. To better understand the role of immune cell components in CNS tumors, we applied a deconvolution approach to bulk DNA methylation array data in a large set of newly profiled samples (n = 741) as well as samples from external data sources (n = 3311) of methylation-defined glial and glioneuronal tumors. Using the cell-type proportion data as input, we used dimensionality reduction to visualize sample-wise patterns that emerge from the cell type proportion estimations. In IDH-wildtype glioblastomas (n = 2,072), we identified distinct tumor clusters based on immune cell proportion and demonstrated an association with oncogenic alterations such as EGFR amplification and CDKN2A/B homozygous deletion. We also investigated the immune cluster-specific distribution of four malignant cellular states (AC-like, OPC-like, MES-like and NPC-like) in the IDH-wildtype cohort. We identified two major immune-based subgroups of IDH-mutant gliomas, which largely aligned with 1p/19q co-deletion status. Non-codeleted gliomas showed distinct proportions of a key genomic aberration (CDKN2A/B loss) among immune cell-based groups. We also observed significant positive correlations between monocyte proportion and expression of PD-L1 and PD-L2 (R = 0.54 and 0.68, respectively). Overall, the findings highlight specific roles of the TME in biology and classification of CNS tumors, where specific immune cell admixtures correlate with tumor types and genomic alterations.


Asunto(s)
Biomarcadores de Tumor/inmunología , Neoplasias Encefálicas/inmunología , Metilación de ADN/fisiología , Glioma/inmunología , Inmunidad Celular/fisiología , Microambiente Tumoral/fisiología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Estudios de Cohortes , Variaciones en el Número de Copia de ADN/fisiología , Análisis de Datos , Glioma/genética , Glioma/metabolismo , Humanos , Monocitos/inmunología , Monocitos/metabolismo
13.
Nat Rev Neurosci ; 22(11): 657-673, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34545240

RESUMEN

Almost 60 years have passed since the initial discovery by Hubel and Wiesel that changes in neuronal activity can elicit developmental rewiring of the central nervous system (CNS). Over this period, we have gained a more comprehensive picture of how both spontaneous neural activity and sensory experience-induced changes in neuronal activity guide CNS circuit development. Here we review activity-dependent synaptic pruning in the mammalian CNS, which we define as the removal of a subset of synapses, while others are maintained, in response to changes in neural activity in the developing nervous system. We discuss the mounting evidence that immune and cell-death molecules are important mechanistic links by which changes in neural activity guide the pruning of specific synapses, emphasizing the role of glial cells in this process. Finally, we discuss how these developmental pruning programmes may go awry in neurodevelopmental disorders of the human CNS, focusing on autism spectrum disorder and schizophrenia. Together, our aim is to give an overview of how the field of activity-dependent pruning research has evolved, led to exciting new questions and guided the identification of new, therapeutically relevant mechanisms that result in aberrant circuit development in neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Sistema Nervioso Central/fisiología , Inmunidad Celular/fisiología , Plasticidad Neuronal/fisiología , Esquizofrenia/fisiopatología , Factores de Edad , Animales , Trastorno del Espectro Autista/inmunología , Sistema Nervioso Central/citología , Humanos , Trastornos del Neurodesarrollo/inmunología , Trastornos del Neurodesarrollo/fisiopatología , Esquizofrenia/inmunología
14.
mBio ; 12(5): e0159921, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34488453

RESUMEN

Cellular immunity may be involved in organ damage and rehabilitation in patients with coronavirus disease 2019 (COVID-19). We aimed to delineate immunological features of COVID-19 patients with pulmonary sequelae (PS) 1 year after discharge. Fifty COVID-19 survivors were recruited and classified according to radiological characteristics, including 24 patients with PS and 26 patients without PS. Phenotypic and functional characteristics of immune cells were evaluated by multiparametric flow cytometry. Patients with PS had an increased proportion of natural killer (NK) cells and a lower percentage of B cells than patients without PS. Phenotypic and functional features of T cells in patients with PS were predominated by the accumulation of CD4-positive (CD4+) T cells secreting interleukin 17A (IL-17A), short-lived effector-like CD8+ T cells (CD27-negative [CD27-] CD62L-), and senescent T cells with excessive secretion of granzyme B/perforin/interferon gamma (IFN-γ). NK cells were characterized by the excessive secretion of granzyme B and perforin and the downregulation of NKP30 and NKP46; highly activated NKT and γδ T cells exhibited NKP30 and TIM-3 upregulation and NKB1 downregulation in patients with PS. However, immunosuppressive cells were comparable between the two groups. The interrelationship of immune cells in COVID-19 was intrinsically identified, whereby T cells secreting IL-2, IL-4, and IL-17A were enriched among CD28+ and CD57- cells and cells secreting perforin/granzyme B/IFN-γ/tumor necrosis factor alpha (TNF-α)-expressed markers of terminal differentiation. CD57+ NK cells, CD4+Perforin+ T cells, and CD8+ CD27+ CD62L+ T cells were identified as the independent predictors for residual lesions. Overall, our findings unveil the profound imbalance of immune landscape that may correlate with organ damage and rehabilitation in COVID-19. IMPORTANCE A considerable proportion of COVID-19 survivors have residual lung lesions such as ground-glass opacity and fiber streak shadow. To determine the relationship between host immunity and residual lung lesions, we performed an extensive analysis of immune responses in convalescent patients with COVID-19 1 year after discharge. We found significant differences in immunological characteristics between patients with pulmonary sequelae and patients without pulmonary sequelae 1 year after discharge. Our study highlights the profound imbalance of immune landscape in the COVID-19 patients with pulmonary sequelae, characterized by the robust activation of cytotoxic T cells, NK cells, and γδ T cells, as well as the deficiencies of immunosuppressive cells. Importantly, CD57+ NK cells, CD4+Perforin+ T cells, and CD8+ CD27+ CD62L+ T cells were identified as the independent predictors for residual lesions.


Asunto(s)
COVID-19/inmunología , Adulto , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD57/metabolismo , Linfocitos T CD8-positivos/metabolismo , COVID-19/metabolismo , Femenino , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Inmunidad Celular/inmunología , Inmunidad Celular/fisiología , Interleucina-17/metabolismo , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Selectina L/metabolismo , Masculino , Persona de Mediana Edad , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Receptor 3 Gatillante de la Citotoxidad Natural/metabolismo
15.
J Neuroimmunol ; 359: 577694, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34450375

RESUMEN

Neuroinflammation plays an important role in the pathogenesis of several neurodegenerative disorders. To elucidate the effects of the mitophagy-related gene Parkin on neuroinflammation, we used a mouse model of experimental autoimmune encephalomyelitis (EAE). Female Parkin-/- and female wild type control mice were immunized with myelin oligodendrocyte glycoprotein to develop active EAE. Compared to the wild type controls, the Parkin-/- mice showed an earlier onset and greater severity of EAE with a greatly increased number of CD8αß+TCRαß+ T cells in the spleen and brain as well as a stronger T-cell proliferative response and an altered cytokine secretion in splenocytes. Furthermore, the Parkin-/- mice showed massive recruitment of monocytes/macrophages and activated microglia in the spinal cord during the acute phase of the disease. They also showed accumulation of microglia co-expressing M1 and M2 markers in the brain and a strong over-expression of A1 reactive astrocytes in the spinal cord. Furthermore, the Parkin-/- mice that developed persistent disease exhibited reduced glial cell numbers and abnormalities in mitochondrial morphology. Our study sheds light on the role of PARKIN protein in modulating peripheral immune cells-mediated immunity during EAE, highlighting its importance in neuroinflammation, and thus elucidating its potential in the development of novel neuroprotective therapies.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/metabolismo , Inmunidad Celular/fisiología , Mediadores de Inflamación/metabolismo , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Secuencia de Aminoácidos , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
16.
Cells ; 10(6)2021 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205262

RESUMEN

COVID-19 is an acute infectious disease of the respiratory system caused by infection with the SARS-CoV-2 virus (Severe Acute Respiratory Syndrome Coronavirus 2). Transmission of SARS-CoV-2 infections occurs through droplets and contaminated objects. A rapid and well-coordinated immune system response is the first line of defense in a viral infection. However, a disturbed and over-activated immune response may be counterproductive, causing damage to the body. Severely ill patients hospitalised with COVID-19 exhibit increased levels of many cytokines, including Interleukin (IL)-1ß, IL-2, IL-6, IL-7, IL-8, IL-10, IL-17, granulocyte colony stimulating factor (G-CSF), monocyte chemoattractant protein 1 (MCP-1) and tumor necrosis factor (TNF). Increasing evidence suggests that Th17 cells play an important role in the pathogenesis of COVID-19, not only by activating cytokine cascade but also by inducing Th2 responses, inhibiting Th1 differentiation and suppressing Treg cells. This review focuses on a Th17 pathway in the course of the immune response in COVID-19, and explores plausible targets for therapeutic intervention.


Asunto(s)
COVID-19/inmunología , Inmunidad Celular/fisiología , Células Th17/fisiología , COVID-19/patología , COVID-19/terapia , Citocinas/metabolismo , Humanos , Inmunoterapia Adoptiva/métodos , SARS-CoV-2/inmunología , Células Th17/metabolismo
17.
Cells ; 10(6)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203839

RESUMEN

The bone marrow (BM) is key to protective immunological memory because it harbors a major fraction of the body's plasma cells, memory CD4+ and memory CD8+ T-cells. Despite its paramount significance for the human immune system, many aspects of how the BM enables decade-long immunity against pathogens are still poorly understood. In this review, we discuss the relationship between BM survival niches and long-lasting humoral immunity, how intrinsic and extrinsic factors define memory cell longevity and show that the BM is also capable of adopting many responsibilities of a secondary lymphoid organ. Additionally, with more and more data on the differentiation and maintenance of memory T-cells and plasma cells upon vaccination in humans being reported, we discuss what factors determine the establishment of long-lasting immunological memory in the BM and what we can learn for vaccination technologies and antigen design. Finally, using these insights, we touch on how this holistic understanding of the BM is necessary for the development of modern and efficient vaccines against the pandemic SARS-CoV-2.


Asunto(s)
Inmunidad Adaptativa/fisiología , Médula Ósea/fisiología , Células Plasmáticas/citología , Linfocitos T/citología , Vacunología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , COVID-19/inmunología , COVID-19/prevención & control , Vacunas contra la COVID-19/inmunología , Humanos , Inmunidad Celular/fisiología , Memoria Inmunológica/fisiología , SARS-CoV-2/inmunología , Linfocitos T/inmunología , Vacunología/métodos , Vacunología/tendencias
19.
Med Oncol ; 38(7): 85, 2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34148185

RESUMEN

Hepatocellular carcinoma (HCC) is among the primary causes of cancer deaths globally. Despite efforts to understand liver cancer, its high morbidity and mortality remain high. Herein, we constructed two nomograms based on competing endogenous RNA (ceRNA) networks and invading immune cells to describe the molecular mechanisms along with the clinical prognosis of HCC patients. RNA maps of tumors and normal samples were downloaded from The Cancer Genome Atlas database. HTseq counts and fragments per megapons per thousand bases were read from 421 samples, including 371 tumor samples and 50 normal samples. We established a ceRNA network based on differential gene expression in normal versus tumor subjects. CIBERSORT was employed to differentiate 22 immune cell types according to tumor transcriptomes. Kaplan-Meier along with Cox proportional hazard analyses were employed to determine the prognosis-linked factors. Nomograms were constructed based on prognostic immune cells and ceRNAs. We employed Receiver operating characteristic (ROC) and calibration curve analyses to estimate these nomogram. The difference analysis found 2028 messenger RNAs (mRNAs), 128 micro RNAs (miRNAs), and 136 long non-coding RNAs (lncRNAs) to be significantly differentially expressed in tumor samples relative to normal samples. We set up a ceRNA network containing 21 protein-coding mRNAs, 12 miRNAs, and 3 lncRNAs. In Kaplan-Meier analysis, 21 of the 36 ceRNAs were considered significant. Of the 22 cell types, resting dendritic cell levels were markedly different in tumor samples versus normal controls. Calibration and ROC curve analysis of the ceRNA network, as well as immune infiltration of tumor showed restful accuracy (3-year survival area under curve (AUC): 0.691, 5-year survival AUC: 0.700; 3-year survival AUC: 0.674, 5-year survival AUC: 0.694). Our data suggest that Tregs, CD4 T cells, mast cells, SNHG1, HMMR and hsa-miR-421 are associated with HCC based on ceRNA immune cells co-expression patterns. On the basis of ceRNA network modeling and immune cell infiltration analysis, our study offers an effective bioinformatics strategy for studying HCC molecular mechanisms and prognosis.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/inmunología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , ARN/genética , ARN/inmunología , Carcinoma Hepatocelular/mortalidad , Regulación Neoplásica de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Humanos , Inmunidad Celular/fisiología , Neoplasias Hepáticas/mortalidad , MicroARNs/genética , MicroARNs/inmunología , Nomogramas , ARN Largo no Codificante/genética , ARN Largo no Codificante/inmunología , ARN Mensajero/genética , ARN Mensajero/inmunología , Análisis de Supervivencia
20.
Circ Res ; 128(7): 908-933, 2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33793336

RESUMEN

Elevated cardiovascular risk including stroke, heart failure, and heart attack is present even after normalization of blood pressure in patients with hypertension. Underlying immune cell activation is a likely culprit. Although immune cells are important for protection against invading pathogens, their chronic overactivation may lead to tissue damage and high blood pressure. Triggers that may initiate immune activation include viral infections, autoimmunity, and lifestyle factors such as excess dietary salt. These conditions activate the immune system either directly or through their impact on the gut microbiome, which ultimately produces chronic inflammation and hypertension. T cells are central to the immune responses contributing to hypertension. They are activated in part by binding specific antigens that are presented in major histocompatibility complex molecules on professional antigen-presenting cells, and they generate repertoires of rearranged T-cell receptors. Activated T cells infiltrate tissues and produce cytokines including interleukin 17A, which promote renal and vascular dysfunction and end-organ damage leading to hypertension. In this comprehensive review, we highlight environmental, genetic, and microbial associated mechanisms contributing to both innate and adaptive immune cell activation leading to hypertension. Targeting the underlying chronic immune cell activation in hypertension has the potential to mitigate the excess cardiovascular risk associated with this common and deadly disease.


Asunto(s)
Hipertensión/inmunología , Inmunidad Celular/fisiología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Antihipertensivos/uso terapéutico , Linfocitos B/inmunología , Proteínas del Sistema Complemento/inmunología , Citocinas/inmunología , Células Dendríticas/inmunología , Resistencia a Medicamentos , Femenino , Microbioma Gastrointestinal/inmunología , Factores de Riesgo de Enfermedad Cardiaca , Interacciones Microbiota-Huesped , Humanos , Hipertensión/tratamiento farmacológico , Fenómenos del Sistema Inmunológico , Inmunidad Innata , Inflamasomas/inmunología , Inflamación/genética , Inflamación/inmunología , Macrófagos/inmunología , Masculino , Monocitos/inmunología , Factores Sexuales , Cloruro de Sodio Dietético/efectos adversos , Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Virosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA