Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 25(6): 1020-1032, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38831106

RESUMEN

The efficacy of T cell-based immunotherapies is limited by immunosuppressive pressures in the tumor microenvironment. Here we show a predominant role for the interaction between BTLA on effector T cells and HVEM (TNFRSF14) on immunosuppressive tumor microenvironment cells, namely regulatory T cells. High BTLA expression in chimeric antigen receptor (CAR) T cells correlated with poor clinical response to treatment. Therefore, we deleted BTLA in CAR T cells and show improved tumor control and persistence in models of lymphoma and solid malignancies. Mechanistically, BTLA inhibits CAR T cells via recruitment of tyrosine phosphatases SHP-1 and SHP-2, upon trans engagement with HVEM. BTLA knockout thus promotes CAR signaling and subsequently enhances effector function. Overall, these data indicate that the BTLA-HVEM axis is a crucial immune checkpoint in CAR T cell immunotherapy and warrants the use of strategies to overcome this barrier.


Asunto(s)
Inmunoterapia Adoptiva , Receptores Quiméricos de Antígenos , Receptores Inmunológicos , Miembro 14 de Receptores del Factor de Necrosis Tumoral , Microambiente Tumoral , Animales , Humanos , Inmunoterapia Adoptiva/métodos , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Ratones , Microambiente Tumoral/inmunología , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Linfocitos T Reguladores/inmunología , Transducción de Señal , Línea Celular Tumoral , Neoplasias/inmunología , Neoplasias/terapia , Ratones Noqueados
2.
Cell ; 184(7): 1858-1864.e10, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33631096

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread within the human population. Although SARS-CoV-2 is a novel coronavirus, most humans had been previously exposed to other antigenically distinct common seasonal human coronaviruses (hCoVs) before the coronavirus disease 2019 (COVID-19) pandemic. Here, we quantified levels of SARS-CoV-2-reactive antibodies and hCoV-reactive antibodies in serum samples collected from 431 humans before the COVID-19 pandemic. We then quantified pre-pandemic antibody levels in serum from a separate cohort of 251 individuals who became PCR-confirmed infected with SARS-CoV-2. Finally, we longitudinally measured hCoV and SARS-CoV-2 antibodies in the serum of hospitalized COVID-19 patients. Our studies indicate that most individuals possessed hCoV-reactive antibodies before the COVID-19 pandemic. We determined that ∼20% of these individuals possessed non-neutralizing antibodies that cross-reacted with SARS-CoV-2 spike and nucleocapsid proteins. These antibodies were not associated with protection against SARS-CoV-2 infections or hospitalizations, but they were boosted upon SARS-CoV-2 infection.


Asunto(s)
Alphacoronavirus/inmunología , Anticuerpos Antivirales , Betacoronavirus/inmunología , COVID-19/inmunología , Adolescente , Adulto , Animales , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Prueba Serológica para COVID-19 , Niño , Preescolar , Chlorocebus aethiops , Protección Cruzada , Reacciones Cruzadas , Susceptibilidad a Enfermedades , Células HEK293 , Humanos , Lactante , Recién Nacido , Células Vero
3.
Nat Immunol ; 24(2): 359-370, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36536105

RESUMEN

Understanding the complexity of the long-lived HIV reservoir during antiretroviral therapy (ART) remains a considerable impediment in research towards a cure for HIV. To address this, we developed a single-cell strategy to precisely define the unperturbed peripheral blood HIV-infected memory CD4+ T cell reservoir from ART-treated people living with HIV (ART-PLWH) via the presence of integrated accessible proviral DNA in concert with epigenetic and cell surface protein profiling. We identified profound reservoir heterogeneity within and between ART-PLWH, characterized by new and known surface markers within total and individual memory CD4+ T cell subsets. We further uncovered new epigenetic profiles and transcription factor motifs enriched in HIV-infected cells that suggest infected cells with accessible provirus, irrespective of reservoir distribution, are poised for reactivation during ART treatment. Together, our findings reveal the extensive inter- and intrapersonal cellular heterogeneity of the HIV reservoir, and establish an initial multiomic atlas to develop targeted reservoir elimination strategies.


Asunto(s)
Infecciones por VIH , VIH-1 , Humanos , VIH-1/fisiología , Linfocitos T CD4-Positivos , Latencia del Virus/genética , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/genética , Epigénesis Genética , Carga Viral , Antirretrovirales/uso terapéutico
4.
Nat Immunol ; 24(11): 1947-1959, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37845489

RESUMEN

Age-associated changes in the T cell compartment are well described. However, limitations of current single-modal or bimodal single-cell assays, including flow cytometry, RNA-seq (RNA sequencing) and CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing), have restricted our ability to deconvolve more complex cellular and molecular changes. Here, we profile >300,000 single T cells from healthy children (aged 11-13 years) and older adults (aged 55-65 years) by using the trimodal assay TEA-seq (single-cell analysis of mRNA transcripts, surface protein epitopes and chromatin accessibility), which revealed that molecular programming of T cell subsets shifts toward a more activated basal state with age. Naive CD4+ T cells, considered relatively resistant to aging, exhibited pronounced transcriptional and epigenetic reprogramming. Moreover, we discovered a novel CD8αα+ T cell subset lost with age that is epigenetically poised for rapid effector responses and has distinct inhibitory, costimulatory and tissue-homing properties. Together, these data reveal new insights into age-associated changes in the T cell compartment that may contribute to differential immune responses.


Asunto(s)
Subgrupos de Linfocitos T , Transcriptoma , Niño , Humanos , Anciano , Envejecimiento/genética , Epítopos/metabolismo , Análisis de la Célula Individual
5.
Cell ; 183(7): 1946-1961.e15, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33306960

RESUMEN

Lymphocyte migration is essential for adaptive immune surveillance. However, our current understanding of this process is rudimentary, because most human studies have been restricted to immunological analyses of blood and various tissues. To address this knowledge gap, we used an integrated approach to characterize tissue-emigrant lineages in thoracic duct lymph (TDL). The most prevalent immune cells in human and non-human primate efferent lymph were T cells. Cytolytic CD8+ T cell subsets with effector-like epigenetic and transcriptional signatures were clonotypically skewed and selectively confined to the intravascular circulation, whereas non-cytolytic CD8+ T cell subsets with stem-like epigenetic and transcriptional signatures predominated in tissues and TDL. Moreover, these anatomically distinct gene expression profiles were recapitulated within individual clonotypes, suggesting parallel differentiation programs independent of the expressed antigen receptor. Our collective dataset provides an atlas of the migratory immune system and defines the nature of tissue-emigrant CD8+ T cells that recirculate via TDL.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Animales , Diferenciación Celular , Células Clonales , Citotoxicidad Inmunológica , Epigénesis Genética , Humanos , Memoria Inmunológica , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Macaca mulatta , Subgrupos de Linfocitos T/inmunología , Transcripción Genética , Transcriptoma/genética
6.
Nat Immunol ; 23(8): 1183-1192, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35902637

RESUMEN

Anti-programmed death-1 (anti-PD-1) immunotherapy reinvigorates CD8 T cell responses in patients with cancer but PD-1 is also expressed by other immune cells, including follicular helper CD4 T cells (Tfh) which are involved in germinal centre responses. Little is known, however, about the effects of anti-PD-1 immunotherapy on noncancer immune responses in humans. To investigate this question, we examined the impact of anti-PD-1 immunotherapy on the Tfh-B cell axis responding to unrelated viral antigens. Following influenza vaccination, a subset of adults receiving anti-PD-1 had more robust circulating Tfh responses than adults not receiving immunotherapy. PD-1 pathway blockade resulted in transcriptional signatures of increased cellular proliferation in circulating Tfh and responding B cells compared with controls. These latter observations suggest an underlying change in the Tfh-B cell and germinal centre axis in a subset of immunotherapy patients. Together, these results demonstrate dynamic effects of anti-PD-1 therapy on influenza vaccine responses and highlight analytical vaccination as an approach that may reveal underlying immune predisposition to adverse events.


Asunto(s)
Vacunas contra la Influenza , Adulto , Humanos , Inmunidad Humoral , Estaciones del Año , Linfocitos T Colaboradores-Inductores , Vacunación
8.
Immunity ; 51(5): 840-855.e5, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31606264

RESUMEN

TCF-1 is a key transcription factor in progenitor exhausted CD8 T cells (Tex). Moreover, this Tex cell subset mediates responses to PD-1 checkpoint pathway blockade. However, the role of the transcription factor TCF-1 in early fate decisions and initial generation of Tex cells is unclear. Single-cell RNA sequencing (scRNA-seq) and lineage tracing identified a TCF-1+Ly108+PD-1+ CD8 T cell population that seeds development of mature Tex cells early during chronic infection. TCF-1 mediated the bifurcation between divergent fates, repressing development of terminal KLRG1Hi effectors while fostering KLRG1Lo Tex precursor cells, and PD-1 stabilized this TCF-1+ Tex precursor cell pool. TCF-1 mediated a T-bet-to-Eomes transcription factor transition in Tex precursors by promoting Eomes expression and drove c-Myb expression that controlled Bcl-2 and survival. These data define a role for TCF-1 in early-fate-bifurcation-driving Tex precursor cells and also identify PD-1 as a protector of this early TCF-1 subset.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Redes Reguladoras de Genes , Factor 1 de Transcripción de Linfocitos T/metabolismo , Transcripción Genética , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Enfermedad Crónica , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Factor 1 de Transcripción de Linfocitos T/genética , Virosis/genética , Virosis/inmunología , Virosis/virología
9.
J Infect Dis ; 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38437622

RESUMEN

Patients with B-cell lymphomas have altered cellular components of vaccine responses due to malignancy and therapy, and the optimal timing of vaccination relative to therapy remains unknown. SARS-CoV-2 vaccines created an opportunity for new insights in vaccine timing because patients were challenged with a novel antigen across multiple phases of treatment. We studied serologic mRNA vaccine response in retrospective and prospective cohorts with lymphoma and CLL, paired with clinical and research immune parameters. Reduced serologic response was observed more frequently during active therapies, but non-response was also common within observation and post-treatment groups. Total IgA and IgM correlated with successful vaccine response. In individuals treated with CART-19, non-response was associated with reduced B and T follicular helper cells. Predictors of vaccine response varied by disease and therapeutic group, and therefore further studies of immune health during and after cancer therapies are needed to allow individualized vaccine timing.

10.
Pediatr Transplant ; 26(3): e14220, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34994041

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) is an important cause of morbidity and mortality in pediatric solid organ transplant (SOT) recipients. However, the impact of asymptomatic CMV infections (ie, DNAemia) on clinical outcomes is not well established. METHODS: We performed a retrospective cohort study of children undergoing first SOT at our institution from January 2012 to June 2018. We evaluated the epidemiology of CMV infections and performed multivariable Cox regression to assess the association between CMV DNAemia without disease or CMV disease (syndrome or end-organ disease) on negative outcomes (death, re-transplantation, or moderate/severe rejection) within the first year after SOT. RESULTS: Among 271 individuals, 43 (15.9%) developed ≥1 CMV infection during the first year after SOT. There were 56 unique CMV infections including 14 episodes of CMV disease. In 167 patients offered CMV prophylaxis, only 8 (4.8%) developed their first CMV DNAemia episode while on prophylaxis 32 developed CMV DNAemia after prophylaxis completion; only 1 episode of CMV disease occurred while on antiviral prophylaxis. When accounting for receipt of ATG, oral steroids, and number of immunosuppressives on a given day, CMV disease was more strongly associated with negative outcomes (Hazard Ratio (HR): 3.28, 95% CI: 0.73-14.64; p = .12) than CMV DNAemia without disease (HR 1.42, 95% CI: 0.19- 10.79; p = .74), although not to a statistically significant degree. CONCLUSIONS: Most CMV infections occurred after completion of antiviral prophylaxis. CMV disease was more strongly associated with negative outcomes than asymptomatic CMV DNAemia and should be the focus of CMV prevention practices.


Asunto(s)
Infecciones por Citomegalovirus , Trasplante de Órganos , Antivirales/uso terapéutico , Niño , Citomegalovirus , Infecciones por Citomegalovirus/diagnóstico , Infecciones por Citomegalovirus/epidemiología , Infecciones por Citomegalovirus/etiología , Humanos , Trasplante de Órganos/efectos adversos , Estudios Retrospectivos , Receptores de Trasplantes
11.
Pediatr Blood Cancer ; 67(11): e28693, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32885904

RESUMEN

There are no proven safe and effective therapies for children who develop life-threatening complications of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Convalescent plasma (CP) has demonstrated potential benefit in adults with SARS-CoV-2, but has theoretical risks.We present the first report of CP in children with life-threatening coronavirus disease 2019 (COVID-19), providing data on four pediatric patients with acute respiratory distress syndrome. We measured donor antibody levels and recipient antibody response prior to and following CP infusion. Infusion of CP was not associated with antibody-dependent enhancement (ADE) and did not suppress endogenous antibody response. We found CP was safe and possibly efficacious. Randomized pediatric trials are needed.


Asunto(s)
COVID-19/terapia , Síndrome de Dificultad Respiratoria/terapia , Adolescente , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/uso terapéutico , COVID-19/complicaciones , Humanos , Inmunización Pasiva/métodos , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Síndrome de Dificultad Respiratoria/etiología , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad , Sueroterapia para COVID-19
12.
medRxiv ; 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38293197

RESUMEN

Multisystem Inflammatory Syndrome in Childhood (MIS-C) follows SARS-CoV-2 infection and frequently leads to intensive care unit admission. The inability to rapidly discriminate MIS-C from similar febrile illnesses delays treatment and leads to misdiagnosis. To identify diagnostic discriminators at the time of emergency department presentation, we enrolled 104 children who met MIS-C screening criteria, 14 of whom were eventually diagnosed with MIS-C. Before treatment, we collected breath samples for volatiles and peripheral blood for measurement of plasma proteins and immune cell features. Clinical and laboratory features were used as inputs for a machine learning model to determine diagnostic importance. MIS-C was associated with significant changes in breath volatile organic compound (VOC) composition as well as increased plasma levels of secretory phospholipase A2 (PLA2G2A) and lipopolysaccharide binding protein (LBP). In an integrated model of all analytes, the proportion of TCRVß21.3+ non-naive CD4 T cells expressing Ki-67 had a high sensitivity and specificity for MIS-C, with diagnostic accuracy further enhanced by low sodium and high PLA2G2A. We anticipate that accurate diagnosis will become increasingly difficult as MIS-C becomes less common. Clinical validation and application of this diagnostic model may improve outcomes in children presenting with multisystem febrile illnesses.

13.
Sci Immunol ; 8(83): eade2335, 2023 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-37235682

RESUMEN

The ability of most patients with selective immunoglobulin A (IgA) deficiency (SIgAD) to remain apparently healthy has been a persistent clinical conundrum. Compensatory mechanisms, including IgM, have been proposed, yet it remains unclear how secretory IgA and IgM work together in the mucosal system and, on a larger scale, whether the systemic and mucosal anti-commensal responses are redundant or have unique features. To address this gap in knowledge, we developed an integrated host-commensal approach combining microbial flow cytometry and metagenomic sequencing (mFLOW-Seq) to comprehensively define which microbes induce mucosal and systemic antibodies. We coupled this approach with high-dimensional immune profiling to study a cohort of pediatric patients with SIgAD and household control siblings. We found that mucosal and systemic antibody networks cooperate to maintain homeostasis by targeting a common subset of commensal microbes. In IgA-deficiency, we find increased translocation of specific bacterial taxa associated with elevated levels of systemic IgG targeting fecal microbiota. Associated features of immune system dysregulation in IgA-deficient mice and humans included elevated levels of inflammatory cytokines, enhanced follicular CD4 T helper cell frequency and activation, and an altered CD8 T cell activation state. Although SIgAD is clinically defined by the absence of serum IgA, the symptomatology and immune dysregulation were concentrated in the SIgAD participants who were also fecal IgA deficient. These findings reveal that mucosal IgA deficiency leads to aberrant systemic exposures and immune responses to commensal microbes, which increase the likelihood of humoral and cellular immune dysregulation and symptomatic disease in patients with IgA deficiency.


Asunto(s)
Deficiencia de IgA , Humanos , Niño , Ratones , Animales , Inmunoglobulina A Secretora , Inmunoglobulina M , Homeostasis
14.
Proc Natl Acad Sci U S A ; 106(33): 14010-5, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19666607

RESUMEN

We previously identified the aberrantly expressed cell cycle regulator cyclin B1 as a tumor antigen recognized by antibodies and T cells from patients with breast, lung, and head and neck cancers. Ordinarily expressed only transiently in the G2/M stage of the cell cycle in normal cells, cyclin B1 is constitutively expressed at high levels in the cytoplasm of these and many other tumor types, leading to its recognition by the cancer patient's immune system. We report here an unexpected observation that cyclin B1-specific antibody and memory CD4 and CD8 T cells are also found in many healthy individuals who have no history of cancer. Moreover, young as well as older healthy people have these responses suggesting that events other than cancer, which occur either early in life or throughout life, may lead to aberrant cyclin B1 expression and anti-cyclin B1 immunity. The role, if any, of immunity to this tumor-associated antigen is not known. We wanted to determine specifically whether immunity to cyclin B1 might be important in the immunosurveillance of cyclin B1+ tumors. We therefore tested in mice the effectiveness of vaccine-elicited anti-cyclin B1 immunity against a cyclin B1+ mouse tumor that was chosen based on our published observation that cyclin B1 overexpression is associated with the lack of p53 function. We found that cyclin B1 DNA prime-protein boost vaccine protected mice from a challenge with a tumor cell line that was established from a tumor arising in the p53(-/-) mouse that spontaneously overexpresses cyclin B1.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Ciclina B/inmunología , Regulación Neoplásica de la Expresión Génica , Neoplasias/inmunología , Adulto , Anciano , Animales , Antígenos de Neoplasias , Linfocitos T CD4-Positivos/metabolismo , Ciclina B/metabolismo , Ciclina B1 , Células Dendríticas/metabolismo , Genes p53 , Humanos , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Monocitos/metabolismo
15.
Front Immunol ; 13: 834988, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35309299

RESUMEN

Patients with COVID-19 present with a wide variety of clinical manifestations. Thromboembolic events constitute a significant cause of morbidity and mortality in patients infected with SARS-CoV-2. Severe COVID-19 has been associated with hyperinflammation and pre-existing cardiovascular disease. Platelets are important mediators and sensors of inflammation and are directly affected by cardiovascular stressors. In this report, we found that platelets from severely ill, hospitalized COVID-19 patients exhibited higher basal levels of activation measured by P-selectin surface expression and had poor functional reserve upon in vitro stimulation. To investigate this question in more detail, we developed an assay to assess the capacity of plasma from COVID-19 patients to activate platelets from healthy donors. Platelet activation was a common feature of plasma from COVID-19 patients and correlated with key measures of clinical outcome including kidney and liver injury, and APACHEIII scores. Further, we identified ferritin as a pivotal clinical marker associated with platelet hyperactivation. The COVID-19 plasma-mediated effect on control platelets was highest for patients that subsequently developed inpatient thrombotic events. Proteomic analysis of plasma from COVID-19 patients identified key mediators of inflammation and cardiovascular disease that positively correlated with in vitro platelet activation. Mechanistically, blocking the signaling of the FcγRIIa-Syk and C5a-C5aR pathways on platelets, using antibody-mediated neutralization, IgG depletion or the Syk inhibitor fostamatinib, reversed this hyperactivity driven by COVID-19 plasma and prevented platelet aggregation in endothelial microfluidic chamber conditions. These data identified these potentially actionable pathways as central for platelet activation and/or vascular complications and clinical outcomes in COVID-19 patients. In conclusion, we reveal a key role of platelet-mediated immunothrombosis in COVID-19 and identify distinct, clinically relevant, targetable signaling pathways that mediate this effect.


Asunto(s)
Plaquetas/inmunología , COVID-19/inmunología , Complemento C5a/metabolismo , Receptor de Anafilatoxina C5a/metabolismo , Receptores de IgG/metabolismo , SARS-CoV-2/fisiología , Tromboembolia/inmunología , Adulto , Aminopiridinas/farmacología , Células Cultivadas , Femenino , Hospitalización , Humanos , Masculino , Morfolinas/farmacología , Activación Plaquetaria , Pirimidinas/farmacología , Índice de Severidad de la Enfermedad , Transducción de Señal , Quinasa Syk/antagonistas & inhibidores
16.
Curr Pediatr Rep ; 9(4): 83-92, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34692237

RESUMEN

Purpose of Review: We highlight the new clinical entity multisystem inflammatory syndrome in children (MIS-C), the progress in understanding its immunopathogenesis, and compare and contrast the clinical and immunologic features of MIS-C with Kawasaki disease (KD). Recent Findings: Studies show immune dysregulation in MIS-C including T lymphocyte depletion and activation, T cell receptor Vbeta skewing, elevated plasmablast frequencies, increased markers of vascular pathology, and decreased numbers and functional profiles of antigen-presenting cells. Summary: MIS-C is a late manifestation of infection with SARS-CoV-2 associated with marked immune activation and many potential mechanisms of immunopathogenesis. MIS-C and KD have clinical similarities but are distinct. Myocardial dysfunction with or without mild coronary artery dilation can occur in MIS-C but generally corrects within weeks. In contrast, the coronary arteries are the primary target in KD, and coronary artery sequelae can be lifelong. Supportive care and anti-inflammatory therapy appear to hasten improvement in children with MIS-C, and there is hope that vaccines will prevent its development.

17.
Cell Rep Med ; 2(5): 100262, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34095875

RESUMEN

Humoral immune responses are dysregulated with aging, but the cellular and molecular pathways involved remain incompletely understood. In particular, little is known about the effects of aging on T follicular helper (Tfh) CD4 cells, the key cells that provide help to B cells for effective humoral immunity. We performed transcriptional profiling and cellular analysis on circulating Tfh before and after influenza vaccination in young and elderly adults. First, whole-blood transcriptional profiling shows that ICOS+CD38+ cTfh following vaccination preferentially enriches in gene sets associated with youth versus aging compared to other circulating T cell types. Second, vaccine-induced ICOS+CD38+ cTfh from the elderly had increased the expression of genes associated with inflammation, including tumor necrosis factor-nuclear factor κB (TNF-NF-κB) pathway activation. Finally, vaccine-induced ICOS+CD38+ cTfh display strong enrichment for signatures of underlying age-associated biological changes. These data highlight the ability to use vaccine-induced cTfh as cellular "biosensors" of underlying inflammatory and/or overall immune health.


Asunto(s)
Factores de Edad , Linfocitos B/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Inflamación/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Humanos , Inmunidad Humoral/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Inflamación/inmunología , Activación de Linfocitos/inmunología , Recuento de Linfocitos/métodos , Vacunación/métodos , Vacunas/metabolismo
18.
Nat Commun ; 12(1): 7222, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34893640

RESUMEN

Multi-system Inflammatory Syndrome in Children (MIS-C) is a major complication of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection in pediatric patients. Weeks after an often mild or asymptomatic initial infection with SARS-CoV-2 children may present with a severe shock-like picture and marked inflammation. Children with MIS-C present with varying degrees of cardiovascular and hyperinflammatory symptoms. Here we perform a comprehensive analysis of the plasma proteome of more than 1400 proteins in children with SARS-CoV-2. We hypothesize that the proteome would reflect heterogeneity in hyperinflammation and vascular injury, and further identify pathogenic mediators of disease. We show that protein signatures demonstrate overlap between MIS-C, and the inflammatory syndromes macrophage activation syndrome (MAS) and thrombotic microangiopathy (TMA). We demonstrate that PLA2G2A is an important marker of MIS-C that associates with TMA. We find that IFNγ responses are dysregulated in MIS-C patients, and that IFNγ levels delineate clinical heterogeneity.


Asunto(s)
COVID-19/complicaciones , Endotelio Vascular/fisiopatología , Interferón gamma/inmunología , Proteoma , Síndrome de Respuesta Inflamatoria Sistémica/patología , Biomarcadores , COVID-19/metabolismo , COVID-19/patología , Estudios de Casos y Controles , Quimiocina CXCL9 , Niño , Fosfolipasas A2 Grupo II , Humanos , Inflamación , Interleucina-10 , Proteómica , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Enfermedades Vasculares
19.
medRxiv ; 2021 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33907759

RESUMEN

Multi-system Inflammatory Syndrome in Children (MIS-C) is a major complication of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic in pediatric patients. Weeks after an often mild or asymptomatic initial infection with SARS-CoV-2 children may present with a severe shock-like picture and marked inflammation. Children with MIS-C present with varying degrees of cardiovascular and hyperinflammatory symptoms. We performed a comprehensive analysis of the plasma proteome of more than 1400 proteins in children with SARS-CoV-2. We hypothesized that the proteome would reflect heterogeneity in hyperinflammation and vascular injury, and further identify pathogenic mediators of disease. Protein signatures demonstrated overlap between MIS-C, and the inflammatory syndromes macrophage activation syndrome (MAS) and thrombotic microangiopathy (TMA). We demonstrate that PLA2G2A is a key marker of MIS-C that associates with TMA. We found that IFNγ responses are dysregulated in MIS-C patients, and that IFNγ levels delineate clinical heterogeneity.

20.
J Pediatric Infect Dis Soc ; 10(5): 669-673, 2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-33263756

RESUMEN

Severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) antibody responses in children remain poorly characterized. Here, we show that pediatric patients with multisystem inflammatory syndrome in children (MIS-C) possess higher SARS-CoV-2 spike immunoglobulin G (IgG) titers compared with those with severe coronavirus disease 2019, likely reflecting a longer time since the onset of infection in MIS-C patients.


Asunto(s)
Anticuerpos Antivirales/inmunología , Formación de Anticuerpos , COVID-19/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Prueba Serológica para COVID-19 , Niño , Femenino , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Masculino , SARS-CoV-2 , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA