Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
JCI Insight ; 4(9)2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31045577

RESUMEN

Human antibody-secreting cells (ASCs) triggered by immunization are globally recognized as CD19loCD38hiCD27hi. Yet, different vaccines give rise to antibody responses of different longevity, suggesting ASC populations are heterogeneous. We define circulating-ASC heterogeneity in vaccine responses using multicolor flow cytometry, morphology, VH repertoire, and RNA transcriptome analysis. We also tested differential survival using a human cell-free system that mimics the bone marrow (BM) microniche. In peripheral blood, we identified 3 CD19+ and 2 CD19- ASC subsets. All subsets contributed to the vaccine-specific responses and were characterized by in vivo proliferation and activation. The VH repertoire demonstrated strong oligoclonality with extensive interconnectivity among the 5 subsets and switched memory B cells. Transcriptome analysis showed separation of CD19+ and CD19- subsets that included pathways such as cell cycle, hypoxia, TNF-α, and unfolded protein response. They also demonstrated similar long-term in vitro survival after 48 days. In summary, vaccine-induced ASCs with different surface markers (CD19 and CD138) are derived from shared proliferative precursors yet express distinctive transcriptomes. Equal survival indicates that all ASC compartments are endowed with long-lived potential. Accordingly, in vivo survival of peripheral long-lived plasma cells may be determined in part by their homing and residence in the BM microniche.


Asunto(s)
Células Productoras de Anticuerpos/inmunología , Células Plasmáticas/inmunología , Transcriptoma , Adulto , Anciano , Formación de Anticuerpos , Antígenos CD19/inmunología , Linfocitos B/inmunología , Médula Ósea/inmunología , Células de la Médula Ósea/inmunología , Femenino , Humanos , Inmunización , Inmunoglobulina G , Cinética , Masculino , Persona de Mediana Edad , Fenotipo , Tétanos/inmunología , Vacunación , Adulto Joven
2.
J Virol ; 93(8)2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30728266

RESUMEN

Memory B cells (MBCs) are key determinants of the B cell response to influenza virus infection and vaccination, but the effect of different forms of influenza antigen exposure on MBC populations has received little attention. We analyzed peripheral blood mononuclear cells and plasma collected following human H3N2 influenza infection to investigate the relationship between hemagglutinin-specific antibody production and changes in the size and character of hemagglutinin-reactive MBC populations. Infection produced increased concentrations of plasma IgG reactive to the H3 head of the infecting virus, to the conserved stalk, and to a broad chronological range of H3s consistent with original antigenic sin responses. H3-reactive IgG MBC expansion after infection included reactivity to head and stalk domains. Notably, expansion of H3 head-reactive MBC populations was particularly broad and reflected original antigenic sin patterns of IgG production. Findings also suggest that early-life H3N2 infection "imprints" for strong H3 stalk-specific MBC expansion. Despite the breadth of MBC expansion, the MBC response included an increase in affinity for the H3 head of the infecting virus. Overall, our findings indicate that H3-reactive MBC expansion following H3N2 infection is consistent with maintenance of response patterns established early in life, but nevertheless includes MBC adaptation to the infecting virus.IMPORTANCE Rapid and vigorous virus-specific antibody responses to influenza virus infection and vaccination result from activation of preexisting virus-specific memory B cells (MBCs). Understanding the effects of different forms of influenza virus exposure on MBC populations is therefore an important guide to the development of effective immunization strategies. We demonstrate that exposure to the influenza hemagglutinin via natural infection enhances broad protection through expansion of hemagglutinin-reactive MBC populations that recognize head and stalk regions of the molecule. Notably, we show that hemagglutinin-reactive MBC expansion reflects imprinting by early-life infection and that this might apply to stalk-reactive, as well as to head-reactive, MBCs. Our findings provide experimental support for the role of MBCs in maintaining imprinting effects and suggest a mechanism by which imprinting might confer heterosubtypic protection against avian influenza viruses. It will be important to compare our findings to the situation after influenza vaccination.


Asunto(s)
Linfocitos B/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Memoria Inmunológica , Subtipo H3N2 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Estaciones del Año , Anticuerpos Antivirales/inmunología , Humanos , Inmunoglobulina G/inmunología , Subtipo H1N1 del Virus de la Influenza A
3.
Vaccine ; 35(38): 5163-5171, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28807607

RESUMEN

BACKGROUND: Preterm (PT) infants are at greater risk for severe influenza infection and experience decrements in long-term antibody responses to vaccines. This may related to defects in antibody secreting cell (ASC) generation. OBJECTIVE: To investigate the relationships among the frequencies of influenza-specific antibody secreting cells, ASC numbers and subsets, and antibody responses to influenza vaccines (IV) among PT and full-term (FT) infants. DESIGN/METHODS: We enrolled 11 former PT (≤32weeks' gestation, ≤1500 g' birth weight) and 11FT infants, 6-17months of age, receiving their first influenza immunizations. Infants received two doses of inactivated trivalent (T)IV or quadrivalent (Q)IV during the 2012-2013 and 2013-2014 influenza seasons, respectively, at 0 and 28days, and blood was drawn at 0, 10, 35, and 56days and 9months. Vaccine-specific antibody was measured by hemagglutination inhibition (HAI) at 0 and 56days and 9months, vaccine-specific ASC numbers by enzyme linked immunospot (ELISPOT) at 10 and 35days, and ASC subsets by flow cytometry at 0, 10 and 35days. RESULTS: PT infants had post-vaccine HAI titers to all 4 vaccine strains at least equal to FT infants at 56days and 9months after beginning immunization. Influenza-specific ASC ELISPOT responses at 35days were higher among PT than FT infants (median 100 v. 30 per 106 PBMC, p=0.04). ASC numbers at 35days were positively correlated with serum HAI titers at 56days (ρ=0.50-0.80). There were no statistical differences between PT and FT infants in the frequency of five ASC subsets and no specific ASC subset correlated with durability of serum antibody titers. CONCLUSIONS: Influenza-specific ASC numbers in both FT and PT infants correlated with peak antibody titers, but ASC subsets did not correlate with durability of antibody response.


Asunto(s)
Formación de Anticuerpos/fisiología , Vacunas contra la Influenza/uso terapéutico , Células Productoras de Anticuerpos/metabolismo , Niño , Femenino , Citometría de Flujo , Humanos , Lactante , Recién Nacido , Vacunas contra la Influenza/efectos adversos , Masculino , Nacimiento Prematuro , Estudios Prospectivos
4.
Immunity ; 43(1): 132-45, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26187412

RESUMEN

Antibody responses to viral infections are sustained for decades by long-lived plasma cells (LLPCs). However, LLPCs have yet to be characterized in humans. Here we used CD19, CD38, and CD138 to identify four PC subsets in human bone marrow (BM). We found that the CD19(-)CD38(hi)CD138(+) subset was morphologically distinct, differentially expressed PC-associated genes, and exclusively contained PCs specific for viral antigens to which the subjects had not been exposed for more than 40 years. Protein sequences of measles- and mumps-specific circulating antibodies were encoded for by CD19(-)CD38(hi)CD138(+) PCs in the BM. Finally, we found that CD19(-)CD38(hi)CD138(+) PCs had a distinct RNA transcriptome signature and human immunoglobulin heavy chain (VH) repertoire that was relatively uncoupled from other BM PC subsets and probably represents the B cell response's "historical record" of antigenic exposure. Thus, our studies define human LLPCs and provide a mechanism for the life-long maintenance of anti-viral antibodies in the serum.


Asunto(s)
Anticuerpos Antivirales/inmunología , Células de la Médula Ósea/inmunología , Virus del Sarampión/inmunología , Virus de la Parotiditis/inmunología , Células Plasmáticas/inmunología , ADP-Ribosil Ciclasa 1/metabolismo , Adulto , Anciano , Anticuerpos Antivirales/sangre , Antígenos CD19/metabolismo , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , ARN Mensajero/genética , Sindecano-1/metabolismo , Adulto Joven
5.
J Infect Dis ; 212(8): 1270-8, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25838266

RESUMEN

Recent studies have shown that live attenuated influenza vaccines (LAIVs) expressing avian influenza virus hemagglutinins (HAs) prime for strong protective antibody responses to an inactivated influenza vaccine (IIV) containing the HA. To better understand this priming effect, we compared H7 HA head and stalk domain-specific B-cell responses in H7N7 LAIV-primed subjects and non-H7-primed controls after a single dose of H7N7 IIV. As previously reported, H7N7 LAIV-primed subjects but not control subjects generated strong hemagglutination-inhibiting and neutralizing antibody responses to the H7N7 IIV. Here, we found that the quantity, epitope diversity, and affinity of H7 head-specific antibodies increased rapidly in only H7N7 LAIV-primed subjects after receipt of the IIV. However, all cohorts generated a vigorous, high-affinity, stalk-specific antibody response. Consistent increases in circulating memory B-cell frequencies after receipt of the IIV reflected the specificity of high-affinity antibody production. Our findings emphasize the value of LAIVs as a vehicle for prepandemic vaccination.


Asunto(s)
Anticuerpos Antivirales/sangre , Subtipo H7N7 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Anticuerpos Neutralizantes/sangre , Linfocitos B/inmunología , Estudios de Cohortes , Pruebas de Inhibición de Hemaglutinación , Humanos , Gripe Humana/prevención & control , Gripe Humana/virología , Pruebas de Neutralización , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/inmunología
6.
J Immunol ; 186(9): 5514-21, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21441455

RESUMEN

Surges of serum Abs after immunization and infection are highly specific for the offending Ag, and recent studies demonstrate that vaccines induce transient increases in circulating Ab-secreting cells (ASCs). These ASCs are highly enriched but not universally specific for the immunizing Ag, suggesting that a fraction of these ASCs could arise from polyclonal bystander stimulation of preexisting memory cells to unrelated Ags. This model is proposed to explain maintenance of long-lived serological memory in the absence of Ag exposure. To test this model, we measure the ability of respiratory syncytial virus and influenza virus infection or immunizations to influenza virus, tetanus toxoid, hepatitis B Ag, and human papillomavirus to stimulate bystander memory cells specific for other major environmental Ags that represent a large fraction of the preexisting memory B compartment. Bystander or nonspecific ASC responses to respiratory syncytial virus and tetanus could not be detected above the background levels in healthy adults, despite the presence of circulating memory B cells specific for the corresponding Ags. Nonspecific ASC responses in the healthy subjects and cord blood samples were similar. In contrast, both vaccination and infection induce massive expansion of circulating Ag-specific ASCs without significant increases in the frequencies of ASCs against unrelated Ags. Hence, nonspecific stimulation of memory B cells is unlikely to contribute to the mechanisms of long-term serological memory against major human pathogens. Additionally, high specificity of circulating ASCs after antigenic challenge highlights the diagnostic value of interrogating ASCs as an ideal single-time-point diagnostic immune surrogate for serology during acute infection.


Asunto(s)
Células Productoras de Anticuerpos/inmunología , Efecto Espectador/inmunología , Memoria Inmunológica , Infecciones del Sistema Respiratorio/inmunología , Vacunación , Virosis/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos Virales/inmunología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
7.
J Infect Dis ; 202(11): 1659-66, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20979459

RESUMEN

BACKGROUND: The specificity and duration of circulating human antibody-secreting cells (ASCs) after vaccination have been well described, but characteristics of ASCs during acute respiratory infections have not been well studied. METHODS: Circulating antigen-specific ASCs were measured at 3 time points (enrollment, days 10-16, and days 22-45) in 40 adults during respiratory syncytial virus (RSV) infection. RESULTS: Of the 40 patients, 36 (90%) had detectable circulating RSV F protein-specific ASCs within 11 days after illness onset. The magnitude of the RSV-specific ASCs was 1-1500 spots per 106 peripheral blood mononuclear cells (mean frequency [± standard deviation], 200 ± 256 spots per 106 peripheral blood mononuclear cells). ASCs were detected on day 8-16 and day 22-45 after symptom onset in 78% and 48% of subjects, respectively. Subjects shedding virus for >10 days were more likely to have a positive response to ASC enzyme-linked immunospot assay at the late time point than those shedding for ≤10 days (8 of 12 subjects vs 2 of 11 subjects; P = .02). CONCLUSIONS: The kinetics of ASC circulation during acute mucosal viral infections was more prolonged than that we had observed after a single intramuscular injection with inactivated influenza vaccine in a study reported elsewhere. The association between the duration of virus shedding and the persistence of detectable viral-specific ASCs suggests that ongoing antigen persistence induces a prolonged temporal pattern of ASC generation.


Asunto(s)
Anticuerpos Antivirales/sangre , Células Productoras de Anticuerpos/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Antígenos Virales , Linfocitos B , Ensayo de Immunospot Ligado a Enzimas , Femenino , Humanos , Masculino , Persona de Mediana Edad , New York , Estudios Prospectivos , Infecciones por Virus Sincitial Respiratorio/sangre , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
8.
Cytometry B Clin Cytom ; 78 Suppl 1: S69-82, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20839340

RESUMEN

BACKGROUND: Advances in multiparameter flow cytometry (FCM) now allow for the independent detection of larger numbers of fluorochromes on individual cells, generating data with increasingly higher dimensionality. The increased complexity of these data has made it difficult to identify cell populations from high-dimensional FCM data using traditional manual gating strategies based on single-color or two-color displays. METHODS: To address this challenge, we developed a novel program, FLOCK (FLOw Clustering without K), that uses a density-based clustering approach to algorithmically identify biologically relevant cell populations from multiple samples in an unbiased fashion, thereby eliminating operator-dependent variability. RESULTS: FLOCK was used to objectively identify seventeen distinct B-cell subsets in a human peripheral blood sample and to identify and quantify novel plasmablast subsets responding transiently to tetanus and other vaccinations in peripheral blood. FLOCK has been implemented in the publically available Immunology Database and Analysis Portal-ImmPort (http://www.immport.org)-for open use by the immunology research community. CONCLUSIONS: FLOCK is able to identify cell subsets in experiments that use multiparameter FCM through an objective, automated computational approach. The use of algorithms like FLOCK for FCM data analysis obviates the need for subjective and labor-intensive manual gating to identify and quantify cell subsets. Novel populations identified by these computational approaches can serve as hypotheses for further experimental study.


Asunto(s)
Subgrupos de Linfocitos B/patología , Vacuna contra Difteria y Tétanos/inmunología , Citometría de Flujo/métodos , Inmunofenotipificación/métodos , Adulto , Anciano , Algoritmos , Subgrupos de Linfocitos B/clasificación , Subgrupos de Linfocitos B/inmunología , Análisis por Conglomerados , Biología Computacional/métodos , Vacuna contra Difteria y Tétanos/administración & dosificación , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador , Masculino , Persona de Mediana Edad , Reconocimiento de Normas Patrones Automatizadas , Vacunación/métodos , Adulto Joven
9.
Vaccine ; 28(20): 3582-7, 2010 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-20298818

RESUMEN

UNLABELLED: Upon vaccination, B cells differentiate into antibody secreting cells (ASCs) that migrate via the circulation to tissues. The kinetics of this response and the relationship of circulating ASCs to protective antibody titers have not been completely explored. METHODS: Influenza-specific and total-IgG ASCs were enumerated by Elispot and flow cytometry daily in the blood in 6 healthy adults after trivalent influenza vaccination (TIV). RESULTS: Peak H1-specific IgG ASC frequencies occurred variably from day 5 to 8 and correlated with the fold-rise rise in hemagglutination inhibition (HAI titers); r=0.91, p=0.006. H3-specific IgG ASC frequencies correlated less well, perhaps due to a mismatch of the H3 protein in the vaccine and that used in the Elispot assay. Peak frequencies of vaccine-specific and total-IgG ASCs were 0.3% and 0.8%, respectively, of peripheral blood mononuclear cells (PBMC). Peak TIV-, H1-, H3-, and total-IgG ASC frequencies were 1736+/-1133, 626+/-520, 592+/-463, and 4091+/-2019 spots/10(6) PBMC, respectively. Peak TIV-, H1-, and H3-specific IgG ASC of total-IgG ASC frequencies constituted 63%+/-21, 26%+/-10, 22%+/-17, respectively. CONCLUSION: After immunization with inactivated influenza vaccine the peak in influenza-specific ASC frequencies is variable but correlates well with the magnitude of protective HAI responses.


Asunto(s)
Formación de Anticuerpos/inmunología , Células Productoras de Anticuerpos/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Adulto , Femenino , Pruebas de Inhibición de Hemaglutinación , Humanos , Inmunoglobulina G/sangre , Masculino , Persona de Mediana Edad , Vacunación , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA