Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 437
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 204(10): 2641-2650, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32253245

RESUMEN

Although STAT1 tyrosine-701 phosphorylation (designated STAT1-pY701) is indispensable for STAT1 function, the requirement for STAT1 serine-727 phosphorylation (designated STAT1-pS727) during systemic autoimmune and antipathogen responses remains unclear. Using autoimmune-prone B6.Sle1b mice expressing a STAT1-S727A mutant in which serine is replaced by alanine, we report in this study that STAT1-pS727 promotes autoimmune Ab-forming cell (AFC) and germinal center (GC) responses, driving autoantibody production and systemic lupus erythematosus (SLE) development. In contrast, STAT1-pS727 is not required for GC, T follicular helper cell (Tfh), and Ab responses to various foreign Ags, including pathogens. STAT1-pS727 is also not required for gut microbiota and dietary Ag-driven GC and Tfh responses in B6.Sle1b mice. By generating B cell-specific bone marrow chimeras, we demonstrate that STAT1-pS727 plays an important B cell-intrinsic role in promoting autoimmune AFC, GC, and Tfh responses, leading to SLE-associated autoantibody production. Our analysis of the TLR7-accelerated B6.Sle1b.Yaa SLE disease model expressing a STAT1-S727A mutant reveals STAT1-pS727-mediated regulation of autoimmune AFC and GC responses and lupus nephritis development. Together, we identify previously unrecognized differential regulation of systemic autoimmune and antipathogen responses by STAT1-pS727. Our data implicate STAT1-pS727 as a therapeutic target for SLE without overtly affecting STAT1-mediated protection against pathogenic infections.


Asunto(s)
Linfocitos B/inmunología , Centro Germinal/inmunología , Lupus Eritematoso Sistémico/metabolismo , Factor de Transcripción STAT1/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Autoanticuerpos/sangre , Autoantígenos/inmunología , Autoinmunidad , Linfocitos B/trasplante , Humanos , Lupus Eritematoso Sistémico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fosforilación , Dominios Proteicos/genética , Factor de Transcripción STAT1/genética , Serina/genética , Activación Transcripcional , Quimera por Trasplante
2.
Rheumatology (Oxford) ; 60(10): 4850-4854, 2021 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33410495

RESUMEN

OBJECTIVE: Mevalonic aciduria represents the most severe form of mevalonate kinase deficiency (MKD). Patients with mevalonic aciduria have an incomplete response even to high doses of anti-cytokine drugs such as anakinra or canakinumab and stem cell transplantation (SCT) represents a possible therapy for this severe disease. METHODS: We report the first two children affected by severe MKD who received haploidentical α/ß T-cell and B-cell depleted SCT. Both patients received a treosulfan-based conditioning regimen and one received a second haploidentical-SCT for secondary rejection of the first. RESULTS: Both patients obtained a stable full donor engraftment with a complete regression of clinical and biochemical inflammatory signs, without acute organ toxicity or acute and chronic GvHD. In both, the urinary excretion of mevalonic acid remained high post-transplant in the absence of any inflammatory signs. CONCLUSION: Haploidentical α/ß T-cell and B-cell depleted SCT represents a potential curative strategy in patients affected by MKD. The persistence of urinary excretion of mevalonic acid after SCT, probably related to the ubiquitous expression of MVK enzyme, suggests that these patients should be carefully monitored after SCT to exclude MKD clinical recurrence. Prophylaxis with anakinra in the acute phase after transplant could represent a safe and effective approach. Further biological studies are required to clarify the pathophysiology of inflammatory attacks in MKD in order to better define the therapeutic role of SCT.


Asunto(s)
Deficiencia de Mevalonato Quinasa/terapia , Trasplante de Células Madre/métodos , Trasplante Haploidéntico/métodos , Enfermedad Aguda , Linfocitos B/trasplante , Femenino , Humanos , Recién Nacido , Masculino , Linfocitos T/trasplante
3.
Public Health Nurs ; 36(4): 541-544, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30945355

RESUMEN

Newborn screening (NBS) is a public health program that detects genetic conditions in neonates enabling treatment before clinical symptoms manifest. Severe combined immune deficiency (SCID) is a primary immune deficiency found in the absence of functioning T and B lymphocytes. Hematopoietic cell transplantation is a potentially curative treatment if received within the first 42 months of life; without treatment, this condition is fatal in the first 2 years of life due to severe opportunistic infections. SCID was added to the recommended uniform panel of conditions for inclusion in state NBS programs in 2010. This manuscript examines the societal costs and benefits of NBS for SCID in Arkansas and implications to health services and social welfare. Total cost per year of all NBS for SCID and resulting early treatment for one patient with SCID in Arkansas is estimated at $1,078,714. Cost of late treatment of one patient with SCID is estimated at $1.43 million. Based on an expected diagnosis of one patient per year in Arkansas, this results in an estimated net cost savings for NBS for SCID in Arkansas of $351,286 per year. Based on cost-effectiveness analysis, NBS for SCID in Arkansas is cost-effective, with higher societal benefit than cost.


Asunto(s)
Análisis Costo-Beneficio , Pruebas Genéticas/métodos , Tamizaje Neonatal/economía , Tamizaje Neonatal/métodos , Inmunodeficiencia Combinada Grave/diagnóstico , Arkansas , Linfocitos B/inmunología , Linfocitos B/trasplante , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Recién Nacido , Inmunodeficiencia Combinada Grave/terapia , Linfocitos T/inmunología , Linfocitos T/trasplante
4.
Mol Med ; 24(1): 26, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-30134811

RESUMEN

BACKGROUND: Sepsis morbidity and mortality are aggravated by acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). Mouse B-1a cells are a phenotypically and functionally unique sub-population of B cells, providing immediate protection against infection by releasing natural antibodies and immunomodulatory molecules. We hypothesize that B-1a cells ameliorate sepsis-induced ALI. METHODS: Sepsis was induced in C57BL/6 mice by cecal ligation and puncture (CLP). PBS or B-1a cells were adoptively transferred into the septic mice intraperitoneally. After 20 h of CLP, lungs were harvested and assessed by PCR and ELISA for pro-inflammatory cytokines (IL-6, IL-1ß) and chemokine (MIP-2) expression, by histology for injury, by TUNEL and cleaved caspase-3 for apoptosis, and by myeloperoxidase (MPO) assay for neutrophil infiltration. RESULTS: We found that septic mice adoptively transferred with B-1a cells significantly decreased the mRNA and protein levels of IL-6, IL-1ß and MIP-2 in the lungs compared to PBS-treated mice. Mice treated with B-1a cells showed dramatic improvement in lung injury compared to PBS-treated mice after sepsis. We found apoptosis in the lungs was significantly inhibited in B-1a cell injected mice compared to PBS-treated mice after sepsis. B-1a cell treatment significantly down-regulated MPO levels in the lungs compared to PBS-treated mice in sepsis. The protective outcomes of B-1a cells in ALI was further confirmed by using B-1a cell deficient CD19-/- mice, which showed significant increase in the lung injury scores following sepsis as compared to WT mice. CONCLUSIONS: Our results demonstrate a novel therapeutic potential of B-1a cells to treat sepsis-induced ALI.


Asunto(s)
Lesión Pulmonar Aguda/terapia , Linfocitos B/trasplante , Sepsis/terapia , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Traslado Adoptivo , Animales , Citocinas/inmunología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Peroxidasa/inmunología , Sepsis/complicaciones , Sepsis/inmunología , Sepsis/patología
5.
Eur J Immunol ; 47(2): 269-279, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27873323

RESUMEN

Vaccination with antigen-pulsed CD40-activated B (CD40-B) cells can efficiently lead to the in vivo differentiation of naive CD8+ T cells into fully functional effectors. In contrast to bone marrow-derived dendritic cell (BMDC) vaccination, CD40-B cell priming does not allow for memory CD8+ T-cell generation but the reason for this deficiency is unknown. Here, we show that compared to BMDCs, murine CD40-B cells induce lower expression of several genes regulated by T-cell receptor signaling, costimulation, and inflammation (signals 1-3) in mouse T cells. The reduced provision of signals 1 and 2 by CD40-B cells can be explained by a reduction in the quality and duration of the interactions with naive CD8+ T cells as compared to BMDCs. Furthermore, CD40-B cells produce less inflammatory mediators, such as IL-12 and type I interferon, and increasing inflammation by coadministration of polyriboinosinic-polyribocytidylic acid with CD40-B-cell immunization allowed for the generation of long-lived and functional CD8+ memory T cells. In conclusion, it is possible to manipulate CD40-B-cell vaccination to promote the formation of long-lived functional CD8+ memory T cells, a key step before translating the use of CD40-B cells for therapeutic vaccination.


Asunto(s)
Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Linfocitos T CD8-positivos/inmunología , Inflamación/inmunología , Polinucleótidos/administración & dosificación , Animales , Linfocitos B/trasplante , Antígenos CD40/metabolismo , Ligando de CD40/genética , Ligando de CD40/metabolismo , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Fibroblastos/inmunología , Fibroblastos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Memoria Inmunológica , Interleucina-4/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Poli I-C , Vacunación
6.
J Immunol ; 197(9): 3628-3638, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27647833

RESUMEN

Elucidation of the immune requirements for control or elimination of retroviral infection remains an important aim. We studied the induction of adaptive immunity to neonatal infection with a murine retrovirus, under conditions leading to immunological tolerance. We found that the absence of either maternal or offspring adaptive immunity permitted efficient vertical transmission of the retrovirus. Maternal immunodeficiency allowed the retrovirus to induce central Th cell tolerance in the infected offspring. In turn, this compromised the offspring's ability to mount a protective Th cell-dependent B cell response. However, in contrast to T cells, offspring B cells were not centrally tolerized and retained their ability to respond to the infection when provided with T cell help. Thus, escape of retrovirus-specific B cells from deletional tolerance offers the opportunity to induce protective retroviral immunity by restoration of retrovirus-specific T cell help, suggesting similar T cell immunotherapies for persistent viral infections.


Asunto(s)
Traslado Adoptivo , Linfocitos B/inmunología , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Virus de la Leucemia Murina/inmunología , Leucemia Experimental/prevención & control , Infecciones por Retroviridae/prevención & control , Linfocitos T/inmunología , Infecciones Tumorales por Virus/prevención & control , Animales , Animales Recién Nacidos , Linfocitos B/trasplante , Linfocitos B/virología , Células Cultivadas , Tolerancia Central , Femenino , Leucemia Experimental/inmunología , Masculino , Exposición Materna/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/transmisión , Linfocitos T/trasplante , Linfocitos T/virología , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/transmisión
7.
J Immunol ; 196(10): 4172-84, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27053762

RESUMEN

Inactivated influenza vaccines have two formulations, whole- and split-virion types; however, how differential formulations impact their booster effects remain unknown. In this study, we demonstrate that whole-virion vaccines recall two waves of Ab responses, early T cell-independent (TI) and late T cell-dependent responses, whereas split-virion vaccines elicit the late T cell-dependent response only. Notably, higher-affinity Abs with improved neutralizing activity are provided from the early TI response, which emphasizes the important contribution of the formulation-dependent response in the protective immunity. Moreover, we show that the early TI response completely requires B cell-intrinsic TLR7 signaling, which can be delivered through viral RNAs within whole-virion vaccine. Thus, our results indicate that TLR agonists in whole-virion type improve recall Ab responses by directly targeting memory B cells, a finding with important implications for vaccine strategies aimed at the prompt recall of high-affinity neutralizing Abs.


Asunto(s)
Linfocitos B/inmunología , Vacunas contra la Influenza/inmunología , ARN Viral/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Virión/inmunología , Animales , Anticuerpos Antivirales/metabolismo , Afinidad de Anticuerpos , Linfocitos B/trasplante , Células Cultivadas , Humanos , Inmunidad Humoral , Memoria Inmunológica , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/metabolismo , Vacunación
8.
Immunology ; 152(2): 276-286, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28557002

RESUMEN

The mammalian target of rapamycin (mTOR) is a serine-threonine kinase that has been shown to be essential for the differentiation and function of various immune cells. Earlier in vitro studies showed that mTOR signalling regulates B-cell biology by supporting their activation and proliferation. However, how mTOR signalling temporally regulates in vivo germinal centre B (GCB) cell development and differentiation into short-lived plasma cells, long-lived plasma cells and memory cells is still not well understood. In this study, we used a combined conditional/inducible knock-out system to investigate the temporal regulation of mTOR complex 1 (mTORC1) in the GCB cell response to acute lymphocytic choriomeningitis virus infection by deleting Raptor, a main component of mTORC1, specifically in B cells in pre- and late GC phase. Early Raptor deficiency strongly inhibited GCB cell proliferation and differentiation and plasma cell differentiation. Nevertheless, late GC Raptor deficiency caused only decreases in the size of memory B cells and long-lived plasma cells through poor maintenance of GCB cells, but it did not change their differentiation. Collectively, our data revealed that mTORC1 signalling supports GCB cell responses at both early and late GC phases during viral infection but does not regulate GCB cell differentiation into memory B cells and plasma cells at the late GC stage.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Linfocitos B/enzimología , Centro Germinal/enzimología , Coriomeningitis Linfocítica/enzimología , Virus de la Coriomeningitis Linfocítica/inmunología , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/trasplante , Linfocitos B/virología , Trasplante de Médula Ósea , Diferenciación Celular , Proliferación Celular , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Centro Germinal/inmunología , Centro Germinal/virología , Interacciones Huésped-Patógeno , Inmunidad Humoral , Memoria Inmunológica , Activación de Linfocitos , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/deficiencia , Complejos Multiproteicos/genética , Complejos Multiproteicos/inmunología , Fenotipo , Células Plasmáticas/enzimología , Células Plasmáticas/inmunología , Células Plasmáticas/virología , Proteína Reguladora Asociada a mTOR , Transducción de Señal , Serina-Treonina Quinasas TOR/deficiencia , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Factores de Tiempo , Quimera por Trasplante
9.
Int J Cancer ; 140(6): 1356-1363, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-27935045

RESUMEN

Patient-derived cancer xenografts (PDX) are widely used to identify and evaluate novel therapeutic targets, and to test therapeutic approaches in preclinical mouse avatar trials. Despite their widespread use, potential caveats of PDX models remain considerably underappreciated. Here, we demonstrate that EBV-associated B-lymphoproliferations frequently develop following xenotransplantation of human colorectal and pancreatic carcinomas in highly immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl /SzJ (NSG) mice (18/47 and 4/37 mice, respectively), and in derived cell cultures in vitro. Strikingly, even PDX with carcinoma histology can host scarce EBV-infected B-lymphocytes that can fully overgrow carcinoma cells during serial passaging in vitro and in vivo. As serial xenografting is crucial to expand primary tumor tissue for biobanks and cohorts for preclinical mouse avatar trials, the emerging dominance of B-lymphoproliferations in serial PDX represents a serious confounding factor in these models. Consequently, repeated phenotypic assessments of serial PDX are mandatory at each expansion step to verify "bona fide" carcinoma xenografts.


Asunto(s)
Linfocitos B/trasplante , Carcinoma Ductal Pancreático/patología , Neoplasias Colorrectales/patología , Infecciones por Virus de Epstein-Barr/patología , Trastornos Linfoproliferativos/etiología , Neoplasias Pancreáticas/patología , Ensayo de Capsula Subrrenal , Animales , Antígenos de Neoplasias/análisis , Linfocitos B/patología , Linfocitos B/virología , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/virología , División Celular , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/virología , Medio de Cultivo Libre de Suero , Infecciones por Virus de Epstein-Barr/inmunología , Xenoinjertos/inmunología , Xenoinjertos/patología , Humanos , Huésped Inmunocomprometido , Antígenos Comunes de Leucocito/análisis , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/virología , Ratones , Ratones Endogámicos NOD , Especificidad de Órganos , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/virología , Esferoides Celulares , Ensayo de Capsula Subrrenal/métodos
10.
Eur J Immunol ; 46(12): 2719-2729, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27701733

RESUMEN

The contribution of B cells to immunity against many infectious diseases is unquestionably important and well characterized. Here, we sought to determine the role of B cells in the induction of T-helper 1 (TH 1) CD4+ T cells upon vaccination with a tuberculosis (TB) antigen combined with a TLR4 agonist. We used B-cell deficient mice (µMT-/- ), tetramer-positive CD4+ T cells, markers of memory "precursor" effector cells (MPECs), and T-cell adoptive transfers and demonstrated that the early antigen-specific cytokine-producing TH 1 responses are unaffected in the absence of B cells, however MPEC induction is strongly impaired resulting in a deficiency of the memory TH 1 response in µMT-/- mice. We further show that antigen-presentation by B cells is necessary for their role in MPEC generation using B-cell adoptive transfers from wt or MHC class II knock-out mice into µMT-/- mice. Our study challenges the view that B-cell deficiency exclusively alters the TH 1 response at memory time-points. Collectively, our results provide new insights on the multifaceted roles of B cells that will have a high impact on vaccine development against several pathogens including those requiring TH 1 cell-mediated immunity.


Asunto(s)
Presentación de Antígeno , Linfocitos B/fisiología , Factores Inmunológicos/inmunología , Subgrupos de Linfocitos T/inmunología , Células TH1/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/inmunología , Traslado Adoptivo , Animales , Linfocitos B/trasplante , Diferenciación Celular , Células Cultivadas , Humanos , Cadenas mu de Inmunoglobulina/genética , Memoria Inmunológica , Activación de Linfocitos , Ratones , Ratones Noqueados , Subgrupos de Linfocitos T/trasplante , Receptor Toll-Like 4/agonistas , Tuberculosis/prevención & control
11.
J Virol ; 90(7): 3439-45, 2016 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-26764000

RESUMEN

UNLABELLED: Previous experiments carried out in a sheep scrapie model demonstrated that the transfusion of 200 µl of prion-infected whole blood has an apparent 100% efficacy for disease transmission. These experiments also indicated that, despite the apparent low infectious titer, the intravenous administration of white blood cells (WBC) resulted in efficient disease transmission. In the study presented here, using the same transmissible spongiform encephalopathy (TSE) animal model, our aim was to determine the minimal number of white blood cells and the specific abilities of mononucleated cell populations to transmit scrapie by the transfusion route. Our results confirmed that the transfusion of 100 µl, but not 10 µl, of fresh whole blood collected in asymptomatic scrapie-infected donor sheep can transmit the disease. The data also show that the intravenous administration of 10(5) WBCs is sufficient to cause scrapie in recipient sheep. Cell-sorted CD45R(+) (predominantly B lymphocytes), CD4(+)/CD8(+) (T lymphocytes), and CD14(+) (monocytes/macrophages) blood cell subpopulations all were shown to contain prion infectivity by bioassays in ovine PrP transgenic mice. However, while the intravenous administration of 10(6) CD45(+) or CD4(+)/8(+) living cells was able to transmit the disease, similar numbers of CD14(+) cells failed to infect the recipients. These data support the contention that mononucleated blood cell populations display different abilities to transmit TSE by the transfusion route. They also represent an important input for the risk assessment of blood-borne prion disease transmission and for refining the target performance of leukoreduction processes that currently are applied to mitigate the transmission risk in transfusion medicine. IMPORTANCE: Interindividual variant Creutzfeldt-Jakob disease (vCJD) transmission through blood and blood-derived products is considered a major public health issue in transfusion medicine. Over the last decade, TSE in sheep has emerged as a relevant model for assessing the blood-borne vCJD transmission risk. In this study, using a sheep TSE model, we characterized the ability of different peripheral blood mononucleated cell populations to infect TSE-free recipients by the transfusion route. Our results indicate that as little as 10(5) WBC and 100 µl of blood collected from asymptomatic scrapie infected animals can transmit the disease. They also demonstrate unambiguously that peripheral blood mononuclear cell subpopulations display dramatically different abilities to transmit the disease. These data represent an important input for the risk assessment of blood-borne prion disease transmission and for refining the target performance of leukoreduction processes that currently are applied to mitigate the transmission risk in transfusion medicine.


Asunto(s)
Leucocitos Mononucleares/trasplante , Scrapie/sangre , Scrapie/transmisión , Reacción a la Transfusión , Animales , Linfocitos B/trasplante , Síndrome de Creutzfeldt-Jakob/sangre , Síndrome de Creutzfeldt-Jakob/transmisión , Modelos Animales de Enfermedad , Macrófagos/trasplante , Ratones , Ovinos , Linfocitos T/trasplante
12.
J Immunol ; 195(6): 2900-7, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26268652

RESUMEN

Emerging evidence suggests a role of B cells in host defense against primary pulmonary tuberculosis (TB). However, the role of B cells in TB vaccine-induced protective T cell immunity still remains unknown. Using a viral-vectored model TB vaccine and a number of experimental approaches, we have investigated the role of B cells in respiratory mucosal vaccine-induced T cell responses and protection against pulmonary TB. We found that respiratory mucosal vaccination activated Ag-specific B cell responses. Whereas respiratory mucosal vaccination elicited Ag-specific T cell responses in the airway and lung interstitium of genetic B cell-deficient (Jh(-/-) knockout [KO]) mice, the levels of airway T cell responses were lower than in wild-type hosts, which were associated with suboptimal protection against pulmonary Mycobacterium tuberculosis challenge. However, mucosal vaccination induced T cell responses in the airway and lung interstitium and protection in B cell-depleted wild-type mice to a similar extent as in B cell-competent hosts. Furthermore, by using an adoptive cell transfer approach, reconstitution of B cells in vaccinated Jh(-/-) KO mice did not enhance anti-TB protection. Moreover, respiratory mucosal vaccine-activated T cells alone were able to enhance anti-TB protection in SCID mice, and the transfer of vaccine-primed B cells alongside T cells did not further enhance such protection. Alternatively, adoptively transferring vaccine-primed T cells from Jh(-/-) KO mice into SCID mice only provided suboptimal protection. These data together suggest that B cells play a minimal role, and highlight a central role by T cells, in respiratory mucosal vaccine-induced protective immunity against M. tuberculosis.


Asunto(s)
Aciltransferasas/inmunología , Antígenos Bacterianos/inmunología , Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Traslado Adoptivo , Animales , Linfocitos B/trasplante , Femenino , Inmunidad Mucosa/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , Mycobacterium tuberculosis/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/microbiología , Tuberculosis Pulmonar/prevención & control , Vacunación
13.
J Immunol ; 194(6): 2607-15, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25672753

RESUMEN

Autophagy is required for the long-term maintenance of Ag-specific memory B cells. However, whether autophagy is also important for the initial formation of memory B cells remains unclear. In this study, we show that newly generated memory B cells do not display active autophagy but are capable of forming Ab-secreting cells after rechallenge with Ags. Increases in autophagy took place over time after the initial formation of memory B cells. The expression of transcription factors involved in autophagy, but not changes in epigenetic regulation by DNA methylation, was required for autophagy gene expression and the development of active autophagy in memory B cells. This indicates that autophagy is not critical for the initial generation of memory B cells but is required for their long-term persistence. Our results suggest that promoting autophagy to improve Ab-dependent immunological memory is more effective during memory B cell maintenance stage.


Asunto(s)
Autofagia/inmunología , Linfocitos B/inmunología , Inmunización/métodos , Memoria Inmunológica/inmunología , Traslado Adoptivo , Animales , Autofagia/genética , Proteína 7 Relacionada con la Autofagia , Linfocitos B/metabolismo , Linfocitos B/trasplante , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Femenino , Citometría de Flujo , Expresión Génica/inmunología , Haptenos , Hemocianinas/inmunología , Inmunohistoquímica , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/inmunología
14.
Immunology ; 147(1): 97-113, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26456931

RESUMEN

We describe a protective early acquired immune response to pneumococcal pneumonia that is mediated by a subset of B1a cells. Mice deficient in B1 cells (xid), or activation-induced cytidine deaminase (AID(-/-) ), or invariant natural killer T (iNKT) cells (Jα18(-/-) ), or interleukin-13 (IL-13(-/-) ) had impaired early clearance of pneumococci in the lung, compared with wild-type mice. In contrast, AID(-/-) mice adoptively transferred with AID(+/+) B1a cells, significantly cleared bacteria from the lungs as early as 3 days post infection. We show that this early bacterial clearance corresponds to an allergic contact sensitivity-like cutaneous response, probably due to a subpopulation of initiating B1a cells. In the pneumonia model, these B1a cells were found to secrete higher affinity antigen-specific IgM. In addition, as in contact sensitivity, iNKT cells were required for the anti-pneumococcal B1a cell initiating response, probably through early production of IL-13, given that IL-13(-/-) mice also failed to clear infection. Our study is the first to demonstrate the importance of AID in generating an appropriate B1a cell response to pathogenic bacteria. Given the antibody affinity and pneumonia resistance data, natural IgM produced by conventional B1a cells are not responsible for pneumonia clearance compared with the AID-dependent subset.


Asunto(s)
Inmunidad Adaptativa , Linfocitos B/enzimología , Citidina Desaminasa/metabolismo , Pulmón/enzimología , Fagocitosis , Neumonía Neumocócica/enzimología , Streptococcus pneumoniae/inmunología , Traslado Adoptivo , Agammaglobulinemia Tirosina Quinasa , Animales , Antígenos Bacterianos/inmunología , Linfocitos B/inmunología , Linfocitos B/microbiología , Linfocitos B/trasplante , Activación de Complemento , Citidina Desaminasa/deficiencia , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citocinas/inmunología , Citocinas/metabolismo , Dermatitis por Contacto/enzimología , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/microbiología , Modelos Animales de Enfermedad , Genotipo , Inmunoglobulina M/inmunología , Inmunoglobulina M/metabolismo , Interleucina-13/deficiencia , Interleucina-13/genética , Pulmón/inmunología , Pulmón/microbiología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/microbiología , Fenotipo , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/microbiología , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Bazo/enzimología , Bazo/inmunología , Bazo/microbiología , Streptococcus pneumoniae/patogenicidad , Factores de Tiempo
15.
BMC Cancer ; 16: 269, 2016 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-27067989

RESUMEN

BACKGROUND: Interrogate the impact of IKZF1 deletion on therapy-outcomes of adults with common B-cell acute lymphoblastic leukemia. METHODS: One hundred sixty-five consecutive adults with common B-cell ALL were tested for IKZF1 deletion and for BCR/ABL. Deletions in IKZF1 were detected using multiplex RQ-PCR, multiplex fluorescent PCR, sequence analysis and multiplex ligation-dependent probe amplification (MLPA). BCR/ABL was detected using RQ-PCR. All subjects received chemotherapy and some also received an allotransplant and tyrosine kinase-inhibitors. Multivariate analyses were done to identify associations between IKZF1 deletion and other variables on non-relapse mortality (NRM), cumulative incidence of relapse (CIR), leukemia-free survival (LFS) and survival. RESULTS: Amongst subjects achieving complete remission those with IKZF1 deletion had similar 5-year non-relapse mortality (NRM) (11% [2-20%] vs. 16% [4-28%]; P = 0.736), a higher 5-year cumulative incidence of relapse (CIR) (55% [35-76%] vs. 25% [12-38%]; P = 0.004), and worse 5-year leukemia-free survival (LFS) (33% [16-52%] vs. 59% [42-73%]; P = 0.012) and survival (48% [33-62%] vs. 75% [57-86%]; P = 0.002). In multivariate analyses IKZF1 deletion was associated with an increased relapse (relative risk [RR] =2.7, [1.4-5.2]; P = 0.002), a higher risk of treatment-failure (inverse of LFS; RR = 2.1, [1.2-3.6]; P = 0.007) and a higher risk of death (RR = 2.8, [1.5-5.5]; P = 0.002). The adverse impact of IKZF1 deletion on outcomes was stronger in subjects without vs. with BCR-ABL1 and in subjects receiving chemotherapy-only vs. an allotransplant. CONCLUSIONS: IKZF1 deletion was independently-associated with a higher relapse risk and worse LFS and survival in adults with common B-cell ALL after adjusting for other prognostic variables and differences in therapies. These data suggest IKZF1 deletion may be a useful prognostic variable in adults with common B-cell ALL, especially in persons without BCR-ABL1 and those receiving chemotherapy-only. Transplants appear to overcome the adverse impact of IKZF1 deletion on therapy-outcomes but confirmation in a randomized study is needed. The trial was registered in 2007 with the Beijing Municipal Government (Beijing Municipal Health Bureau Registration N: 2007-1007).


Asunto(s)
Proteínas de Fusión bcr-abl/genética , Factor de Transcripción Ikaros/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Trasplante Homólogo , Adolescente , Adulto , Anciano , Linfocitos B/patología , Linfocitos B/trasplante , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico , Eliminación de Secuencia
16.
J Immunol ; 193(6): 2753-63, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25108021

RESUMEN

Immune cells regulate cell surface receptor expression during their maturation, activation, and motility. Although many of these receptors are regulated largely at the level of expression, protease-mediated ectodomain shedding represents an alternative means of refashioning the surface of immune cells. Shedding is largely attributed to a family of a disintegrin and metalloprotease domain (ADAM) metalloproteases, including ADAM17. Although ADAM17 is well known to contribute to the innate immune response, mainly by releasing TNF-α, much less is known about whether/how this metalloprotease regulates adaptive immunity. To determine whether ADAM17 contributes to regulating adaptive immune responses, we took advantage of ADAM17 hypomorphic (ADAM17(ex/ex)) mice, in which ADAM17 expression is reduced by 90-95% compared with wild-type littermates. In this study, we show that that ADAM17 deficiency results in spleen and lymph node enlargement, as well as increased levels of Ag-specific class-switched Ig production following immunization with OVA together with anti-CD40 mAbs and polyinosinic-polycytidylic acid. Moreover, we demonstrate that the costimulatory ligand ICOS ligand (ICOSL) is selectively downregulated on the surface of B cells in an ADAM17-specific manner, although it is not proteolitically processed by recombinant ADAM17 in vitro. Finally, we show that higher cell surface levels of ICOSL in ADAM17(ex/ex) mice may contribute to the development of excessive Ab responses. Therefore, our data suggest a functional link between ADAM17 and ICOSL in controlling adaptive immune responses.


Asunto(s)
Proteínas ADAM/inmunología , Linfocitos B/inmunología , Inmunidad Humoral , Ligando Coestimulador de Linfocitos T Inducibles/inmunología , Proteínas ADAM/genética , Proteína ADAM17 , Animales , Anticuerpos Monoclonales/inmunología , Formación de Anticuerpos/inmunología , Linfocitos B/trasplante , Antígenos CD40/inmunología , Células Cultivadas , Femenino , Cambio de Clase de Inmunoglobulina , Isotipos de Inmunoglobulinas/biosíntesis , Isotipos de Inmunoglobulinas/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/biosíntesis , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/inmunología , Poli I-C/inmunología , Bazo/patología
17.
J Immunol ; 193(11): 5637-48, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25339664

RESUMEN

Many metabolic diseases, including atherosclerosis, type 2 diabetes, pulmonary alveolar proteinosis, and obesity, have a chronic inflammatory component involving both innate and adaptive immunity. Mice lacking the ATP-binding cassette transporter G1 (ABCG1) develop chronic inflammation in the lungs, which is associated with the lipid accumulation (cholesterol, cholesterol ester, and phospholipid) and cholesterol crystal deposition that are characteristic of atherosclerotic lesions and pulmonary alveolar proteinosis. In this article, we demonstrate that specific lipids, likely oxidized phospholipids and/or sterols, elicit a lung-specific immune response in Abcg1(-/-) mice. Loss of ABCG1 results in increased levels of specific oxysterols, phosphatidylcholines, and oxidized phospholipids, including 1-palmitoyl-2-(5'-oxovaleroyl)-sn-glycero-3-phosphocholine, in the lungs. Further, we identify a niche-specific increase in natural Ab (NAb)-secreting B-1 B cells in response to this lipid accumulation that is paralleled by increased titers of IgM, IgA, and IgG against oxidation-specific epitopes, such as those on oxidized low-density lipoprotein and malondialdehyde-modified low-density lipoprotein. Finally, we identify a cytokine/chemokine signature that is reflective of increased B cell activation, Ab secretion, and homing. Collectively, these data demonstrate that the accumulation of lipids in Abcg1(-/-) mice induces the specific expansion and localization of B-1 B cells, which secrete NAbs that may help to protect against the development of atherosclerosis. Indeed, despite chronic lipid accumulation and inflammation, hyperlipidemic mice lacking ABCG1 develop smaller atherosclerotic lesions compared with controls. These data also suggest that Abcg1(-/-) mice may represent a new model in which to study the protective functions of B-1 B cells/NAbs and suggest novel targets for pharmacologic intervention and treatment of disease.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Aterosclerosis/inmunología , Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Lipoproteínas/metabolismo , Pulmón/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Traslado Adoptivo , Animales , Anticuerpos/metabolismo , Proteínas Aviares/metabolismo , Subgrupos de Linfocitos B/trasplante , Linfocitos B/trasplante , Células Cultivadas , Citocinas/metabolismo , Perfilación de la Expresión Génica , Homeostasis/genética , Errores Innatos del Metabolismo Lipídico/genética , Lipoproteínas/genética , Pulmón/inmunología , Activación de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Fosfolípidos/metabolismo
18.
Infect Immun ; 83(2): 743-58, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25452554

RESUMEN

HIV infection results in a complex immunodeficiency due to loss of CD4(+) T cells, impaired type I interferon (IFN) responses, and B cell dysfunctions causing susceptibility to opportunistic infections such as Pneumocystis murina pneumonia and unexplained comorbidities, including bone marrow dysfunctions. Type I IFNs and B cells critically contribute to immunity to Pneumocystis lung infection. We recently also identified B cells as supporters of on-demand hematopoiesis following Pneumocystis infection that would otherwise be hampered due to systemic immune effects initiated in the context of a defective type I IFN system. While studying the role of type I IFNs in immunity to Pneumocystis infection, we discovered that mice lacking both lymphocytes and type I IFN receptor (IFrag(-/-)) developed progressive bone marrow failure following infection, while lymphocyte-competent type I IFN receptor-deficient mice (IFNAR(-/-)) showed transient bone marrow depression and extramedullary hematopoiesis. Lymphocyte reconstitution of lymphocyte-deficient IFrag(-/-) mice pointed to B cells as a key player in bone marrow protection. Here we define how B cells protect on-demand hematopoiesis following Pneumocystis lung infection in our model. We demonstrate that adoptive transfer of B cells into IFrag(-/-) mice protects early hematopoietic progenitor activity during systemic responses to Pneumocystis infection, thus promoting replenishment of depleted bone marrow cells. This activity is independent of CD4(+) T cell help and B cell receptor specificity and does not require B cell migration to bone marrow. Furthermore, we show that B cells protect on-demand hematopoiesis in part by induction of interleukin-10 (IL-10)- and IL-27-mediated mechanisms. Thus, our data demonstrate an important immune modulatory role of B cells during Pneumocystis lung infection that complement the modulatory role of type I IFNs to prevent systemic complications.


Asunto(s)
Linfocitos B/inmunología , Hematopoyesis/inmunología , Pneumocystis/inmunología , Neumonía por Pneumocystis/inmunología , Receptor de Interferón alfa y beta/genética , Traslado Adoptivo , Anemia Aplásica , Animales , Linfocitos B/trasplante , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Linfocitos T CD4-Positivos/inmunología , Hemoglobinuria Paroxística/inmunología , Hemoglobinuria Paroxística/microbiología , Interferón Tipo I/genética , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Interleucinas/biosíntesis , Interleucinas/inmunología , Pulmón/inmunología , Pulmón/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID
19.
Eur J Immunol ; 44(11): 3453-63, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25154613

RESUMEN

NOD/LtSzscid/IL-2Rγ(-/-) (NSG) mice have advantages in establishing humanized mouse models. However, transferring human PBMCs into these mice often causes lethal GVH disease. In this study, we discovered an improved method for the engraftment of normal or pathological human PBMCs into NSG mice and examined the subsequent induction of specific immune responses. We sequentially transferred human CD4+ memory T (Tm) and B cells obtained from PBMCs of healthy adults or patients with autoimmune diseases into NSG mice. Removing naïve CD4+ T cells from the transferred PBMCs allowed successful engraftment without lethal GVH disease. The transferred Tm cells were found to reside mainly in the spleen and the lymphoid nodules, where they expressed MHC class II molecules and produced cytokines, including IL-21. Surprisingly, the transferred B cells were also well maintained in the lymphoid organs, underwent de novo class-switch recombination, and secreted all isotypes of human Igs at significant levels. Moreover, transferring patient-derived Tm and B cells resulted in sustained production of IgM-rheumatoid factor and antiaminoacyl transfer RNA synthetase Abs in these mice. These results suggest that transfer of Tm and B cells derived from human PBMCs into NSG mice could be a useful method for the study of human autoimmune mechanisms.


Asunto(s)
Autoanticuerpos/biosíntesis , Linfocitos B/trasplante , Linfocitos T CD4-Positivos/trasplante , Trasplante Heterólogo/métodos , Animales , Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Cambio de Clase de Inmunoglobulina/genética , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Interleucinas/biosíntesis , Antígenos Comunes de Leucocito/biosíntesis , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Monocitos/inmunología , Monocitos/trasplante
20.
Cell Immunol ; 296(2): 133-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25959608

RESUMEN

Tumor necrosis factor (TNF) is a key cytokine in rheumatoid arthritis (RA) pathogenesis, as underscored by the clinical effectiveness of TNF antagonists. While several of TNF's key targets in RA are well understood, its many pleiotropic effects remain to be elucidated. TNF-transgenic mice develop inflammatory-erosive arthritis associated with disruption of draining lymph node histology and function, and accumulation of B cells with unique phenotypic and functional features consistent with contribution to pathogenesis (B cells in inflamed nodes, Bin). Bin cell induction depends on the inflamed microenvironment, but the specific signals are unknown. Using anti-TNF treatment and TNF-receptor-deficient mice, here we show that Bin cells are induced and maintained independently of B cell-intrinsic TNF signals.


Asunto(s)
Anticuerpos/farmacología , Artritis Experimental/inmunología , Linfocitos B/inmunología , Ganglios Linfáticos/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Traslado Adoptivo , Animales , Antígenos CD1d/genética , Antígenos CD1d/inmunología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/genética , Artritis Experimental/patología , Artritis Reumatoide/genética , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Linfocitos B/trasplante , Microambiente Celular/inmunología , Expresión Génica , Humanos , Inmunofenotipificación , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Complemento 3b/genética , Receptores de Complemento 3b/inmunología , Receptores de Complemento 3d/genética , Receptores de Complemento 3d/inmunología , Receptores de IgE/genética , Receptores de IgE/inmunología , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA