Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 437
Filtrar
1.
Comput Math Methods Med ; 2022: 8361665, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35047059

RESUMEN

OBJECTIVE: To explore the effect of autoimmune cell therapy on immune cells in patients with chronic obstructive pulmonary disease (COPD) and to provide a reference for clinical treatment of COPD. METHODS: Sixty patients with stable COPD were randomly divided into control group and treatment group (n = 30). The control group was given conventional treatment, and the treatment group was given one autoimmune cell therapy on the basis of conventional treatment. The serum levels of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the peripheral blood were detected by flow cytometry. Possible adverse reactions were detected at any time during treatment. RESULTS: There were no significant differences in the contents of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the serum of the control group (P > 0.05). Compared with before treatment, the contents of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the serum of the treatment group were significantly increased (P < 0.05). The ratio of CD4 + /CD8+ T cells in both control and treatment groups did not change significantly during treatment (P > 0.05). There were no significant differences in serum CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the treatment group at 30 days and 90 days after treatment (P > 0.05), but they were significantly higher than those in the control group (P < 0.05). CONCLUSION: Autoimmune cell therapy can significantly increase the level of immune cells in the body and can be maintained for a long period of time, which has certain clinical benefits for recurrent respiratory tract infections and acute exacerbation in patients with COPD.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/terapia , Anciano , Anciano de 80 o más Años , Autoinmunidad , Linfocitos B/inmunología , Linfocitos B/trasplante , Transfusión de Sangre Autóloga/métodos , Transfusión de Sangre Autóloga/estadística & datos numéricos , Tratamiento Basado en Trasplante de Células y Tejidos/estadística & datos numéricos , Biología Computacional , Femenino , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/trasplante , Transfusión de Leucocitos/métodos , Transfusión de Leucocitos/estadística & datos numéricos , Masculino , Persona de Mediana Edad , Enfermedad Pulmonar Obstructiva Crónica/sangre , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/trasplante
2.
Front Immunol ; 12: 730471, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34566992

RESUMEN

The ligand-binding surface of the B cell receptor (BCR) is formed by encoded and non-encoded antigen complementarity determining regions (CDRs). Genetically reproducible or 'public' antibodies can arise when the encoded CDRs play deterministic roles in antigen recognition, notably within human broadly neutralizing antibodies against HIV and influenza virus. We sought to exploit this by engineering virus-like-particle (VLP) vaccines that harbor multivalent affinity against gene-encoded moieties of the BCR antigen binding site. As proof of concept, we deployed a library of RNA bacteriophage VLPs displaying random peptides to identify a multivalent antigen that selectively triggered germline BCRs using the human VH gene IGVH1-2*02. This VLP selectively primed IGHV1-2*02 BCRs that were present within a highly diversified germline antibody repertoire within humanized mice. Our approach thus provides methodology to generate antigens that engage specific BCR configurations of interest, in the absence of structure-based information.


Asunto(s)
Linfocitos B/inmunología , Ingeniería de Proteínas , Fagos ARN/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Anticuerpos de Dominio Único/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Traslado Adoptivo , Animales , Especificidad de Anticuerpos , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/trasplante , Femenino , Biblioteca de Genes , Humanos , Ligandos , Masculino , Ratones Transgénicos , Prueba de Estudio Conceptual , Fagos ARN/genética , Fagos ARN/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Anticuerpos de Dominio Único/administración & dosificación , Anticuerpos de Dominio Único/genética , Anticuerpos de Dominio Único/metabolismo , Vacunación , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/metabolismo
3.
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
4.
Nat Commun ; 11(1): 5851, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33203857

RESUMEN

HIV viremia can be controlled by chronic antiretroviral therapy. As a potentially single-shot alternative, B cells engineered by CRISPR/Cas9 to express anti-HIV broadly neutralizing antibodies (bNAbs) are capable of secreting high antibody titers. Here, we show that, upon immunization of mice, adoptively transferred engineered B cells home to germinal centers (GC) where they predominate over the endogenous response and differentiate into memory and plasma cells while undergoing class switch recombination (CSR). Immunization with a high affinity antigen increases accumulation in GCs and CSR rates. Boost immunization increases the rate of engineered B cells in GCs and antibody secretion, indicating memory retention. Finally, antibody sequences of engineered B cells in the spleen show patterns of clonal selection. Therefore, B cells can be engineered into what could be a living and evolving drug.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Monoclonales/genética , Linfocitos B/inmunología , Anticuerpos ampliamente neutralizantes/genética , Anticuerpos Anti-VIH/genética , Memoria Inmunológica/genética , Vacunas contra el SIDA/genética , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Linfocitos B/fisiología , Linfocitos B/trasplante , Anticuerpos ampliamente neutralizantes/sangre , Anticuerpos ampliamente neutralizantes/inmunología , Ingeniería Genética/métodos , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/inmunología , Inmunización , Isotipos de Inmunoglobulinas/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación
5.
Nat Commun ; 11(1): 5850, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33203876

RESUMEN

HIV broadly neutralizing antibodies (bnAbs) can suppress viremia and protect against HIV infection. However, their elicitation is made difficult by low frequencies of appropriate precursor B cell receptors and the complex maturation pathways required to generate bnAbs from these precursors. Antibody genes can be engineered into B cells for expression as both a functional antigen receptor on cell surfaces and as secreted antibody. Here, we show that HIV bnAb-engineered primary mouse B cells can be adoptively transferred and vaccinated in immunocompetent mice resulting in the expansion of durable bnAb memory and long-lived plasma cells. Somatic hypermutation after immunization indicates that engineered cells have the capacity to respond to an evolving pathogen. These results encourage further exploration of engineered B cell vaccines as a strategy for durable elicitation of HIV bnAbs to protect against infection and as a contributor to a functional HIV cure.


Asunto(s)
Vacunas contra el SIDA/inmunología , Linfocitos B/inmunología , Anticuerpos ampliamente neutralizantes/inmunología , Animales , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Linfocitos B/fisiología , Linfocitos B/trasplante , Anticuerpos ampliamente neutralizantes/sangre , Anticuerpos ampliamente neutralizantes/genética , Femenino , Ingeniería Genética/métodos , Células HEK293 , Anticuerpos Anti-VIH/sangre , Anticuerpos Anti-VIH/genética , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH , Humanos , Inmunización , Memoria Inmunológica/genética , Activación de Linfocitos , Ratones Endogámicos C57BL , Hipermutación Somática de Inmunoglobulina
6.
Transfus Med Rev ; 34(3): 178-187, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32826130

RESUMEN

Passenger lymphocyte syndrome (PLS) is caused by the transfer of B-lymphocytes present in the donor graft into the recipient circulation following solid organ or hematopoietic stem cell transplantation. These cells may produce antibodies against the recipient's red blood cells, thereby triggering antibody dependent cytotoxicity and erythroid clearance, with potential resulting hemolysis and jaundice. Although uncommon, the true incidence is unknown because many cases are subclinical, with only serologic findings or with non significant levels of hemolysis detectable clinically or by laboratory monitoring. Thus, these cases may not be detected in the immediate perioperative period. No standardized consensus exists on screening for PLS in patients. Through a review of the literature from 2009 to 2019, we aim to approximate the incidence of this condition in different solid organ transplant settings, as well as to streamline recognition, detection, and management of PLS early in the disease course to prevent adverse outcomes and minimize invasive therapy. The resultant literature review yielded 22 case reports and 8 case series comprising 71 solid organ transplant patients. Hematopoietic stem cell transplant cases were excluded, as PLS cases related to solid organ transplant were the primary focus of this review. Our institution has traditionally handled PLS on a case-by-case basis, although we hope to improve this process through an introduction of an algorithm based on review of the literature and formalized communication with primary caregivers.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/trasplante , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Enfermedades del Sistema Inmune/etiología , Isoanticuerpos/inmunología , Trasplante de Órganos/efectos adversos , Complicaciones Posoperatorias , Humanos , Enfermedades del Sistema Inmune/diagnóstico , Enfermedades del Sistema Inmune/inmunología , Enfermedades del Sistema Inmune/terapia , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/inmunología , Complicaciones Posoperatorias/terapia , Síndrome
7.
Pharmacol Ther ; 214: 107603, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32553789

RESUMEN

Immunotherapy approaches consisting of genetically modified immune cells have become a promising platform for cancer treatment. Such 'living' therapies targeting tumor antigens have shown success in many cancer patients in the form of durable responses in a growing number of clinical studies. Besides, a large number of ongoing studies have been designed to introduce reliable methods for identification of tumor antigens. In addition, technical and biotechnological developments are being applied to the generation and expansion of genetically modified immune cells. In this review, we summarize and discuss the latest progress and current challenges in the tumor antigen landscape and in the generation of genetically modified immune cells in view of their clinical efficacy, either as monotherapy or combinational therapy.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Células Dendríticas/trasplante , Terapia Genética , Inmunoterapia Adoptiva , Subgrupos Linfocitarios/trasplante , Macrófagos/trasplante , Neoplasias/terapia , Receptores Quiméricos de Antígenos/genética , Animales , Antígenos de Neoplasias/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/trasplante , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/trasplante , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Resultado del Tratamiento , Microambiente Tumoral
8.
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
9.
Transplantation ; 103(10): 2065-2074, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31343579

RESUMEN

BACKGROUND: Interleukin 21 (IL-21) is involved in regulating the expansion and effector function of a broad range of leukocytes, including T cells and B cells. In transplantation, the exact role of IL-21 in the process of allograft rejection is unknown. To further explore this, the aim of this study is to test the effect of an IL-21 receptor (IL-21R) blocking antibody on the early phase of allograft rejection in a humanized skin transplantation model in mice reconstituted with human T and B cells. METHODS: Immunodeficient Balb/c IL2rγRag2 mice were transplanted with human skin followed by adoptive transfer of human allogeneic splenocytes. Control animals were treated with a phosphate buffered saline vehicle while the other group was treated with a humanized anti-IL-21R antibody (αIL-21R). RESULTS: In the phosphate buffered saline-treated animals, human skin allografts were infiltrated with lymphocytes and developed a thickened epidermis with increased expression of the inflammatory markers Keratin 17 (Ker17) and Ki67. In mice treated with αIL-21R, these signs of allograft reactivity were significantly reduced. Concordantly, STAT3 phosphorylation was inhibited in this group. Of note, treatment with αIL-21R attenuated the process of T and B cell reconstitution after adoptive cellular transfer. CONCLUSIONS: These findings demonstrate that blockade of IL-21 signaling can delay allograft rejection in a humanized skin transplantation model.


Asunto(s)
Rechazo de Injerto/inmunología , Inmunosupresores/administración & dosificación , Subunidad alfa del Receptor de Interleucina-21/antagonistas & inhibidores , Trasplante de Piel/efectos adversos , Aloinjertos/efectos de los fármacos , Aloinjertos/inmunología , Aloinjertos/patología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/trasplante , Proteínas de Unión al ADN/genética , Rechazo de Injerto/patología , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad alfa del Receptor de Interleucina-21/inmunología , Ratones Noqueados , Piel/efectos de los fármacos , Piel/inmunología , Piel/patología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Quimera por Trasplante , Trasplante Homólogo/efectos adversos
11.
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
12.
J Neurotrauma ; 36(17): 2579-2589, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30997843

RESUMEN

Cerebral contusion causes neurological dysfunction mediated in part by inflammatory responses to injury. B lymphocytes are dynamic regulators of the immune system that have not been systematically studied in traumatic brain injury (TBI). We showed previously that topically applied mature B cells have immunomodulatory properties and strongly promote tissue regeneration, including cutaneous nerve growth, in acute and chronic skin wounds. Using a mouse controlled cortical impact (CCI) model, we assessed a possible beneficial role of exogenously applied B cells on histopathological and functional outcome after TBI. Mice were injected intraparenchymally at the lesion site with 2 × 106 mature naïve syngeneic splenic B cells, then subjected to CCI. Control CCI mice received equal numbers of T cells or saline, and sham-injured mice (craniotomy only) were given B cells or saline. Sham-injured groups performed similarly in motor and learning tests. Injured mice administered B cells showed significantly improved post-injury rotarod, Y maze, and Morris water maze (MWM) performance compared with saline- or T-cell-treated CCI groups. Moreover, lesion volume in mice treated with B cells was significantly reduced by 40% at 35 days post-TBI compared with saline and T cell controls, and astrogliosis and microglial activation were decreased. In vivo tracking of exogenous B cells showed that they have a limited life span of approximately 14 days in situ and do not appear to proliferate. The data suggest proof of principle that local administration of B lymphocytes may represent a therapeutic option for treatment of cerebral contusion, especially when clinical management involves procedures that allow access to the injury site.


Asunto(s)
Linfocitos B/trasplante , Contusión Encefálica/patología , Contusión Encefálica/fisiopatología , Recuperación de la Función/fisiología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
13.
JCI Insight ; 3(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30429370

RESUMEN

Allograft tolerance, in which a graft is accepted without long-term immunosuppression, could overcome numerous obstacles in transplantation. Human allograft tolerance has been intentionally induced across HLA barriers via combined kidney and bone marrow transplantation (CKBMT) with a regimen that induces only transient chimerism. Tregs are enriched early after CKBMT. While deletional tolerance contributes to long-term tolerance, the role of Tregs remains unclear. We have optimized a method for identifying the donor-specific Treg repertoire and used it to interrogate the fate of donor-specific Tregs after CKBMT. We expanded Tregs with several different protocols. Using functional analyses and T cell receptor sequencing, we found that expanding sorted Tregs with activated donor B cells identified the broadest Treg repertoire with the greatest potency and donor specificity of suppression. This method outperformed both alloantigen stimulation with CTLA4Ig and sequencing of CFSElo cells from the primary mixed lymphocyte reaction. In 3 tolerant and 1 nontolerant CKBMT recipients, we sequenced donor-specific Tregs before transplant and tracked them after transplant. Preexisting donor-specific Tregs were expanded at 6 months after CKBMT in tolerant patients and were reduced in the nontolerant patient. These results suggest that early expansion of donor-specific Tregs is involved in tolerance induction following CKBMT.


Asunto(s)
Trasplante de Riñón , Linfocitos T Reguladores/trasplante , Tolerancia al Trasplante , Linfocitos B/inmunología , Linfocitos B/trasplante , Trasplante de Médula Ósea , Recuento de Linfocito CD4 , Antígeno CTLA-4/inmunología , Humanos , Prueba de Cultivo Mixto de Linfocitos , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Donantes de Tejidos
14.
Blood Adv ; 2(21): 2798-2810, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30355579

RESUMEN

Precursor B-cell acute lymphoblastic leukemia (B-ALL) is associated with recurrent mutations that occur in cancer-initiating cells. There is a need to understand how driver mutations influence clonal evolution of leukemia. The E26-transformation-specific (ETS) transcription factors PU.1 and Spi-B (encoded by Spi1 and Spib) execute a critical role in B-cell development and serve as complementary tumor suppressors. Here, we used a mouse model to conditionally delete Spi1 and Spib genes in developing B cells. These mice developed B-ALL with a median time to euthanasia of 18 weeks. We performed RNA and whole-exome sequencing (WES) on leukemias isolated from Mb1-CreΔPB mice and identified single nucleotide variants (SNVs) in Jak1, Jak3, and Ikzf3 genes, resulting in amino acid sequence changes. Jak3 mutations resulted in amino acid substitutions located in the pseudo-kinase (R653H, V670A) and in the kinase (T844M) domains. Introduction of Jak3 T844M into Spi1/Spib-deficient precursor B cells was sufficient to promote proliferation in response to low IL-7 concentrations in culture, and to promote proliferation and leukemia-like disease in transplanted mice. We conclude that mutations in Janus kinases represent secondary drivers of leukemogenesis that cooperate with Spi1/Spib deletion. This mouse model represents a useful tool to study clonal evolution in B-ALL.


Asunto(s)
Janus Quinasa 1/genética , Janus Quinasa 3/genética , Leucemia Linfocítica Crónica de Células B/patología , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Secuencia de Aminoácidos , Animales , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/trasplante , Proliferación Celular , Modelos Animales de Enfermedad , Factor de Transcripción Ikaros , Interleucina-7/farmacología , Janus Quinasa 1/química , Janus Quinasa 3/química , Leucemia Linfocítica Crónica de Células B/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Receptores de Interleucina-7/metabolismo , Eliminación de Secuencia , Transactivadores/química
15.
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
17.
Biosci Trends ; 12(3): 247-256, 2018 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-29806632

RESUMEN

The humanized mouse system is a promising tool for analyzing human immune responses in vivo. Recently, we developed a new humanized mouse system using the severely immunodeficient NOD/Shi-scid-IL2rγnull (NOG)-hIL-4-Tg mouse, which enabled us to evaluate the human humoral immune response after peripheral blood mononuclear cell (PBMC) transplantation. However, the mechanism by which hIL-4 enhances antigen-specific IgG production in these mice is not clear. In this study, we analyzed the relationship between human lymphocyte subsets and the expression level of the glucocorticoid receptor (GR) to clarify the humoral immune condition in human PBMC-transplanted NOG-hIL-4 mice. The results showed that the human GR mRNA level was significantly lower in NOG-hIL-4-Tg splenocytes than in conventional NOG splenocytes after immunization. Whereas no obvious difference of the proportion of T helper-cell subsets was observed between the NOG and NOG-hIL-4-Tg mouse strains, the B-cell proportion and antigen-specific IgG concentration in plasma showed strong negative correlations with the GR mRNA level. These results suggest that the GR expression level was changed in PBMCs in the humanized NOG-hIL-4-Tg mice, which may support B-cell survival and function in the mouse system.


Asunto(s)
Linfocitos B/inmunología , Enfermedad Injerto contra Huésped/inmunología , Interleucina-4/inmunología , Receptores de Glucocorticoides/metabolismo , Quimera por Trasplante/inmunología , Animales , Linfocitos B/metabolismo , Linfocitos B/trasplante , Supervivencia Celular/inmunología , Modelos Animales de Enfermedad , Voluntarios Sanos , Humanos , Inmunidad Humoral , Interleucina-4/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , ARN Mensajero/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/inmunología , Bazo/citología , Bazo/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología
18.
Virulence ; 9(1): 173-184, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28837391

RESUMEN

IgM and B-1 cell deficient mice exhibit early C. neoformans dissemination from lungs to brain, but a definitive role for B cells in conferring resistance to C. neoformans dissemination has not been established. To address this question, we developed an intranasal (i.n.) C. neoformans infection model in B and T cell deficient Rag1-/- mice and found they also exhibit earlier fungal dissemination and higher brain CFU than wild-type C57Bl/6 (wild-type) mice. To probe the effect of B cells on fungal dissemination, Rag1-/- mice were given splenic (intravenously) or peritoneal (intraperitoneally) B cells from wild-type mice and infected i.n. with C. neoformans 7 d later. Mice that received B cells had lung histopathology resembling wild type mice 14 d post-infection, and B-1, not B-2 or T cells in their lungs, and serum and lung IgM and IgG 21 d post-infection. Lung CFU were comparable in wild-type, Rag1-/-, and Rag1-/- mice that received B cells 21 d post-infection, but brain CFU were significantly lower in mice that received B cells than Rag1-/- mice that did not. To determine if natural antibody can promote immunity in our model, we measured alveolar macrophage phagocytosis of C. neoformans in Rag1-/- mice treated with naive wild-type IgM-sufficient or sIgM-/- IgM-deficient sera before infection. Compared to IgM-deficient sera, IgM-sufficient sera significantly increased phagocytosis. Our data establish B cells are able to reduce early C. neoformans dissemination in mice and suggest natural IgM may be a key mediator of early antifungal immunity in the lungs.


Asunto(s)
Linfocitos B/inmunología , Criptococosis/inmunología , Cryptococcus neoformans/crecimiento & desarrollo , Pulmón/microbiología , Traslado Adoptivo , Animales , Linfocitos B/trasplante , Encéfalo/microbiología , Recuento de Colonia Microbiana , Criptococosis/microbiología , Criptococosis/patología , Cryptococcus neoformans/inmunología , Citocinas/inmunología , Genes RAG-1/genética , Inmunoglobulina M/inmunología , Pulmón/inmunología , Pulmón/patología , Macrófagos Alveolares/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagocitosis/inmunología
20.
Autoimmunity ; 50(6): 363-369, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28686488

RESUMEN

The underlying cellular and molecular mechanism for the development of Type 1 diabetes is still to be fully revealed. We have previously demonstrated that the NOD mouse, a model for Type 1 diabetes, display a prolonged and enhanced immune response to both self and non-self-antigens. The molecular explanation for this defect however, has not been determined. In this study we immunized NOD and C57BL/6 (B6) with the conventional antigen i.e. hen egg lysozyme (HEL) and analyzed B cell activation, germinal center reaction and antibody clearance. Corroborating our previous observations NOD mice responded robustly to a single immunization of HEL. Immunofluorescence analysis of the spleen revealed an increased number of germinal centers in unimmunized NOD compared to B6. However, post immunization germinal center numbers were similar in NOD and B6. NOD mice showed lower response to BCR stimulation with anti-IgM, in particular at lower concentrations of anti-IgM. Antibody clearance in vivo did not differ between the strains. To determine the cell type that is responsible for the prolonged and enhance immune response, we reconstituted NOD-RAGs with cells from primed donors in different combinations. NOD B cells were required to reproduce the phenotype; however the non-lymphoid compartment of NOD origin also played a role. Based on our results we propose that preexisting GCs in the NOD promote the robust response and alteration in the BCR signaling could promote survival of stimulated cells. Overall, this mechanism could in turn also contribute to the activation and maintenance of autoreactive B cells in the NOD mouse.


Asunto(s)
Linfocitos B/inmunología , Diabetes Mellitus Experimental/inmunología , Centro Germinal/inmunología , Inmunización , Muramidasa/administración & dosificación , Receptores de Antígenos de Linfocitos B/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antiidiotipos/farmacología , Autoinmunidad/efectos de los fármacos , Autoinmunidad/genética , Linfocitos B/efectos de los fármacos , Linfocitos B/patología , Linfocitos B/trasplante , Pollos , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Progresión de la Enfermedad , Expresión Génica , Centro Germinal/efectos de los fármacos , Centro Germinal/patología , Humanos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Receptores de Antígenos de Linfocitos B/genética , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...