Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transplantation ; 107(11): 2353-2363, 2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37871273

RESUMEN

BACKGROUND: "Natural" ABO antibodies (Abs) are produced without known exposure to A/B carbohydrate antigens, posing significant risks for hyperacute rejection during ABO-incompatible transplantation. We investigated anti-A "natural" ABO antibodies versus intentionally induced Abs with regard to the need for T-cell help, the impact of sex, and stimulation by the microbiome. METHODS: Anti-A was measured by hemagglutination assay of sera from untreated C57BL/6 wild-type (WT) or T cell-deficient mice of both sexes. Human ABO-A reagent blood cell membranes were injected intraperitoneally to induce anti-A Abs. The gut microbiome was eliminated by maintenance of mice in germ-free housing. RESULTS: Compared with WT mice, CD4 + T-cell knockout (KO), major histocompability complex-II KO, and αß/γδ T-cell receptor KO mice produced much higher levels of anti-A nAbs; females produced dramatically more anti-A nAbs than males, rising substantially with puberty. Sensitization with human ABO-A reagent blood cell membranes did not induce additional anti-A in KO mice, unlike WT. Sex-matched CD4 + T-cell transfer significantly suppressed anti-A nAbs in KO mice and rendered mice responsive to A-sensitization. Even under germ-free conditions, WT mice of several strains produced anti-A nAbs, with significantly higher anti-A nAbs levels in females than males. CONCLUSIONS: Anti-A nAbs were produced without T-cell help, without microbiome stimulation, in a sex- and age-dependent manner, suggestive of a role for sex hormones in regulating anti-A nAbs. Although CD4 + T cells were not required for anti-A nAbs, our findings indicate that T cells regulate anti-A nAb production. In contrast to anti-A nAbs, induced anti-A production was T-cell dependent without a sex bias.


Asunto(s)
Formación de Anticuerpos , Microbiota , Masculino , Femenino , Ratones , Animales , Humanos , Ratones Endogámicos C57BL , Anticuerpos , Linfocitos T CD4-Positivos , Ratones Noqueados
2.
RSC Chem Biol ; 3(10): 1260-1275, 2022 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-36320887

RESUMEN

Synthetic glycoconjugates are used in the development of vaccines and the design of inhibitors for glycan-protein interactions. The in vivo persistence of synthetic glycoconjugates is an important factor in their efficacy, especially when prolonged interactions with specific cell types may be required. In this study, we applied a strategy for non-covalent association of an active compound with serum proteins for extension of glycoconjugate half-life in serum. The small molecule, AG10, has previously been used to extend the half-life of small molecules through its high affinity for transthyretin (TTR), a serum protein. Using a tetravalent polyethylene glycol (PEG)-based scaffold we developed a synthetic strategy for glycoconjugates that allowed for controlled addition of multiple tags, such as a TTR affinity tag or fluorophore. We designed a version of AG10 modified at the pyrazole core, named GD10, amenable to our conjugation strategy and introduced to glycoconjugates using a tri-functional linker. This approach allowed for attachment of GD10 and fluorophore tags, as well as carbohydrate antigens. We then tested the influence of the GD10 tag on glycoconjugate half-life in vivo using a mouse model. Our results suggest that the combination of the GD10 tag and the PEG scaffold extended the half-life of glycoconjugates by as much as 10-fold when compared to proteins of similar molecular weight. The GD10 tag was able to extend the half-life of similar glycoconjugates by as much as 2-fold. We observed a role for the terminal saccharide residue of the carbohydrate antigen and confirmed that conjugates were able to penetrate multiple compartments in vivo including bone marrow, lymph nodes, and other organs. The introduction of the GD10 tag did not obstruct the ability of conjugates to interact with lectin receptors. We conclude that serum protein binders can be used to extend the persistence of glycoconjugates in vivo.

3.
Pediatr Transplant ; 26(4): e14252, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35187796

RESUMEN

BACKGROUND: Atopic disorders are more common in children after heart transplant (HTx). We hypothesized that HTx at an early age and thymus excision (TE) affect development of T and B cells, especially regulatory T cells (Tregs), which help maintain tolerance. METHODS: In this single-center study including 24 patients transplanted between 2013 and 2018, we investigated lymphocyte patterns in relation to these factors using flow cytometry. Clinical data were collected from standardized questionnaires and medical charts. Patients were stratified into TE and non-TE groups as well as patients with and without post-transplant atopy development/worsening. RESULTS: 64% of TE patients experienced new or worsening asthma/eczema post-transplant compared to 20% of non-TE patients. TE patients had higher total Treg proportions (CD4+CD25+CD127lo) than non-TE patients (p = .043), but borderline significantly lower naïve Tregs (CD45RA+CD27-) (p = .057). Memory CD4+ T cells were higher in TE patients in trend (p = .084). Total Tregs did not differ between atopic/nonatopic groups, although naïve Tregs were significantly lower in atopic patients (p = .028). Memory CD4+ T cells were higher in atopic patients in trend (p = .082). IgM+IgD+ B cells were higher in nonatopic patients in trend (p = .064). CONCLUSIONS: New/worsening atopy is more common in thymectomized HTx children and is associated with alterations in T-cell profiles. Avoiding TE may prevent these alterations and reduce incidence of atopy post-HTx.


Asunto(s)
Trasplante de Corazón , Humanos , Inmunofenotipificación , Fenotipo , Linfocitos T Reguladores , Timectomía
4.
Virulence ; 13(1): 30-45, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-34967260

RESUMEN

Since December 2019, the coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread throughout the world. To eradicate it, it is crucial to acquire a strong and long-lasting anti-SARS-CoV-2 immunity, by either natural infection or vaccination. We collected blood samples 12-305 days after positive polymerase chain reactions (PCRs) from 35 recovered individuals infected by SARS-CoV-2. Peripheral blood mononuclear cells were stimulated with SARS-CoV-2-derived peptide pools, such as the spike (S), nucleocapsid (N) and membrane (M) proteins, and we quantified anti-S immunoglobulins in plasma. After 10 months post-infection, we observed a sustained SARS-CoV-2-specific CD4+ T-cell response directed against M-protein, but responses against S- or N-proteins were lost over time. Besides, we demonstrated that O-group individuals presented significantly lower frequencies of specific CD4+ T-cell responses against Pep-M than non O-group individuals. The non O-group subjects also needed longer to clear the virus, and they lost cellular immune responses over time, compared to the O-group individuals, who showed a persistent specific immune response against SARS-CoV-2. Therefore, the S-specific immune response was lost over time, and individual factors might determine the sustainability of the body's defenses, which must be considered in the future design of vaccines to achieve continuous anti-SARS-CoV-2 immunity.


Asunto(s)
Sistema del Grupo Sanguíneo ABO , COVID-19/sangre , Inmunidad Humoral , Células T de Memoria , SARS-CoV-2/inmunología , Humanos , Inmunidad Celular , Leucocitos Mononucleares , Glicoproteína de la Espiga del Coronavirus
5.
Am J Transplant ; 21(11): 3649-3662, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34101982

RESUMEN

ABO-incompatible (ABOi) transplantation requires preemptive antibody reduction; however, the relationship between antibody-mediated rejection (AMR) and ABO-antibodies, quantified by hemagglutination (HA), is inconsistent, possibly reflecting variable graft resistance to AMR or HA assay limitations. Using an ABH-glycan microarray, we quantified ABO-A antigen-subtype (A-subtype)-specific IgM and IgG in 53 ABO-O recipients of ABO-A kidneys, before and after antibody removal (therapeutic plasma exchange [TPE] or ABO-A-trisaccharide immunoadsorption [IA]) and 1-year posttransplant. IgM binding to all A-subtypes correlated highly (R2  ≥ .90) and A-subtype antibody specificities was reduced equally by IA versus TPE. IgG binding to the A-subtypes (II-IV) expressed in kidney correlated poorly (.27 ≤ R2  ≤ .69). Reduction of IgG specific to A-subtype-II was equivalent for IA and TPE, whereas IgG specific to A-subtypes-III/IV was not as greatly reduced by IA (p < .005). One-year posttransplant, IgG specific to A-II remained the most reduced antibody. Immunostaining revealed only A-II on vascular endothelium but A-subtypes II-III/IV on tubular epithelium. These results show that ABO-A-trisaccharide is sufficient for IgM binding to all A-subtypes; this is true for IgG binding to A-II, but not subtypes-III/IV, which exhibits varying degrees of specificity. We identify A-II as the major, but importantly not the sole, antigen relevant to treatment and immune modulation in adult ABO-A-incompatible kidney transplantation.


Asunto(s)
Trasplante de Riñón , Sistema del Grupo Sanguíneo ABO , Adulto , Incompatibilidad de Grupos Sanguíneos , Rechazo de Injerto , Humanos , Donadores Vivos
6.
Front Immunol ; 11: 597433, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329589

RESUMEN

Newborns are highly susceptible to infectious diseases. The underlying mechanism of neonatal infection susceptibility has generally been related to their under-developed immune system. Nevertheless, this notion has recently been challenged by the discovery of the physiological abundance of immunosuppressive erythroid precursors CD71+erythroid cells (CECs) in newborn mice and human cord blood. Here, as proof of concept, we show that these cells are also abundant in the peripheral blood of human newborns. Although their frequency appears to be more variable compared to their counterparts in mice, they rapidly decline by 4 weeks of age. However, their proportion remains significantly higher in infants up to six months of age compared to older infants. We found CD45 expressing CECs, as erythroid progenitors, were the prominent source of reactive oxygen species (ROS) production in both humans and mice. Interestingly, a higher proportion of CD45+CECs was observed in the spleen versus bone marrow of neonatal mice, which was associated with a higher ROS production by splenic CECs compared to their siblings in the bone marrow. CECs from human newborns suppressed cytokine production by CD14 monocytes and T cells, which was partially abrogated by apocynin in vitro. Moreover, the depletion of CECs in neonatal mice increased the number of activated effector immune cells in their spleen and liver, which rendered them more resistant to Listeria monocytogenes infection. This was evident by a significant reduction in the bacteria load in the spleen, liver and brain of treated-mice compared to the control group, which enhanced their survival rate. Our finding highlights the immunoregulatory processes mediated by CECs in newborns. Thus, such tightly regulated immune system in newborns/infants may explain one potential mechanism for the asymptomatic or mild COVID-19 infection in this population.


Asunto(s)
Antígenos CD/inmunología , Células Precursoras Eritroides , Terapia de Inmunosupresión , Listeria monocytogenes/inmunología , Listeriosis , Receptores de Transferrina/inmunología , Animales , Animales Recién Nacidos , COVID-19/inmunología , COVID-19/patología , Células Precursoras Eritroides/inmunología , Células Precursoras Eritroides/patología , Células Precursoras Eritroides/trasplante , Femenino , Xenoinjertos , Humanos , Recién Nacido , Listeriosis/inmunología , Listeriosis/patología , Listeriosis/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , SARS-CoV-2/inmunología
7.
Hum Vaccin Immunother ; 16(4): 779-792, 2020 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-31687875

RESUMEN

Chimigen® HBV Immunotherapeutic Vaccine (C-HBV), a recombinant chimeric fusion protein comprising hepatitis B virus (HBV) S1 and S2 surface antigen fragments, Core antigen and a murine monoclonal antibody heavy chain fragment (Fc), was designed and produced in Sf9 insect cells. C-HBV targets the host immune system through specific receptors present on dendritic cells (DCs) which facilitates antigen internalization, processing, and presentation on MHC class I and II to induce both cellular and humoral immune responses against HBV antigens. T cell responses, previously assessed by ex vivo antigen presentation assays using human peripheral blood mononuclear cell (PBMC)-derived DCs and T cells from uninfected and HBV chronic-infected donors, demonstrated that C-HBV was highly immunogenic. A vaccine dose response study was performed in sheep to analyze the immunogenicity of C-HBV in vivo. Sheep (n = 8/group) received three consecutive subcutaneous injections of each dose of C-HBV at four-week intervals. Analysis of serum antibody levels confirmed C-HBV induced a dose-dependent antibody response to C-HBV and S1/S2-Core. Kinetics of the S1/S2-Core specific antibody response was similar to hepatitis B surface antigen (HBsAg)-specific antibody responses induced by ENGERIX-B. Analysis of cell-mediated immune responses (CMI) confirmed C-HBV induced both dose-dependent S1/S2-Core-specific lymphocyte proliferative responses and IFN-γ secretion. These responses were stronger with blood lymphocytes than with cells isolated from the lymph node draining the vaccination site. No correlation was seen between antibody titers and CMI. The results confirm C-HBV is an effective delivery vehicle for the induction of T cell responses and may be an appropriate candidate for immunotherapy for chronic HBV infections.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B , Animales , Células Dendríticas , Anticuerpos contra la Hepatitis B , Antígenos de Superficie de la Hepatitis B , Vacunas contra Hepatitis B , Inmunidad Humoral , Inmunoterapia , Leucocitos Mononucleares , Ratones , Ovinos
8.
Hum Vaccin Immunother ; 16(4): 756-778, 2020 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-31687879

RESUMEN

In chronic Hepatitis B Virus (HBV) infections HBV-specific T cells are functionally impaired. Immunotherapy may restore HBV-specific T cell responses essential for sustained disease remission off-treatment and induction of a functional cure. Chimigen® Molecules are fusion proteins of antigen(s) with the Fc fragment of a xenotypic antibody designed to target specific receptors on dendritic cells (DCs). Here we describe the production and pre-clinical evaluation of Chimigen® HBV (C-HBV), containing HBV PreS1 and PreS2 peptide fragments, HBV core and murine Fc, produced in insect cells. C-HBV binding to immature DCs and internalization by endocytosis was FcγRII (CD32) and mannose receptor (CD206) dependent and led to increased MHC I and MHC II surface expression. Upon exposure of human T cells isolated from HBV un-infected healthy and chronically HBV-infected donors to C-HBV-pulsed mature DCs ex vivo, C-HBV induced vigorous T cell proliferation and enhanced expression of IFN-γ, TNF-α, perforin and granzyme B in both CD4+ and CD8+ T cell subsets. Re-stimulation of C-HBV-activated T cells from chronically infected donors with HBV PreS1/PreS2 and core overlapping peptides induced IFN-γ production in both CD4+ and CD8+ populations. C-HBV-activation of peripheral blood mononuclear cells (PBMCs) from chronically HBV-infected patients stimulated granzyme B production by CD4+CD25- T responder (Tresp) cells, accompanied by an increase in Annexin V staining on CD4+CD25+ T regulatory (Treg) cell phenotype, consistent with apoptosis. The observed HBV-specific cellular responses induced by C-HBV ex vivo suggest that C-HBV is a promising immunotherapeutic candidate for the treatment of chronic HBV infections.


Asunto(s)
Células Dendríticas , Hepatitis B Crónica , Inmunoterapia , Animales , Linfocitos T CD8-positivos , Virus de la Hepatitis B , Hepatitis B Crónica/terapia , Humanos , Leucocitos Mononucleares , Ratones
9.
Transplantation ; 100(6): 1228-37, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27120451

RESUMEN

BACKGROUND: ABO-incompatible (ABOi) organ transplantation is performed owing to unremitting donor shortages. Defining mechanisms of antibody-mediated rejection, accommodation, and tolerance of ABOi grafts is limited by lack of a suitable animal model. We report generation and characterization of a murine model to enable study of immunobiology in the setting of ABOi transplantation. METHODS: Transgenesis of a construct containing human A1- and H-transferases under control of the ICAM-2 promoter was performed in C57BL/6 (B6) mice. A-transgenic (A-Tg) mice were assessed for A-antigen expression by histology and flow cytometry. B6 wild-type (WT) mice were sensitized with blood group A-human erythrocytes; others received passive anti-A monoclonal antibody and complement after heart transplant. Serum anti-A antibodies were assessed by hemagglutination. "A-into-O" transplantation (major histocompatibility complex syngeneic) was modeled by transplanting hearts from A-Tg mice into sensitized or nonsensitized WT mice. Antibody-mediated rejection was assessed by morphology/immunohistochemistry. RESULTS: A-Tg mice expressed A-antigen on vascular endothelium and other cells including erythrocytes. Antibody-mediated rejection was evident in 15/17 A-Tg grafts in sensitized WT recipients (median titer, 1:512), with 2 showing hyperacute rejection and rapid cessation of graft pulsation. Hyperacute rejection was observed in 8/8 A-Tg grafts after passive transfer of anti-A antibody and complement into nonsensitized recipients. Antibody-mediated rejection was not observed in A-Tg grafts transplanted into nonsensitized mice. CONCLUSIONS: A-Tg heart grafts transplanted into WT mice with abundant anti-A antibody manifests characteristic features of antibody-mediated rejection. These findings demonstrate an effective murine model to facilitate study of immunologic features of ABOi transplantation and to improve potential diagnostic and therapeutic strategies.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/inmunología , Incompatibilidad de Grupos Sanguíneos/inmunología , Rechazo de Injerto , Trasplante de Corazón , Animales , Anticuerpos Monoclonales/inmunología , Antígenos/inmunología , Antígenos CD/genética , Moléculas de Adhesión Celular/genética , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/citología , Eritrocitos/inmunología , Citometría de Flujo , Glicosiltransferasas/genética , Supervivencia de Injerto , Humanos , Tolerancia Inmunológica , Inmunohistoquímica , Inmunofenotipificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas
10.
Hum Gene Ther ; 21(7): 877-90, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20163247

RESUMEN

The ABO histo-blood group system is the most important antigen system in transplantation medicine, yet no small animal model of the ABO system exists. To determine the feasibility of developing a murine model, we previously subcloned the human alpha-1,2-fucosyltransferase (H-transferase, EC 2.4.1.69) cDNA and the human alpha-1,3-N-acetylgalactosaminyltransferase (A-transferase, EC 2.4.1.40) cDNA into lentiviral vectors to study their ability to induce human histo-blood group A antigen expression on mouse cells. Herein we investigated the optimal conditions for human A and H antigen expression in murine cells. We determined that transduction of a bicistronic lentiviral vector (LvEF1-AH-trs) resulted in the expression of A antigen in a mouse endothelial cell line. We also studied the in vivo utility of this vector to induce human A antigen expression in mouse liver. After intrahepatic injection of LvEF1-AH-trs, A antigen expression was observed on hepatocytes as detected by immunohistochemistry and real-time RT-PCR. In human group A erythrocyte-sensitized mice, A antigen expression in the liver was associated with tissue damage, and deposition of antibody and complement. These results suggest that this gene transfer strategy can be used to simulate the human ABO blood group system in a murine model. This model will facilitate progress in the development of interventions for ABO-incompatible transplantation and transfusion scenarios, which are difficult to develop in clinical or large animal settings.


Asunto(s)
Sistema del Grupo Sanguíneo ABO , Vectores Genéticos , Antígenos HLA-A/metabolismo , Lentivirus/genética , Modelos Biológicos , Transducción Genética/métodos , Animales , Células Cultivadas , Femenino , Citometría de Flujo , Vectores Genéticos/genética , Humanos , Inmunohistoquímica , Hígado/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Dev Comp Immunol ; 34(7): 749-58, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20156479

RESUMEN

Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4, CD152) is an inhibitory T cell receptor predominately expressed on activated T cells. The duck CTLA-4 (DuCTLA-4) cDNA and a transcript lacking the predicted transmembrane encoding region (DuCTLA-4DeltaTM) were isolated from splenocytes using RT-PCR. The predicted DuCTLA-4 protein showed an identity of 92%, 49% and 47% with chicken, human and mouse homologues, respectively. Sequence comparison revealed conservation of residues implicated in the B7 ligand binding, disulfide linkages, glycosylation and intracellular signaling. DuCTLA-4 mRNA was predominately expressed in primary and secondary immune organs. DuCTLA-4 and DuCTLA-4DeltaTM transcripts were differentially regulated in PBMCs. Flow cytometric analysis showed constitutive expression of DuCTLA-4 protein on freshly isolated PBMCs and a modest increase upon mitogen stimulation. Our observations suggest that DuCTLA-4 and its isoform DuCTLA-4DeltaTM evolved before the divergence of birds and mammals. Both DuCTLA-4 isoforms have significant structural homology to mammalian CTLA-4 proteins but their individual roles in the regulation of duck immune responses remains to be elucidated.


Asunto(s)
Antígenos CD/aislamiento & purificación , Patos/inmunología , Inmunidad Adaptativa/inmunología , Secuencia de Aminoácidos , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Secuencia de Bases , Antígeno CTLA-4 , Clonación Molecular , Patos/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Filogenia , Isoformas de Proteínas , ARN Mensajero/química , ARN Mensajero/genética , Técnica del ADN Polimorfo Amplificado Aleatorio , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia , Análisis de Secuencia de ADN
12.
J Immunol ; 180(6): 3766-74, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18322182

RESUMEN

IFN-gamma, a pleiotropic immune regulator, is implicated in both tumor immune surveillance and selection of tumor variants resistant to immune control, i.e., immunoediting. In uveal melanoma patients, elevated serum levels of IFN-gamma correlate with the spread of metastasis and represent a negative prognostic marker. Treatment with IFN-gamma boosted the MHC class I presentation machinery in uveal melanoma cells but suppressed their MHC class I-restricted CTL lysis. Tumor cells exposed to IFN-gamma efficiently activated specific CTL but were less susceptible to permeabilization by perforin and exhibited a decreased capacity to bind and incorporate granzyme B. These results define a novel mechanism of resistance to granule-mediated CTL lysis in human tumors. Furthermore, the data suggest that immunoediting is not limited to genetic or epigenetic changes resulting in stable cellular phenotypes but also involves an inducible modulation of tumor cells in response to a microenvironment associated with immune activation.


Asunto(s)
Citotoxicidad Inmunológica , Granzimas/fisiología , Inmunofenotipificación , Interferón gamma/fisiología , Melanoma/inmunología , Linfocitos T Citotóxicos/inmunología , Neoplasias de la Úvea/inmunología , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/fisiología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular/inmunología , Células Clonales , Regulación hacia Abajo/inmunología , Granzimas/antagonistas & inhibidores , Humanos , Tolerancia Inmunológica , Inmunidad Innata/inmunología , Interferón gamma/sangre , Melanoma/metabolismo , Melanoma/patología , Melanoma/secundario , Perforina/fisiología , Pronóstico , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/metabolismo , Regulación hacia Arriba/inmunología , Neoplasias de la Úvea/enzimología , Neoplasias de la Úvea/patología
13.
J Immunol ; 177(8): 5051-8, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17015688

RESUMEN

Sertoli cells have long since been recognized for their ability to suppress the immune system and protect themselves as well as other cell types from harmful immune reaction. However, the exact mechanism or product produced by Sertoli cells that affords this immunoprotection has never been fully elucidated. We examined the effect of mouse Sertoli cell-conditioned medium on human granzyme B-mediated killing and found that there was an inhibitory effect. We subsequently found that a factor secreted by Sertoli cells inhibited killing through the inhibition of granzyme B enzymatic activity. SDS-PAGE analysis revealed that this factor formed an SDS-insoluble complex with granzyme B. Immunoprecipitation and mass spectroscopic analysis of the complex identified a proteinase inhibitor, serpina3n, as a novel inhibitor of human granzyme B. We cloned serpina3n cDNA, expressed it in Jurkat cells, and confirmed its inhibitory action on granzyme B activity. Our studies have led to the discovery of a new inhibitor of granzyme B and have uncovered a new mechanism used by Sertoli cells for immunoprotection.


Asunto(s)
Proteínas de Fase Aguda/aislamiento & purificación , Granzimas/antagonistas & inhibidores , Serpinas/aislamiento & purificación , Células de Sertoli/metabolismo , Proteínas de Fase Aguda/inmunología , Proteínas de Fase Aguda/metabolismo , Animales , Factores Biológicos/inmunología , Factores Biológicos/aislamiento & purificación , Factores Biológicos/metabolismo , Células Cultivadas , Clonación Molecular , Humanos , Sistema Inmunológico , Células Jurkat , Masculino , Ratones , Unión Proteica , Serpinas/inmunología , Serpinas/metabolismo , Células de Sertoli/citología , Células de Sertoli/inmunología
14.
Mol Biol Cell ; 17(2): 623-33, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16280358

RESUMEN

During granule-mediated killing by cytotoxic T lymphocytes or natural killer cells, the serine protease granzyme B enters the target cell by endocytosis and induces apoptosis. Previous studies suggested a role for the mannose 6-phosphate receptor, but further experiments with purified granzyme B indicated this was not essential. Additionally, it is now clear that grB is exocytosed from killer cells in a high-molecular-weight complex with the proteoglycan serglycin. Here granzyme B was delivered as a purified monomer, or in complex with either glycosaminoglycans or serglycin, and killing was evaluated. When granzyme B was a monomer, soluble mannose 6-phosphate had a limited impact, whereas apoptosis induced by the complexed grB was effectively inhibited by mannose 6-phosphate. Most importantly, when granzyme B and perforin were delivered together from granules, inhibition by mannose 6-phosphate was also observed. In pulldown assays mediated by the cation-independent mannose 6-phosphate receptor, granzyme B bound to the receptor more intensely in the presence of immobilized heparan sulfate. We therefore propose the model that under physiological conditions serglycin-bound granzyme B is critically endocytosed by a mannose 6-phosphate receptor, and receptor binding is enhanced by cell surface heparan sulfate.


Asunto(s)
Heparitina Sulfato/fisiología , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/fisiología , Receptor IGF Tipo 2/fisiología , Vesículas Secretoras/fisiología , Serina Endopeptidasas/fisiología , Linfocitos T Citotóxicos/inmunología , Animales , Apoptosis , Línea Celular , Glicosaminoglicanos/metabolismo , Granzimas , Heparitina Sulfato/química , Humanos , Células Jurkat , Ratones , Modelos Biológicos , Perforina , Proteínas Citotóxicas Formadoras de Poros , Proteoglicanos/fisiología , Vesículas Secretoras/enzimología , Linfocitos T Citotóxicos/enzimología , Proteínas de Transporte Vesicular/fisiología
15.
Arterioscler Thromb Vasc Biol ; 24(12): 2245-50, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15472125

RESUMEN

OBJECTIVE: T cell-induced cytotoxicity, of which granzyme B is a key mediator, is believed to contribute to the pathogenesis of inflammatory vascular diseases. In this report, we investigate the mechanism of granzyme B-induced smooth muscle cell (SMC) death. METHODS AND RESULTS: The addition of purified granzyme B alone to cultured SMCs caused a significant reduction in cell viability. Chromatin condensation, phosphatidylserine externalization, and membrane blebbing were observed, indicating that the mechanism of granzyme B-induced SMC death was through apoptosis. Activated splenocytes from perforin-knockout mice induced SMC death through a granzyme B-mediated pathway. Inhibition of the proteolytic activities of caspases and granzyme B prevented granzyme B-induced SMC death, whereas attenuation of granzyme B internalization with mannose-6-phosphate (M6P) did not. Further, granzyme B induced the cleavage of several SMC extracellular proteins, including fibronectin, and reduced focal adhesion kinase phosphorylation. CONCLUSIONS: These results indicate that granzyme B can induce apoptosis of SMCs in the absence of perforin by cleaving extracellular proteins, such as fibronectin.


Asunto(s)
Apoptosis/fisiología , Matriz Extracelular/metabolismo , Glicoproteínas de Membrana/deficiencia , Miocitos del Músculo Liso/fisiología , Serina Endopeptidasas/fisiología , Animales , Caspasa 3 , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/fisiología , Vasos Coronarios/enzimología , Vasos Coronarios/metabolismo , Matriz Extracelular/enzimología , Proteínas de la Matriz Extracelular/metabolismo , Granzimas , Células HeLa/química , Células HeLa/metabolismo , Humanos , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/enzimología , Perforina , Proteínas Citotóxicas Formadoras de Poros , Ratas , Serina Endopeptidasas/metabolismo , Bazo/citología , Bazo/metabolismo
16.
Blood ; 103(10): 3845-53, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-14739229

RESUMEN

Cytotoxic T lymphocytes and natural killer cells destroy target cells via the directed exocytosis of lytic effector molecules such as perforin and granzymes. The mechanism by which these proteins enter targets is uncertain. There is ongoing debate over whether the most important endocytic mechanism is nonspecific or is dependent on the cation-independent mannose 6-phosphate receptor. This study tested whether granzyme B endocytosis is facilitated by dynamin, a key factor in many endocytic pathways. Uptake of and killing by the purified granzyme B molecule occurred by both dynamin-dependent and -independent mechanisms. However most importantly, serglycin-bound granzyme B in high-molecular-weight degranulate material from cytotoxic T lymphocytes predominantly followed a dynamin-dependent pathway to kill target cells. Similarly, killing by live cytotoxic T lymphocytes was attenuated by a defect in the dynamin endocytic pathway, and in particular, the pathways characteristically activated by granzyme B were affected. We therefore propose a model where degranulated serglycin-bound granzymes require dynamin for uptake.


Asunto(s)
Gránulos Citoplasmáticos/inmunología , Citotoxicidad Inmunológica , Dinaminas/fisiología , Endocitosis/inmunología , Proteoglicanos/metabolismo , Serina Endopeptidasas/metabolismo , Apoptosis/inmunología , Gránulos Citoplasmáticos/enzimología , Dinaminas/genética , Granzimas , Células HeLa , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/ultraestructura , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/ultraestructura , Transfección , Proteínas de Transporte Vesicular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...