Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
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.
Am J Transplant ; 23(2): 202-213, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36804130

RESUMEN

Heart transplant and recipient survival are limited by immune cell-mediated injury of the graft vasculature. We examined the role of the phosphoinositide 3-kinase-ß (PI3Kß) isoform in endothelial cells (EC) during coronary vascular immune injury and repair in mice. In minor histocompatibility-antigen mismatched allogeneic heart grafts, a robust immune response was mounted to each wild-type, PI3Kß inhibitor-treated, or endothelial-selective PI3Kß knockout (ECßKO) graft transplanted to wild-type recipients. However, microvascular EC loss and progressive occlusive vasculopathy only developed in control, but not PI3Kß-inactivated hearts. We observed a delay in inflammatory cell infiltration of the ECßKO grafts, particularly in the coronary arteries. Surprisingly, this was accompanied by an impaired display of proinflammatory chemokine and adhesion molecules by the ECßKO ECs. In vitro, tumor necrosis factor α-stimulated endothelial ICAM1 and VCAM1 expression was blocked by PI3Kß inhibition or RNA interference. Selective PI3Kß inhibition also blocked tumor necrosis factor α-stimulated degradation of inhibitor of nuclear factor kappa Bα and nuclear translocation of nuclear factor kappa B p65 in EC. These data identify PI3Kß as a therapeutic target to reduce vascular inflammation and injury.


Asunto(s)
Células Endoteliales , Lesiones del Sistema Vascular , Ratones , Animales , Células Endoteliales/patología , Fosfatidilinositol 3-Quinasa , Fosfatidilinositol 3-Quinasas , Lesiones del Sistema Vascular/patología , Factor de Necrosis Tumoral alfa
3.
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.

4.
Transplantation ; 105(8): 1730-1746, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33273316

RESUMEN

BACKGROUND: In Medawar's murine neonatal tolerance model, injection of adult semiallogeneic lymphohematopoietic cells (spleen cells [SC] and bone marrow cells [BMC]) tolerizes the neonatal immune system. An eventual clinical application would require fully allogeneic (allo) cells, yet little is known about the complex in vivo/in situ interplay between those cells and the nonconditioned neonatal immune system. METHODS: To this end, labeled adult SC and BMC were injected into allogeneic neonates; interactions between donor and host cells were analyzed and modulated by systematic depletion/inactivation of specific donor and host immune effector cell types. RESULTS: Consistent with effector cell compositions, allo-SC and allo-SC/BMC each induced lethal acute graft-versus-host disease, whereas allo-BMC alone did so infrequently. CD8 T cells from SC inoculum appeared naïve, while those of BMC were more memory-like. Age-dependent, cell-type dominance defined the interplay between adult donor cells and the neonatal host immune system such that if the dominant adult effector type was removed, then the equivalent neonatal one became dominant. Depletion of donor/host peripheral T cells protected against acute graft-versus-host disease and prolonged heart allograft survival; peripheral CD8 T-cell depletion together with CD4 T cell-costimulation blockade induced more robust tolerance. CONCLUSIONS: This comprehensive study provides direct observation of the cellular interplay between allogeneic donor and host immune systems, adds to our previous work with semiallogeneic donor cells, and provides important insights for robust tolerance induction. Induction of transplant tolerance in neonates will likely require "crowd sourcing" of multiple tolerizing cell types and involve depletion of immune effector cells with costimulation blockade.


Asunto(s)
Animales Recién Nacidos/inmunología , Tolerancia Inmunológica , Animales , Células de la Médula Ósea/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/prevención & control , Ratones , Ratones Endogámicos , Bazo/inmunología
5.
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
6.
Transplantation ; 96(6): 519-28, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23860083

RESUMEN

BACKGROUND: ABO-incompatible (ABOi) infant heart transplantation results in B-cell tolerance to graft A/B antigens, confirming human susceptibility to acquired immunologic or "neonatal" tolerance as described originally in murine models. Starting with this clinical observation, we sought to model neonatal ABOi organ transplantation to allow mechanistic studies of tolerance. METHODS: Plasma anti-A/B antibodies were measured over time in piglets to establish developmental antibody kinetics. Blood group O piglets received kidney allografts from group A (AO-incompatible) or group O (AO-compatible) donors under cyclosporine immunosuppression. Anti-A antibodies were measured serially after transplantation; A/H antigen expression and allograft rejection were assessed in graft biopsies. RESULTS: Anti-A antibodies developed in naïve piglets in a kinetic pattern analogous to human infants; anti-B remained low. After transplantation, anti-A antibodies developed similarly in AO-incompatible and AO-compatible groups and were not suppressed by cyclosporine. A/H antigen expression was persistent in all graft biopsies; however, A/H antigens were not detected in vascular endothelium. Cellular and antibody-mediated rejection was absent or minimal in early and late biopsies in both groups, with one exception. CONCLUSIONS: Naturally delayed isohemagglutinin production in piglets is analogous to the developmental kinetics in human infants. However, in contrast to deficient anti-A antibody production as seen long-term after "A-into-O" infant heart transplant recipients, normal anti-A antibody production after "A-into-O" piglet kidney transplantation indicates that tolerance did not develop despite graft A antigen persistence. These findings suggest that the impact on the host immune system of exposure to nonself ABH antigens during early life in human heart versus porcine kidney grafts may depend on expression in vascular endothelium.


Asunto(s)
Sistema del Grupo Sanguíneo ABO/inmunología , Linfocitos B/inmunología , Incompatibilidad de Grupos Sanguíneos/inmunología , Histocompatibilidad , Trasplante de Riñón/inmunología , Tolerancia al Trasplante , Animales , Animales Recién Nacidos , Biopsia , Ciclosporina/farmacología , Rechazo de Injerto/inmunología , Supervivencia de Injerto , Histocompatibilidad/efectos de los fármacos , Prueba de Histocompatibilidad , Inmunosupresores/farmacología , Isoanticuerpos/sangre , Trasplante de Riñón/efectos adversos , Cinética , Modelos Animales , Sus scrofa
7.
Ultrasound Med Biol ; 33(6): 870-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17434663

RESUMEN

This study investigated the use of an ultrasound biomicroscope (UBM) to observe murine heterotopic cardiac transplants. By using an UBM (30 MHz), cardiac isografts in eight mice were studied on days 1, 5, 14 and 50 posttransplantation. The same method was tested in allografts in two mice on days 1, 5, 7 and 9. Two-dimensional imaging delineated the graft structures with high spatial resolution. In isografts, M-mode recording showed gradually decreased left ventricular (LV) wall thickness and chamber dimension, but increased LV fractional shortening. Doppler sampling measured blood velocities from the ascending aorta, left coronary artery (LCA), aortic and mitral orifices of grafts. In isografts, LCA forward flow caused by native circulation to perfuse the graft myocardium increased from day 1 to 5, then moderately decreased by day 14 and stabilized thereafter. In allografts, LCA forward flow sharply decreased to almost zero between day 5-9. Therefore, UBM is a reliable method for following the survival status of cardiac grafts in mice.


Asunto(s)
Trasplante de Corazón/diagnóstico por imagen , Microscopía Acústica/métodos , Trasplante Heterotópico , Animales , Aorta/diagnóstico por imagen , Aorta/fisiopatología , Velocidad del Flujo Sanguíneo/fisiología , Circulación Coronaria/fisiología , Vasos Coronarios/diagnóstico por imagen , Vasos Coronarios/fisiopatología , Ecocardiografía Doppler/métodos , Femenino , Corazón/fisiopatología , Ventrículos Cardíacos/diagnóstico por imagen , Masculino , Ratones , Ratones Endogámicos C3H , Estenosis de la Válvula Mitral/diagnóstico por imagen , Estenosis de la Válvula Mitral/fisiopatología , Modelos Animales , Arteria Pulmonar/diagnóstico por imagen , Arteria Pulmonar/fisiopatología
8.
Proc Natl Acad Sci U S A ; 101(48): 16909-14, 2004 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-15550536

RESUMEN

Successful therapy for many inherited disorders could be improved if the intervention were initiated early. This is especially true for lysosomal storage disorders. Earlier intervention may allow metabolic correction to occur before lipid buildup has irreversible consequences and/or before the immune system mounts limiting responses. We have been developing gene therapy to treat lysosomal storage disorders, especially Fabry disease. We describe studies directed toward metabolic correction in neonatal animals mediated by recombinant lentiviral vectors. To develop this method, we first injected a marking lentiviral vector that engineers expression of luciferase into the temporal vein of recipient neonatal animals. The use of a cooled charged-coupled device camera allowed us to track transgene expression over time in live animals. We observed intense luciferase expression in many tissues, including the brain, that did not diminish over 24 weeks. Next, we injected neonatal Fabry mice a single time with a therapeutic lentiviral vector engineered to express human alpha-galactosidase A. The injection procedure was well tolerated. We observed increased plasma levels of alpha-galactosidase A activity starting at our first plasma collection point (4 weeks). Levels of alpha-galactosidase A activity were found to be significantly elevated in many tissues even after 28 weeks. No immune response was observed against the corrective transgene product. Increased levels of enzyme activity also led to significant reduction of globotriaosylceramide in the liver, spleen, and heart. This approach provides a method to treat lysosomal storage disorders and other disorders before destructive manifestations occur.


Asunto(s)
Enfermedad de Fabry/terapia , Vectores Genéticos , Lentivirus/genética , Transgenes , Animales , Animales Recién Nacidos , Línea Celular , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Enfermedad de Fabry/genética , Humanos , Inmunohistoquímica , Ratones
9.
J Heart Lung Transplant ; 23(9): 1069-76, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15454173

RESUMEN

BACKGROUND: Non-donor-specific cardiac allograft acceptance is induced in C3H/He (C3H; H-2k) recipients injected as neonates with allogeneic BALB/c (BALB; H-2d) fetal liver cells (FLC). This occurs despite intact reactivity to donor-type and third-party alloantigens in in vitro assays and skin transplants. To investigate a role for regulatory T cells, we performed adoptive transfer studies and specifically assessed CD4+ and CD4- T cells. METHODS: Three cell populations (splenocytes, CD4+, CD4-) derived from neonatally-treated mice with accepted C57BL/6 (B6; H-2b) third-party cardiac grafts were adoptively transferred into sub-lethally-irradiated C3H mice. Reconstituted mice were challenged with B6 cardiac grafts, B6 skin grafts, or unrelated cardiac grafts. Separated cells were assessed in vitro. RESULTS: B6, BALB, and NZW (H-2z) graft acceptance was transferred by unfractionated splenocytes. CD4+ cells transferred B6 graft acceptance (85% survival > 100 days). CD4- cells, unfractionated cells from naive or only irradiated mice, and unfractionated cells from neonatally-treated non-transplanted C3H mice rejected grafts within 35 days. No inoculum induced skin graft acceptance. Co-cultured assays confirmed the suppressive function of CD4+ cells in vitro. CONCLUSIONS: Cardiac allograft acceptance in our model is regulated by CD4+ cells. The regulatory cell population is induced by the cardiac graft itself and mediates in vivo cardiac graft acceptance in a tissue-specific but not donor-strain-specific manner.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Trasplante de Corazón/inmunología , Traslado Adoptivo , Animales , Animales Recién Nacidos , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Bazo/citología , Inmunología del Trasplante , Trasplante Homólogo
10.
Transplantation ; 78(6): 844-52, 2004 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-15385803

RESUMEN

BACKGROUND: The immune response against xenografts is vigorous and poorly controlled with conventional immunosuppressants. Therefore, success in xenotransplantation will depend on developing additional approaches such as induction of immunologic unresponsiveness or tolerance. Although classic protocols of neonatal tolerance induction in mice are very tolerogenic in many allogeneic models, they have generally failed in xenogeneic models. The purpose of these studies was to determine whether failure results from an intrinsic property of xenogenic major histocompatibility complex (MHC) molecules themselves or, instead, is caused by some limitation in species-specific molecular interactions distinct from the polymorphic domains of xenogenic MHC molecules. METHODS: Our approach was to test the ability of lymphoid cells from a transgenic (Tg) mouse donor expressing a xeno-MHC class I molecule encoding the polymorphic alpha1/alpha2 for human leukocyte antigen (HLA)-B7 to induce neonatal tolerance in non-Tg syngeneic C57BL/6 recipients. Because the donor and recipient strains are genetically identical (C57BL/6, H-2b) except for Tg human MHC HLA-B7, any species-specific molecular incompatibility in this mouse anti-human class I xeno-combination that could potentially interfere with induction of tolerance has been eliminated. RESULTS: Our results show that HLA-B7 Tg-, but not C57BL/6 syngeneic-, injected neonates were unresponsive as adults to HLA-B7-expressing target cells in vitro and specifically accepted HLA-B7-expressing Tg skin grafts. In addition, neonatal injection of donor cells resulted in peripheral chimerism. CONCLUSIONS: These experiments demonstrate that, as long as species-specific molecular interactions are maintained, recognition of the polymorphic domains of xenogeneic MHC does not represent a barrier to neonatal tolerance induction.


Asunto(s)
Antígeno HLA-B7/genética , Antígeno HLA-B7/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Tolerancia al Trasplante/inmunología , Trasplante Heterólogo/inmunología , Animales , Cruzamientos Genéticos , Femenino , Citometría de Flujo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Bazo/inmunología
11.
Am J Transplant ; 2(8): 733-44, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12243494

RESUMEN

Neonatal tolerance is exclusively donor-specific when assessed by skin allograft survival and in vitro alloreactivity assays. In contrast, we reported previously that acceptance of primarily vascularized cardiac allografts was not donor-specific in C3H/He (C3H, H-2(k)) mice treated as neonates with BALB/c-derived (BALB, H-2(d)) lymphohematopoietic cells, but included third-party C57BL/10 (B10, H-2(b)) allografts. The present study examined whether this unusual pattern is limited to heart grafts in this strain combination, and defined the relative importance of the donor cell H-2(d) haplotype for third-party cardiac allograft acceptance. C3H neonates were injected with (C3HxBALB)F1 bone marrow and spleen cells. Tolerance was assessed at age 8-10 weeks by transplantation of heart or skin allografts from several donor strains, and by in vitro assays of proliferation and cytotoxicity. Additionally, cells from H-2(d) and H-2(b)-expressing strains on BALB or non-BALB minor histocompatibility (miH) antigen backgrounds were tested as tolerizing inocula. Prolonged survival of cardiac grafts from all donor strains was observed in neonatally treated mice, whereas skin grafting and in vitro assays demonstrated donor-specific hyporesponsiveness. Both H-2(d) haplotype and non-H-2 miH background of graft donor and tolerizing cell donor were important to third-party cardiac allograft acceptance. These results suggest that the functional alteration in alloreactivity induced by neonatal alloantigen exposure depends partly on method of assessment.


Asunto(s)
Rechazo de Injerto/prevención & control , Trasplante de Corazón/inmunología , Trasplante de Células Madre Hematopoyéticas , Trasplante de Piel/inmunología , Animales , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...