Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Immunol ; 189(1): 222-33, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22649197

RESUMEN

We reported that both donor CD4(+) T and B cells in transplants were required for induction of an autoimmune-like chronic graft-versus-host disease (cGVHD) in a murine model of DBA/2 donor to BALB/c recipient, but mechanisms whereby donor B cells augment cGVHD pathogenesis remain unknown. In this study, we report that, although donor B cells have little impact on acute GVHD severity, they play an important role in augmenting the persistence of tissue damage in the acute and chronic GVHD overlapping target organs (i.e., skin and lung); they also markedly augment damage in a prototypical cGVHD target organ, the salivary gland. During cGVHD pathogenesis, donor B cells are activated by donor CD4(+) T cells to upregulate MHC II and costimulatory molecules. Acting as efficient APCs, donor B cells augment donor CD4(+) T clonal expansion, autoreactivity, IL-7Rα expression, and survival. These qualitative changes markedly augment donor CD4(+) T cells' capacity in mediating autoimmune-like cGVHD, so that they mediate disease in the absence of donor B cells in secondary recipients. Therefore, a major mechanism whereby donor B cells augment cGVHD is through augmenting the clonal expansion, differentiation, and survival of pathogenic CD4(+) T cells.


Asunto(s)
Autoanticuerpos/biosíntesis , Subgrupos de Linfocitos B/trasplante , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Proliferación Celular , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Regulación hacia Arriba/inmunología , Animales , Subgrupos de Linfocitos B/inmunología , Subgrupos de Linfocitos B/patología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Enfermedad Crónica , Células Clonales , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/genética , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Ratones Noqueados , Ratones Transgénicos , Glándulas Salivales/inmunología , Glándulas Salivales/patología , Piel/inmunología , Piel/patología , Regulación hacia Arriba/genética
2.
J Clin Invest ; 134(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38426503

RESUMEN

Tissue-intrinsic mechanisms that regulate severity of systemic pathogenic immune-mediated diseases, such as acute graft-versus-host disease (GVHD), remain poorly understood. Following allogeneic hematopoietic stem cell transplantation, autophagy, a cellular stress protective response, is induced in host nonhematopoietic cells. To systematically address the role of autophagy in various host nonhematopoietic tissues, both specific classical target organs of acute GVHD (intestines, liver, and skin) and organs conventionally not known to be targets of GVHD (kidneys and heart), we generated mice with organ-specific knockout of autophagy related 5 (ATG5) to specifically and exclusively inhibit autophagy in the specific organs. When compared with wild-type recipients, animals that lacked ATG5 in the gastrointestinal tract or liver showed significantly greater tissue injury and mortality, while autophagy deficiency in the skin, kidneys, or heart did not affect mortality. Treatment with the systemic autophagy inducer sirolimus only partially mitigated GVHD mortality in intestine-specific autophagy-deficient hosts. Deficiency of autophagy increased MHC class I on the target intestinal epithelial cells, resulting in greater susceptibility to damage by alloreactive T cells. Thus, autophagy is a critical cell-intrinsic protective response that promotes tissue tolerance and regulates GVHD severity.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Ratones , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Intestinos/patología , Linfocitos T/patología , Células Epiteliales/patología
3.
J Immunol ; 186(2): 856-68, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149609

RESUMEN

Chronic graft-versus-host disease (cGVHD) is considered an autoimmune-like disease mediated by donor CD4(+) T cells, but the origin of the autoreactive T cells is still controversial. In this article, we report that the transplantation of DBA/2 donor spleen cells into thymectomized MHC-matched allogeneic BALB/c recipients induced autoimmune-like cGVHD, although not in control syngeneic DBA/2 recipients. The donor-type CD4(+) T cells from the former but not the latter recipients induced autoimmune-like manifestations in secondary allogeneic BALB/c as well as syngeneic DBA/2 recipients. Transfer of donor-type CD4(+) T cells from secondary DBA/2 recipients with disease into syngeneic donor-type or allogeneic host-type tertiary recipients propagated autoimmune-like manifestations in both. Furthermore, TCR spectratyping revealed that the clonal expansion of the autoreactive CD4(+) T cells in cGVHD recipients was initiated by an alloimmune response. Finally, hybridoma CD4(+) T clones derived from DBA/2 recipients with disease proliferated similarly in response to stimulation by syngeneic donor-type or allogeneic host-type dendritic cells. These results demonstrate that the autoimmune-like manifestations in cGVHD can be mediated by a population of donor CD4(+) T cells in transplants that simultaneously recognize Ags presented by both donor and host APCs.


Asunto(s)
Autoantígenos/metabolismo , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Enfermedad Injerto contra Huésped/inmunología , Antígenos H-2/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/patología , Autoanticuerpos/biosíntesis , Autoantígenos/administración & dosificación , Autoantígenos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular/genética , Enfermedad Crónica , Células Clonales , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/patología , Antígenos H-2/administración & dosificación , Antígenos H-2/genética , Antígeno de Histocompatibilidad H-2D , Antígenos de Histocompatibilidad Clase II/administración & dosificación , Antígenos de Histocompatibilidad Clase II/genética , Prueba de Histocompatibilidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Modelos Animales , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/trasplante
4.
J Immunol ; 186(5): 2739-49, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21263067

RESUMEN

Foxp3(+) regulatory T (Treg) cells include thymic-derived natural Treg and conventional T-derived adaptive Treg cells. Both are proposed to play important roles in downregulating inflammatory immune responses. However, the mechanisms of Treg expansion in inflammatory environments remain unclear. In this study, we report that, in an autoimmune-like graft-versus-host disease model of DBA/2 (H-2(d)) donor to BALB/c (H-2(d)) recipients, donor Treg cells in the recipients predominantly originated from expansion of natural Treg cells and few originated from adaptive Treg cells. In vivo neutralization of IFN-γ resulted in a marked reduction of donor natural Treg expansion and exacerbation of graft-versus-host disease, which was associated with downregulation of host APC expression of B7H1. Furthermore, host APC expression of B7H1 was shown to augment donor Treg survival and expansion. Finally, donor Treg interactions with host APCs via B7.1/B7H1 but not PD-1/B7H1 were demonstrated to be critical in augmenting donor Treg survival and expansion. These studies have revealed a new immune regulation loop consisting of T cell-derived IFN-γ, B7H1 expression by APCs, and B7.1 expression by Treg cells.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos de Superficie/fisiología , Proteínas Reguladoras de la Apoptosis/fisiología , Antígeno B7-1/fisiología , Diferenciación Celular/inmunología , Glicoproteínas de Membrana/fisiología , Péptidos/fisiología , Linfocitos T Reguladores/inmunología , Animales , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/patología , Antígenos de Superficie/biosíntesis , Proteínas Reguladoras de la Apoptosis/biosíntesis , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Antígeno B7-1/biosíntesis , Antígeno B7-H1 , Diferenciación Celular/genética , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Inmunidad Innata/genética , Ligando Coestimulador de Linfocitos T Inducibles , Interferón gamma/antagonistas & inhibidores , Interferón gamma/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Péptidos/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1 , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
5.
Nat Commun ; 14(1): 3164, 2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37258514

RESUMEN

Although battery electric vehicles (BEVs) are climate-friendly alternatives to internal combustion engine vehicles (ICEVs), an important but often ignored fact is that the climate mitigation benefits of BEVs are usually delayed. The manufacture of BEVs is more carbon-intensive than that of ICEVs, leaving a greenhouse gas (GHG) debt to be paid back in the future use phase. Here we analyze millions of vehicle data from the Chinese market and show that the GHG break-even time (GBET) of China's BEVs ranges from zero (i.e., the production year) to over 11 years, with an average of 4.5 years. 8% of China's BEVs produced and sold between 2016 and 2018 cannot pay back their GHG debt within the eight-year battery warranty. We suggest enhancing the share of BEVs reaching the GBET by promoting the effective substitution of BEVs for ICEVs instead of the single-minded pursuit of speeding up the BEV deployment race.

6.
Res Sq ; 2023 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-36945465

RESUMEN

Intestinal stem cells (ISC) encounter inflammatory insults in immune mediated gastro-intestinal (GI) diseases. It remains unknown whether, and how, they adapt, and if the adaptation leaves scars on the ISCs that affects their subsequent regeneration capacity. We investigated the consequences of inflammation on Lgr5+ISCs in well-defined clinically relevant models of gastro-intestinal acute graft-versus-host disease (GI GVHD). Utilizing single cell transcriptomics, organoid, metabolic, epigenomic and in vivo models we found that Lgr5+ISCs undergo metabolic changes that lead to accumulation of succinate, which reprograms its epigenome. These changes reduced the ability of ISCs to differentiate and regenerate ex vivo in serial organoid cultures demonstrating the persistence of the maladaptive impact of an in vivo inflammatory encounter by the ISCs. Thus, inflammation from GI GVHD leaves a memory of its effects on ISCs that persist and are likely to affect their sensitivity to adapt to future stress or challenges.

7.
Sci Total Environ ; 826: 154102, 2022 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-35218846

RESUMEN

Promoting new energy vehicles (NEVs) is the key to achieving net-zero emissions in the transportation sector. NEVs' total life cycle CO2 emissions are mainly determined by average vehicle lifespan, annual mileage traveled, energy carbon intensity and energy mix in the production stage. Current studies mainly adopt assumptions about NEVs' average lifespan due to limited available data. This paper expands on the previous studies by examining the NEVs' age and distribution based on the national representative China Compulsory Traffic Accident Liability Insurance for Motor Vehicles (CTALI) database from 2018 to 2020. Then, the survival patterns and lifespan of NEVs are assessed using Weibull distribution. New energy passenger vehicles' life cycle CO2 emissions are further evaluated based on the reshaped representative survival patterns. The results show that there are significant differences in survival patterns between conventional vehicles and NEVs. NEVs generally show a shorter average lifespan compared with conventional vehicles. Among NEVs, the average lifespan of plug-in hybrid electric vehicles (PHEVs) is better than that of battery electric vehicles (BEVs). The survival patterns of several types of electric vehicles (including passenger battery electric vehicles, non-operating light battery electric buses, and light battery electric trucks) do not have a stable period in their first few years of operation. The life cycle assessment results show that the total life cycle CO2 emissions of passenger BEVs and PHEVs are lower than those of conventional vehicles. However, the short lifespan dramatically increases the passenger BEV and PHEV total life cycle CO2 emissions per kilometer, resulting in passenger BEV total life cycle CO2 emissions per kilometer being higher than those of conventional vehicles.


Asunto(s)
Dióxido de Carbono , Emisiones de Vehículos , Animales , Dióxido de Carbono/análisis , Electricidad , Estadios del Ciclo de Vida , Vehículos a Motor , Emisiones de Vehículos/análisis
8.
Transplant Cell Ther ; 28(8): 472.e1-472.e11, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35643350

RESUMEN

Conditioning regimens used for hematopoietic stem cell transplantation (HCT) can escalate the severity of acute T cell-mediated graft-versus-host disease (GVHD) by disrupting gastrointestinal integrity and initiating lipopolysaccharide (LPS)-dependent innate immune cell activation. Activation of the complement cascade has been associated with murine GVHD, and previous work has shown that alternative pathway complement activation can amplify T cell immunity. Whether and how mannan-binding lectin (MBL), a component of the complement system that binds mannose as well as oligosaccharide components of LPS and lipoteichoic acid, affects GVHD is unknown. In this study, we tested the hypothesis that MBL modulates murine GVHD and examined the mechanisms by which it does so. We adoptively transferred C3.SW bone marrow (BM) cells ± T cells into irradiated wild type (WT) or MBL-deficient C57Bl/6 (B6) recipients with or without inhibiting MBL-initiated complement activation using C1-esterase inhibitor (C1-INH). We analyzed the clinical severity of disease expression and analyzed intestinal gene and cell infiltration. In vitro studies assessed MBL expression on antigen-presenting cells (APCs) and compared LPS-induced responses of WT and MBL-deficient APCs. MBL-deficient recipients of donor BM ± T cells exhibited significantly less weight loss over the first 2 weeks post-transplantation weeks compared with B6 controls (P < .05), with similar donor engraftment in the 2 groups. In recipients of C3.SW BM + T cells, the clinical expression of GVHD was less severe (P < .05) and overall survival was better (P < .05) in MBL-deficient mice compared with WT mice. On day-7 post-transplantation, analyses showed that the MBL-deficient recipients exhibited less intestinal IL1b, IL17, and IL12 p40 gene expression (P < .05 for each) and fewer infiltrating intestinal CD11c+, CD11b+, and F4/80+ cells and TCRß+, CD4+, CD4+IL17+, and CD8+ T cells (P < .05 for each). Ovalbumin or allogeneic cell immunizations induced equivalent T cell responses in MBL-deficient and WT mice, demonstrating that MBL-deficiency does not directly impact T cell immunity in the absence of irradiation conditioning. Administration of C1-INH did not alter the clinical expression of GVHD in preconditioned WT B6 recipients, suggesting that MBL amplifies clinical expression of GVHD via a complement-independent mechanism. WT, but not MBL-deficient, APCs express MBL on their surfaces. LPS-stimulated APCs from MBL-deficient mice produced less proinflammatory cytokines (P < .05) and induced weaker alloreactive T cell responses (P < .05) compared with WT APCs. Together, our data show that MBL modulates murine GVHD, likely by amplifying complement-independent, LPS-initiated gastrointestinal inflammation. The results suggest that devising strategies to block LPS/MBL ligation on APCs has the potential to reduce the clinical expression of GVHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Inflamación , Lectina de Unión a Manosa , Animales , Trasplante de Médula Ósea , Linfocitos T CD8-positivos , Enfermedad Injerto contra Huésped/genética , Inflamación/etiología , Inflamación/genética , Lipopolisacáridos/efectos adversos , Lipopolisacáridos/farmacología , Lectina de Unión a Manosa/genética , Ratones , Ratones Endogámicos C57BL , Trasplante Homólogo
9.
Glia ; 59(3): 486-98, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21264954

RESUMEN

Despite significant infiltration into tumors, the effector function of macrophages (MPs) and microglia (MG) appears to be suppressed in gliomas. Although STAT3 pathway is thought to play a role in this process, the exact mechanism by which gliomas induce STAT3 activation in MPs and MG is not known. Because activation of receptor for advanced glycation end products (RAGE) can induce STAT3, and because gliomas express high levels of S100B, a RAGE ligand, we hypothesized that MP/MG STAT3 activity may be modulated through S100B-RAGE interaction. Exposure of N9 MG and bone marrow-derived monocytes (BMM) to GL261 glioma condition medium (GCM) and low (nM) levels of S100B increased RAGE expression, induced STAT3 and suppressed MG function in vitro. Furthermore, neutralization of S100B in GCM, partially reversed IL-1ß suppression in BMM, suggesting that the inhibitory effect of GCM to be in part due to S100B. Finally, blockage of S100B-RAGE interaction inhibited STAT3 activation in N9 MG and in glioma MG/MP in vivo. These findings suggest that the RAGE pathway may play an important role in STAT3 induction in glioma-associated MG/MPs, and that this process may be mediated through S100B.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Microglía/metabolismo , Factores de Crecimiento Nervioso/fisiología , Proteínas S100/fisiología , Factor de Transcripción STAT3/fisiología , Transducción de Señal/fisiología , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microglía/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/fisiología , Subunidad beta de la Proteína de Unión al Calcio S100 , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética
10.
Blood ; 113(4): 953-62, 2009 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-18922852

RESUMEN

Host dendritic cells (DCs) play a critical role in initiating graft-versus-host disease (GVHD) and graft-versus-leukemia (GVL), and separation of GVL from GVHD remains a major challenge in the treatment of hematologic malignancies by allogeneic hematopoietic cell transplantation (HCT). Here, we show that preconditioning with anti-CD3 monoclonal antibody before conditioning with total body irradiation (TBI) prevents GVHD but retains GVL in a HCT model of major histocompatibility complex (MHC)-mismatched C57BL/6 donor to BALB/c host. Prevention of GVHD is associated with inhibition of donor T-cell expression of homing and chemokine receptors, and inhibition of GVHD target tissue expression of chemokines. Furthermore, inhibition of donor T-cell expression of gut homing alpha4beta7 and chemokine receptor (CCR)9 by anti-CD3 preconditioning results from a reduction of CD103(+) DCs in draining mesenteric lymph nodes (LNs), which is associated with down-regulation of DC expression of CCR7, a receptor required for tissue DC migration to draining LNs. These results indicate that anti-CD3 preconditioning reduces not only tissue release of chemokines but also prevents tissue DC migration to draining LNs and subsequently reduces the capacity of DCs of draining LNs to imprint donor T-cell tissue tropism. Therefore, modulation of host DCs by anti-CD3 preconditioning before HCT represents a new approach for separating GVL from GVHD.


Asunto(s)
Anticuerpos/inmunología , Complejo CD3/inmunología , Células Dendríticas/inmunología , Enfermedad Injerto contra Huésped/inmunología , Leucemia/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Animales , Antígenos CD/inmunología , Donantes de Sangre , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Quimiocinas/metabolismo , Regulación hacia Abajo , Trasplante de Células Madre Hematopoyéticas , Cadenas alfa de Integrinas/inmunología , Leucemia/patología , Leucemia/cirugía , Ratones , Receptores de Quimiocina/inmunología , Linfocitos T/metabolismo , Acondicionamiento Pretrasplante , Trasplante Homólogo , Regulación hacia Arriba , Irradiación Corporal Total
11.
Blood ; 114(14): 3101-12, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19602708

RESUMEN

In acute graft-versus-host disease (GVHD), naive donor CD4(+) T cells recognize alloantigens on host antigen-presenting cells and differentiate into T helper (Th) subsets (Th1, Th2, and Th17 cells), but the role of Th subsets in GVHD pathogenesis is incompletely characterized. Here we report that, in an MHC-mismatched model of C57BL/6 donor to BALB/c recipient, WT donor CD4(+) T cells predominantly differentiated into Th1 cells and preferentially mediated GVHD tissue damage in gut and liver. However, absence of interferon-gamma (IFN-gamma) in CD4(+) T cells resulted in augmented Th2 and Th17 differentiation and exacerbated tissue damage in lung and skin; absence of both IL-4 and IFN-gamma resulted in augmented Th17 differentiation and preferential, although not exclusive, tissue damage in skin; and absence of both IFN-gamma and IL-17 led to further augmentation of Th2 differentiation and idiopathic pneumonia. The tissue-specific GVHD mediated by Th1, Th2, and Th17 cells was in part associated with their tissue-specific migration mediated by differential expression of chemokine receptors. Furthermore, lack of tissue expression of the IFN-gamma-inducible B7-H1 played a critical role in augmenting the Th2-mediated idiopathic pneumonia. These results indicate donor CD4(+) T cells can reciprocally differentiate into Th1, Th2, and Th17 cells that mediate organ-specific GVHD.


Asunto(s)
Diferenciación Celular , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Linfocitos T Colaboradores-Inductores/patología , Células TH1/patología , Células Th2/patología , Animales , Anticuerpos Monoclonales/inmunología , Antígeno B7-1/fisiología , Antígeno B7-H1 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Movimiento Celular , Citometría de Flujo , Enfermedad Injerto contra Huésped/metabolismo , Neumonías Intersticiales Idiopáticas/etiología , Neumonías Intersticiales Idiopáticas/patología , Interferón gamma/fisiología , Interleucina-17/fisiología , Interleucina-4 , Pulmón/inmunología , Pulmón/patología , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Péptidos/fisiología , Piel/inmunología , Piel/patología , Linfocitos T Colaboradores-Inductores/inmunología , Células TH1/inmunología , Células Th2/inmunología
12.
Proc Natl Acad Sci U S A ; 105(12): 4796-801, 2008 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18347343

RESUMEN

In allogeneic hematopoietic cell transplantation (HCT), donor T cell-mediated graft versus host leukemia (GVL) and graft versus autoimmune (GVA) activity play critical roles in treatment of hematological malignancies and refractory autoimmune diseases. However, graft versus host disease (GVHD), which sometimes can be fatal, remains a major obstacle in classical HCT, where recipients are conditioned with total body irradiation or high-dose chemotherapy. We previously reported that anti-CD3 conditioning allows donor CD8(+) T cells to facilitate engraftment and mediate GVL without causing GVHD. However, the clinical application of this radiation-free and GVHD preventative conditioning regimen is hindered by the cytokine storm syndrome triggered by anti-CD3 and the high-dose donor bone marrow (BM) cells required for induction of chimerism. Histone deacetylase (HDAC) inhibitors such as suberoylanilide hydroxamic acid (SAHA) are known to induce apoptosis of cancer cells and reduce production of proinflammatory cytokines by nonmalignant cells. Here, we report that SAHA inhibits the proliferative and cytotoxic activity of anti-CD3-activated T cells. Administration of low-dose SAHA reduces cytokine production and ameliorates the cytokine storm syndrome triggered by anti-CD3. Conditioning with anti-CD3 and SAHA allows induction of chimerism with lower doses of donor BM cells in old nonautoimmune and autoimmune lupus mice. In addition, conditioning with anti-CD3 and SAHA allows donor CD8(+) T cell-mediated GVA activity to reverse lupus glomerulonephritis without causing GVHD. These results indicate that conditioning with anti-CD3 and HDAC inhibitors represent a radiation-free and GVHD-preventative regimen with clinical application potential.


Asunto(s)
Complejo CD3/inmunología , Quimerismo/efectos de los fármacos , Citocinas/inmunología , Trasplante de Células Madre Hematopoyéticas , Ácidos Hidroxámicos/farmacología , Nefritis Lúpica/inmunología , Acondicionamiento Pretrasplante , Envejecimiento/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimerismo/inducido químicamente , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Femenino , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/administración & dosificación , Nefritis Lúpica/patología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Vorinostat
13.
Nat Commun ; 12(1): 805, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547295

RESUMEN

Efforts to improve the prognosis of steroid-resistant gut acute graft-versus-host-disease (SR-Gut-aGVHD) have suffered from poor understanding of its pathogenesis. Here we show that the pathogenesis of SR-Gut-aGVHD is associated with reduction of IFN-γ+ Th/Tc1 cells and preferential expansion of IL-17-IL-22+ Th/Tc22 cells. The IL-22 from Th/Tc22 cells causes dysbiosis in a Reg3γ-dependent manner. Transplantation of IFN-γ-deficient donor CD8+ T cells in the absence of CD4+ T cells produces a phenocopy of SR-Gut-aGVHD. IFN-γ deficiency in donor CD8+ T cells also leads to a PD-1-dependent depletion of intestinal protective CX3CR1hi mononuclear phagocytes (MNP), which also augments expansion of Tc22 cells. Supporting the dual regulation, simultaneous dysbiosis induction and depletion of CX3CR1hi MNP results in full-blown Gut-aGVHD. Our results thus provide insights into SR-Gut-aGVHD pathogenesis and suggest the potential efficacy of IL-22 antagonists and IFN-γ agonists in SR-Gut-aGVHD therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Disbiosis/inmunología , Enfermedad Injerto contra Huésped/inmunología , Interferón gamma/inmunología , Interleucinas/inmunología , Fagocitos/inmunología , Animales , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Modelos Animales de Enfermedad , Disbiosis/genética , Disbiosis/microbiología , Disbiosis/patología , Microbioma Gastrointestinal/inmunología , Regulación de la Expresión Génica , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/microbiología , Enfermedad Injerto contra Huésped/patología , Interferón gamma/deficiencia , Interferón gamma/genética , Interleucina-17/deficiencia , Interleucina-17/genética , Interleucina-17/inmunología , Interleucinas/genética , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Depleción Linfocítica , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/inmunología , Fagocitos/citología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Transducción de Señal , Linfocitos T Colaboradores-Inductores , Linfocitos T Reguladores , Irradiación Corporal Total , Interleucina-22
14.
Blood ; 112(5): 2129-38, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18550852

RESUMEN

CD103 (alphaEbeta7) has been shown to be an excellent marker for identifying in vivo-activated FoxP3(+)CD4(+) regulatory T (Treg) cells. It is unknown whether reinfusion of in vivo-activated donor-type CD103(+) Treg cells from recipient can ameliorate ongoing chronic graft-versus-host disease (GVHD). Here, we showed that, in a chronic GVHD model of DBA/2 (H-2(d)) donor to BALB/c (H-2(d)) recipient, donor-type CD103(+) Treg cells from recipients were much more potent than CD25(hi) natural Treg cells from donors in reversing clinical signs of GVHD and tissue damage. Furthermore, in contrast to CD25(hi) natural Treg cells, CD103(+) Treg cells expressed high levels of CCR5 but low levels of CD62L and directly migrated to GVHD target tissues. In addition, the CD103(+) Treg cells strongly suppressed donor CD4(+) T-cell proliferation; they also induced apoptosis of in vivo-activated CD4(+) T and B cells and significantly reduced pathogenic T and B cells in GVHD target tissues. These results indicate that CD103(+) Treg cells from chronic GVHD recipients are functional, and reinfusion of the CD103(+) Treg cells can shift the balance between Treg cells and pathogenic T cells in chronic GVHD recipients and ameliorate ongoing disease.


Asunto(s)
Antígenos CD/metabolismo , Enfermedad Injerto contra Huésped/terapia , Cadenas alfa de Integrinas/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Animales , Apoptosis/inmunología , Autoanticuerpos/biosíntesis , Enfermedad Crónica , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Células Plasmáticas/inmunología , Células Plasmáticas/patología , Bazo/citología , Bazo/inmunología , Bazo/trasplante , Sindecanos/inmunología , Linfocitos T Reguladores/clasificación , Donantes de Tejidos , Trasplante Homólogo
15.
Blood ; 112(5): 2101-10, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18596226

RESUMEN

Th17 is a newly identified T-cell lineage that secretes proinflammatory cytokine IL-17. Th17 cells have been shown to play a critical role in mediating autoimmune diseases such as EAE, colitis, and arthritis, but their role in the pathogenesis of graft-versus-host disease (GVHD) is still unknown. Here we showed that, in an acute GVHD model of C57BL/6 (H-2(b)) donor to BALB/c (H-2(d)) recipient, IL-17(-/-) donor T cells manifested an augmented Th1 differentiation and IFN-gamma production and induced exacerbated acute GVHD. Severe tissue damage mediated by IL-17(-/-) donor T cells was associated with increased Th1 infiltration, up-regulation of chemokine receptors by donor T cells, and enhanced tissue expression of inflammatory chemokines. Administration of recombinant IL-17 and neutralizing IFN-gamma in the recipients given IL-17(-/-) donor cells ameliorated the acute GVHD. Furthermore, the regulation of Th1 differentiation by IL-17 or Th17 may be through its influence on host DCs. Our results indicate that donor Th17 cells can down-regulate Th1 differentiation and ameliorate acute GVHD in allogeneic recipients, and that treatments neutralizing proinflammatory cytokine IL-17 may augment acute GVHD as well as other inflammatory autoimmune diseases.


Asunto(s)
Enfermedad Injerto contra Huésped/etiología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Células TH1/inmunología , Células TH1/patología , Animales , Secuencia de Bases , Diferenciación Celular , Quimiocinas/genética , Cartilla de ADN/genética , Expresión Génica , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Interferón gamma/antagonistas & inhibidores , Interleucina-17/administración & dosificación , Interleucina-17/biosíntesis , Interleucina-17/deficiencia , Interleucina-17/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Linfocitos T Colaboradores-Inductores/clasificación , Linfocitos T Colaboradores-Inductores/citología , Linfocitos T Colaboradores-Inductores/inmunología , Donantes de Tejidos , Trasplante Homólogo
16.
J Clin Invest ; 128(11): 4970-4979, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30106382

RESUMEN

Graft-versus-host disease (GVHD) in the gastrointestinal (GI) tract remains the major cause of morbidity and nonrelapse mortality after BM transplantation (BMT). The Paneth cell protein regenerating islet-derived 3α (REG3α) is a biomarker specific for GI GVHD. REG3α serum levels rose in the systematic circulation as GVHD progressively destroyed Paneth cells and reduced GI epithelial barrier function. Paradoxically, GVHD suppressed intestinal REG3γ (the mouse homolog of human REG3α), and the absence of REG3γ in BMT recipients intensified GVHD but did not change the composition of the microbiome. IL-22 administration restored REG3γ production and prevented apoptosis of both intestinal stem cells (ISCs) and Paneth cells, but this protection was completely abrogated in Reg3g-/- mice. In vitro, addition of REG3α reduced the apoptosis of colonic cell lines. Strategies that increase intestinal REG3α/γ to promote crypt regeneration may offer a novel, nonimmunosuppressive approach for GVHD and perhaps for other diseases involving the ISC niche, such as inflammatory bowel disease.


Asunto(s)
Apoptosis , Trasplante de Médula Ósea , Colon/metabolismo , Enfermedad Injerto contra Huésped/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Proteínas Asociadas a Pancreatitis/metabolismo , Células de Paneth/metabolismo , Transducción de Señal , Animales , Supervivencia Celular/genética , Colon/patología , Femenino , Enfermedad Injerto contra Huésped/patología , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/genética , Células de Paneth/patología , Estudios Prospectivos , Trasplante Homólogo
17.
World J Gastroenterol ; 11(22): 3431-40, 2005 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-15948250

RESUMEN

AIM: Recent reports have shown the capacity of mesenchymal stem cells (MSCs) to differentiate into hepatocytes in vitro and in vivo. MSCs administration could repair injured liver, lung, or heart through reducing inflammation, collagen deposition, and remodeling. These results provide a clue to treatment of liver fibrosis. The aim of this study was to investigate the effect of infusion of bone marrow (BM)-derived MSCs on the experimental liver fibrosis in rats. METHODS: MSCs isolated from BM in male Fischer 344 rats were infused to female Wistar rats induced with carbon tetrachloride (CCl4) or dimethylnitrosamine (DMN). There were two random groups on the 42nd d of CCl4:CCl4/MSCs, to infuse a dose of MSCs alone; CCl4/saline, to infuse the same volume of saline as control. There were another three random groups after exposure to DMN: DMN10/MSCs, to infuse the same dose of MSCs on d 10; DMN10/saline, to infuse the same volume of saline on d 10; DMN20/MSCs, to infuse the same dose of MSCs on d 20. The morphological and behavioral changes of rats were monitored everyday. After 4-6 wk of MSCs administration, all rats were killed and fibrosis index were assessed by histopathology and radioimmunoassay. Smooth muscle alpha-actin (alpha-SMA) of liver were tested by immunohistochemistry and quantified by IBAS 2.5 software. Male rats sex determination region on the Y chromosome (sry) gene were explored by PCR. RESULTS: Compared to controls, infusion of MSCs reduced the mortality rates of incidence in CCl4-induced model (10% vs 20%) and in DMN-induced model (20-40% vs 90%). The amount of collagen deposition and alpha-SMA staining was about 40-50% lower in liver of rats with MSCs than that of rats without MSCs. The similar results were observed in fibrosis index. And the effect of the inhibition of fibrogenesis was greater in DMN10/MSCs than in DMN20/MSCs. The sry gene was positive in the liver of rats with MSCs treatment by PCR. CONCLUSION: MSCs treatment can protect against experimental liver fibrosis in CCl4-induced or DMN-induced rats and the mechanisms of the anti-fibrosis by MSCs will be studied further.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Células Madre Hematopoyéticas , Hepatocitos/citología , Cirrosis Hepática/terapia , Animales , Tetracloruro de Carbono , Diferenciación Celular , Dimetilnitrosamina , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/prevención & control , Masculino , Mesodermo/citología , Ratas , Ratas Endogámicas F344
18.
Clin Cancer Res ; 17(4): 771-82, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21088258

RESUMEN

PURPOSE: Stimulation of toll-like receptor-9 (TLR9) by CpG oligodeoxynucleotides (CpG) has been shown to counteract the immunosuppressive microenvironment and to inhibit tumor growth in glioma models. Because TLR9 is located intracellularly, we hypothesized that methods that enhance its internalization may also potentiate its immunostimulatory response. The goal of this study was to evaluate carbon nanotubes (CNT) as a CpG delivery vehicle in brain tumor models. EXPERIMENTAL DESIGN: Functionalized single-walled CNTs were conjugated with CpG (CNT-CpG) and evaluated in vitro and in mice bearing intracranial GL261 gliomas. Flow cytometry was used to assess CNT-CpG uptake and antiglioma immune response. Tumor growth was measured by bioluminescent imaging, histology, and animal survival. RESULTS: CNT-CpG was nontoxic and enhanced CpG uptake both in vitro and intracranial gliomas. CNT-mediated CpG delivery also potentiated proinflammatory cytokine production by primary monocytes. Interestingly, a single intracranial injection of low-dose CNT-CpG (but not free CpG or blank CNT) eradicated intracranial GL261 gliomas in half of tumor-bearing mice. Moreover, surviving animals exhibited durable tumor-free remission (>3 months), and were protected from intracranial tumor rechallenge, demonstrating induction of long-term antitumor immunity. CONCLUSIONS: These findings suggest that CNTs can potentiate CpG immunopotency by enhancing its delivery into tumor-associated inflammatory cells.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Islas de CpG , Glioma/tratamiento farmacológico , Nanoconjugados/uso terapéutico , Nanotubos de Carbono , Polinucleótidos/uso terapéutico , Adyuvantes Inmunológicos/farmacocinética , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Quimiocinas/metabolismo , Citocinas/metabolismo , Femenino , Glioma/inmunología , Glioma/patología , Humanos , Células Asesinas Naturales/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Trasplante de Neoplasias , Polinucleótidos/farmacocinética , Receptor Toll-Like 9/metabolismo , Trasplante Heterólogo , Microambiente Tumoral/efectos de los fármacos
19.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 13(3): 397-403, 2005 Jun.
Artículo en Zh | MEDLINE | ID: mdl-15972129

RESUMEN

To investigate effects of rat bone marrow mesenchymal stem cells (rBMMSC) on hematopoiesis after allo-hematopoietic stem cell transplantation (HSCT), allogeneic BMT model from Fischer 344 rats (RT-1Al) to Wistar rats (RT-1Au) was established; effects of MSCs on hematopoietic reconstitution were studied by survival rate, peripheral blood counts, histological analysis and FACS at day 30 after transplantation. The results showed that (1) MSCs from donor Fisher344 could survive in recipient irradiated by lethal dose and could be found in the thymus, spleen and bone marrow of the recipient at 30 days after cotransplantation with BM by measuring EGFP gene. (2) Cotransplanation of MSCs and BM improved hematopoietic reconstitution. Lymphocyte and platelet counts of peripheral blood in cotransplantation group were higher than those in the control group. Active hematopoiesis and increase of bone marrow nucleated cells were observed in cotransplantation group. MSCs significantly enhanced hematopoiesis of B lymphocyte and megakaryocytopoietic lineages by FACS analysis. It is concluded that (1) MSCs of Fisher344 can be found in the thymus, spleen, bone marrow of the recipients at 30 days after cotransplantion by measuring EGFP gene. (2) hematopoietic reconstitution is significantly enhanced by MSCs cotransplanted with BM.


Asunto(s)
Trasplante de Médula Ósea/métodos , Hematopoyesis/fisiología , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Animales , Diferenciación Celular/fisiología , Citometría de Flujo , Recuento de Linfocitos , Masculino , Células Madre Mesenquimatosas/citología , Modelos Animales , Recuento de Plaquetas , Ratas , Ratas Endogámicas F344 , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA