Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 25(9): 1593-1606, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39112630

RESUMEN

The thymus is essential for establishing adaptive immunity yet undergoes age-related involution that leads to compromised immune responsiveness. The thymus is also extremely sensitive to acute insult and although capable of regeneration, this capacity declines with age for unknown reasons. We applied single-cell and spatial transcriptomics, lineage-tracing and advanced imaging to define age-related changes in nonhematopoietic stromal cells and discovered the emergence of two atypical thymic epithelial cell (TEC) states. These age-associated TECs (aaTECs) formed high-density peri-medullary epithelial clusters that were devoid of thymocytes; an accretion of nonproductive thymic tissue that worsened with age, exhibited features of epithelial-to-mesenchymal transition and was associated with downregulation of FOXN1. Interaction analysis revealed that the emergence of aaTECs drew tonic signals from other functional TEC populations at baseline acting as a sink for TEC growth factors. Following acute injury, aaTECs expanded substantially, further perturbing trophic regeneration pathways and correlating with defective repair of the involuted thymus. These findings therefore define a unique feature of thymic involution linked to immune aging and could have implications for developing immune-boosting therapies in older individuals.


Asunto(s)
Envejecimiento , Células Epiteliales , Factores de Transcripción Forkhead , Regeneración , Timo , Timo/inmunología , Animales , Células Epiteliales/inmunología , Regeneración/inmunología , Ratones , Envejecimiento/inmunología , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Transición Epitelial-Mesenquimal/inmunología , Ratones Endogámicos C57BL , Masculino , Timocitos/inmunología , Timocitos/metabolismo , Femenino , Análisis de la Célula Individual
2.
Dev Cell ; 58(22): 2411-2412, 2023 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-37989079

RESUMEN

T cell development relies on a supportive epithelial microenvironment. Embryonic and postnatal epithelial progenitors have been identified in mice, but not humans. In this issue of Developmental Cell, Raggazzini et al. use scRNAseq, spatial sequencing, and clonogenic assays to identify a putative bipotent TEPC in pediatric human thymic tissue.


Asunto(s)
Células Madre , Timo , Humanos , Ratones , Animales , Niño , Diferenciación Celular , Nicho de Células Madre , Células Epiteliales
3.
bioRxiv ; 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36711570

RESUMEN

Endogenous thymic regeneration is a crucial process that allows for the renewal of immune competence following stress, infection or cytoreductive conditioning. Fully understanding the molecular mechanisms driving regeneration will uncover therapeutic targets to enhance regeneration. We previously demonstrated that high levels of homeostatic apoptosis suppress regeneration and that a reduction in the presence of damage-induced apoptotic thymocytes facilitates regeneration. Here we identified that cell-specific metabolic remodeling after ionizing radiation steers thymocytes towards mitochondrial-driven pyroptotic cell death. We further identified that a key damage-associated molecular pattern (DAMP), ATP, stimulates the cell surface purinergic receptor P2Y2 on cortical thymic epithelial cells (cTECs) acutely after damage, enhancing expression of Foxn1, the critical thymic transcription factor. Targeting the P2Y2 receptor with the agonist UTPγS promotes rapid regeneration of the thymus in vivo following acute damage. Together these data demonstrate that intrinsic metabolic regulation of pyruvate processing is a critical process driving thymus repair and identifies the P2Y2 receptor as a novel molecular therapeutic target to enhance thymus regeneration.

4.
Proc Natl Acad Sci U S A ; 119(17): e2121028119, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35439062

RESUMEN

Secondary lymphoid organs (SLOs) (including the spleen and lymph nodes [LNs]) are critical both for the maintenance of naive T (TN) lymphocytes and for the initiation and coordination of immune responses. How they age, including the exact timing, extent, physiological relevance, and the nature of age-related changes, remains incompletely understood. We used "time stamping" to indelibly mark newly generated naive T cells (also known as recent thymic emigrants) (RTEs) in mice, and followed their presence, phenotype, and retention in SLOs. We found that SLOs involute asynchronously. Skin-draining LNs atrophied by 6 to 9 mo in life, whereas deeper tissue-draining LNs atrophied by 18 to 20 mo, as measured by the loss of both TN numbers and the fibroblastic reticular cell (FRC) network. Time-stamped RTEs at all ages entered SLOs and successfully completed postthymic differentiation, but the capacity of older SLOs to maintain TN numbers was reduced with aging, and that trait did not depend on the age of TNs. However, in SLOs of older mice, these cells exhibited an emigration phenotype (CCR7loS1P1hi), which correlated with an increase of the cells of the same phenotype in the blood. Finally, upon intradermal immunization, RTEs generated in mice barely participated in de novo immune responses and failed to produce well-armed effector cells detectable in blood as early as by 7 to 8 mo of age. These results highlight changes in structure and function of superficial secondary lymphoid organs in laboratory mice that are earlier than expected and are consistent with the long-appreciated reduction of cutaneous immunity with aging.


Asunto(s)
Ganglios Linfáticos , Piel , Envejecimiento , Animales , Atrofia/patología , Ratones , Ratones Endogámicos C57BL , Piel/patología
5.
Aging Cell ; 21(4): e13582, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35289071

RESUMEN

Older humans and animals often exhibit reduced immune responses to infection and vaccination, and this often directly correlates to the numbers and frequency of naive T (Tn) cells. We found such a correlation between reduced numbers of blood CD8+ Tn cells and severe clinical outcomes of West Nile virus (WNV) in both humans naturally exposed to, and mice experimentally infected with, WNV. To examine possible causality, we sought to increase the number of CD8 Tn cells by treating C57BL/6 mice with IL-7 complexes (IL-7C, anti-IL-7 mAb bound to IL-7), shown previously to efficiently increase peripheral T-cell numbers by homeostatic proliferation. T cells underwent robust expansion following IL-7C administration to old mice increasing the number of total T cells (>fourfold) and NS4b:H-2Db -restricted antigen-specific CD8 T cells (twofold). This improved the numbers of NS4b-specific CD8 T cells detected at the peak of the response against WNV, but not survival of WNV challenge. IL-7C-treated old animals also showed no improvement in WNV-specific effector immunity (neutralizing antibody and in vivo T-cell cytotoxicity). To test quantitative limits to which CD8 Tn cell restoration could improve protective immunity, we transferred graded doses of Ag-specific precursors into old mice and showed that injection of 5400 (but not of 1800 or 600) adult naive WNV-specific CD8 T cells significantly increased survival after WNV. These results set quantitative limits to the level of Tn reconstitution necessary to improve immune defense in older organisms and are discussed in light of targets of immune reconstitution.


Asunto(s)
Fiebre del Nilo Occidental , Virus del Nilo Occidental , Animales , Linfocitos T CD8-positivos , Recuento de Células , Interleucina-7 , Ratones , Ratones Endogámicos C57BL
6.
Blood ; 139(25): 3655-3666, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35357432

RESUMEN

Prolonged lymphopenia represents a major clinical problem after cytoreductive therapies such as chemotherapy and the conditioning required for hematopoietic stem cell transplant (HCT), contributing to the risk of infections and malignant relapse. Restoration of T-cell immunity depends on tissue regeneration in the thymus, the primary site of T-cell development, although the capacity of the thymus to repair itself diminishes over its lifespan. However, although boosting thymic function and T-cell reconstitution is of considerable clinical importance, there are currently no approved therapies for treating lymphopenia. Here we found that zinc (Zn) is critically important for both normal T-cell development and repair after acute damage. Accumulated Zn in thymocytes during development was released into the extracellular milieu after HCT conditioning, where it triggered regeneration by stimulating endothelial cell production of BMP4 via the cell surface receptor GPR39. Dietary supplementation of Zn was sufficient to promote thymic function in a mouse model of allogeneic HCT, including enhancing the number of recent thymic emigrants in circulation although direct targeting of GPR39 with a small molecule agonist enhanced thymic function without the need for prior Zn accumulation in thymocytes. Together, these findings not only define an important pathway underlying tissue regeneration but also offer an innovative preclinical approach to treat lymphopenia in HCT recipients.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Linfopenia , Receptores Acoplados a Proteínas G , Animales , Diferenciación Celular , Ratones , Receptores Acoplados a Proteínas G/genética , Timo/metabolismo , Trasplante Homólogo , Zinc/metabolismo
7.
Cell Rep ; 37(1): 109789, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34610317

RESUMEN

The thymus, which is the primary site of T cell development, is particularly sensitive to insult but also has a remarkable capacity for repair. However, the mechanisms orchestrating regeneration are poorly understood, and delayed repair is common after cytoreductive therapies. Here, we demonstrate a trigger of thymic regeneration, centered on detecting the loss of dying thymocytes that are abundant during steady-state T cell development. Specifically, apoptotic thymocytes suppressed production of the regenerative factors IL-23 and BMP4 via TAM receptor signaling and activation of the Rho-GTPase Rac1, the intracellular pattern recognition receptor NOD2, and micro-RNA-29c. However, after damage, when profound thymocyte depletion occurs, this TAM-Rac1-NOD2-miR29c pathway is attenuated, increasing production of IL-23 and BMP4. Notably, pharmacological inhibition of Rac1-GTPase enhanced thymic function after acute damage. These findings identify a complex trigger of tissue regeneration and offer a regenerative strategy for restoring immune competence in patients whose thymic function has been compromised.


Asunto(s)
Apoptosis , Regeneración , Timo/fisiología , Animales , Proteína Morfogenética Ósea 4/metabolismo , Femenino , Interleucina-23/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/genética , Fosfatidilserinas/metabolismo , Pironas/farmacología , Quinolinas/farmacología , Regeneración/efectos de los fármacos , Timocitos/citología , Timocitos/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/metabolismo
8.
Semin Immunopathol ; 43(1): 119-134, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33608819

RESUMEN

T cell recognition of unknown antigens relies on the tremendous diversity of the T cell receptor (TCR) repertoire; generation of which can only occur in the thymus. TCR repertoire breadth is thus critical for not only coordinating the adaptive response against pathogens but also for mounting a response against malignancies. However, thymic function is exquisitely sensitive to negative stimuli, which can come in the form of acute insult, such as that caused by stress, infection, or common cancer therapies; or chronic damage such as the progressive decline in thymic function with age. Whether it be prolonged T cell deficiency after hematopoietic cell transplantation (HCT) or constriction in the breadth of the peripheral TCR repertoire with age; these insults result in poor adaptive immune responses. In this review, we will discuss the importance of thymic function for generation of the TCR repertoire and how acute and chronic thymic damage influences immune health. We will also discuss methods that are used to measure thymic function in patients and strategies that have been developed to boost thymic function.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Linfocitos T , Antígenos , Comunicación Celular , Humanos , Receptores de Antígenos de Linfocitos T/genética
10.
Front Immunol ; 11: 1745, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32903477

RESUMEN

Even though the thymus is exquisitely sensitive to acute insults like infection, shock, or common cancer therapies such as cytoreductive chemo- or radiation-therapy, it also has a remarkable capacity for repair. This phenomenon of endogenous thymic regeneration has been known for longer even than its primary function to generate T cells, however, the underlying mechanisms controlling the process have been largely unstudied. Although there is likely continual thymic involution and regeneration in response to stress and infection in otherwise healthy people, acute and profound thymic damage such as that caused by common cancer cytoreductive therapies or the conditioning regimes as part of hematopoietic cell transplantation (HCT), leads to prolonged T cell deficiency; precipitating high morbidity and mortality from opportunistic infections and may even facilitate cancer relapse. Furthermore, this capacity for regeneration declines with age as a function of thymic involution; which even at steady state leads to reduced capacity to respond to new pathogens, vaccines, and immunotherapy. Consequently, there is a real clinical need for strategies that can boost thymic function and enhance T cell immunity. One approach to the development of such therapies is to exploit the processes of endogenous thymic regeneration into novel pharmacologic strategies to boost T cell reconstitution in clinical settings of immune depletion such as HCT. In this review, we will highlight recent work that has revealed the mechanisms by which the thymus is capable of repairing itself and how this knowledge is being used to develop novel therapies to boost immune function.


Asunto(s)
Proliferación Celular , Células Epiteliales/patología , Regeneración , Timocitos/patología , Timo/fisiopatología , Animales , Comunicación Celular , Proliferación Celular/efectos de los fármacos , Microambiente Celular , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Humanos , Factores Inmunológicos/uso terapéutico , Regeneración/efectos de los fármacos , Transducción de Señal , Timocitos/efectos de los fármacos , Timocitos/inmunología , Timocitos/metabolismo , Timo/efectos de los fármacos , Timo/inmunología , Timo/patología
11.
Cancer Cell ; 35(3): 489-503.e8, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30889382

RESUMEN

Many potential targets for CAR-T cells in solid tumors are expressed in some normal tissues, raising concern for off-tumor toxicity. Following lymphodepletion, CAR-T cells targeting the tumor-associated antigen ROR1 lysed tumors in mice but induced lethal bone marrow failure due to recognition of ROR1+ stromal cells. To improve selectivity, we engineered T cells with synthetic Notch (synNotch) receptors specific for EpCAM or B7-H3, which are expressed on ROR1+ tumor cells but not ROR1+ stromal cells. SynNotch receptors induced ROR1 CAR expression selectively within the tumor, resulting in tumor regression without toxicity when tumor cells were segregated from, but not when co-localized with, normal ROR1+ cells. This strategy, thus, permits safe targeting of tumors that are sufficiently separated from normal cells.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Receptores Quiméricos de Antígenos/inmunología , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Células K562 , Ratones , Neoplasias/inmunología , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Blood ; 132(26): 2763-2774, 2018 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-30381375

RESUMEN

Nuclear factor erythroid-derived 2-like 2 (Nrf2) is a ubiquitously expressed transcription factor that is well known for its role in regulating the cellular redox pathway. Although there is mounting evidence suggesting a critical role for Nrf2 in hematopoietic stem cells and innate leukocytes, little is known about its involvement in T-cell biology. In this study, we identified a novel role for Nrf2 in regulating alloreactive T-cell function during allogeneic hematopoietic cell transplantation (allo-HCT). We observed increased expression and nuclear translocation of Nrf2 upon T-cell activation in vitro, especially in CD4+ donor T cells after allo-HCT. Allo-HCT recipients of Nrf2 -/- donor T cells had significantly less acute graft-versus-host disease (GVHD)-induced mortality, morbidity, and pathology. This reduction in GVHD was associated with the persistence of Helios+ donor regulatory T cells in the allograft, as well as defective upregulation of the gut-homing receptor LPAM-1 on alloreactive CD8+ T cells. Additionally, Nrf2 -/- donor CD8+ T cells demonstrated intact cytotoxicity against allogeneic target cells. Tumor-bearing allo-HCT recipients of Nrf2 -/- donor T cells had overall improved survival as a result of preserved graft-versus-tumor activity and reduced GVHD activity. Our findings characterized a previously unrecognized role for Nrf2 in T-cell function, as well as revealed a novel therapeutic target to improve the outcomes of allo-HCT.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Activación de Linfocitos , Factor 2 Relacionado con NF-E2/inmunología , Neoplasias Experimentales/inmunología , Enfermedad Aguda , Aloinjertos , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia
14.
Sci Immunol ; 3(19)2018 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-29330161

RESUMEN

The thymus is not only extremely sensitive to damage but also has a remarkable ability to repair itself. However, the mechanisms underlying this endogenous regeneration remain poorly understood, and this capacity diminishes considerably with age. We show that thymic endothelial cells (ECs) comprise a critical pathway of regeneration via their production of bone morphogenetic protein 4 (BMP4) ECs increased their production of BMP4 after thymic damage, and abrogating BMP4 signaling or production by either pharmacologic or genetic inhibition impaired thymic repair. EC-derived BMP4 acted on thymic epithelial cells (TECs) to increase their expression of Foxn1, a key transcription factor involved in TEC development, maintenance, and regeneration, and its downstream targets such as Dll4, a key mediator of thymocyte development and regeneration. These studies demonstrate the importance of the BMP4 pathway in endogenous tissue regeneration and offer a potential clinical approach to enhance T cell immunity.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Células Endoteliales/metabolismo , Regeneración/fisiología , Timo/metabolismo , Timo/fisiología , Animales , Proliferación Celular/fisiología , Células Endoteliales/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Femenino , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Células Madre/metabolismo , Células Madre/fisiología , Linfocitos T/metabolismo , Linfocitos T/fisiología
15.
Nat Med ; 24(2): 239-246, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29309056

RESUMEN

There is a substantial unmet clinical need for new strategies to protect the hematopoietic stem cell (HSC) pool and regenerate hematopoiesis after radiation injury from either cancer therapy or accidental exposure. Increasing evidence suggests that sex hormones, beyond their role in promoting sexual dimorphism, regulate HSC self-renewal, differentiation, and proliferation. We and others have previously reported that sex-steroid ablation promotes bone marrow (BM) lymphopoiesis and HSC recovery in aged and immunodepleted mice. Here we found that a luteinizing hormone (LH)-releasing hormone antagonist (LHRH-Ant), currently in wide clinical use for sex-steroid inhibition, promoted hematopoietic recovery and mouse survival when administered 24 h after an otherwise-lethal dose of total-body irradiation (L-TBI). Unexpectedly, this protective effect was independent of sex steroids and instead relied on suppression of LH levels. Human and mouse long-term self-renewing HSCs (LT-HSCs) expressed high levels of the LH/choriogonadotropin receptor (LHCGR) and expanded ex vivo when stimulated with LH. In contrast, the suppression of LH after L-TBI inhibited entry of HSCs into the cell cycle, thus promoting HSC quiescence and protecting the cells from exhaustion. These findings reveal a role of LH in regulating HSC function and offer a new therapeutic approach for hematopoietic regeneration after hematopoietic injury.


Asunto(s)
Autorrenovación de las Células/genética , Células Madre Hematopoyéticas/metabolismo , Hormona Luteinizante/metabolismo , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Proliferación Celular/efectos de la radiación , Autorrenovación de las Células/efectos de los fármacos , Autorrenovación de las Células/efectos de la radiación , Hormona Liberadora de Gonadotropina/antagonistas & inhibidores , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Hormona Luteinizante/farmacología , Ratones , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Receptores de HL/genética , Regeneración/efectos de los fármacos , Regeneración/genética , Regeneración/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Irradiación Corporal Total
16.
J Exp Med ; 214(9): 2733-2758, 2017 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-28798028

RESUMEN

In the thymus, hematopoietic progenitors commit to the T cell lineage and undergo sequential differentiation to generate diverse T cell subsets, including major histocompatibility complex (MHC)-restricted αß T cell receptor (TCR) T cells and non-MHC-restricted γδ TCR T cells. The factors controlling precursor commitment and their subsequent maturation and specification into αß TCR versus γδ TCR T cells remain unclear. Here, we show that the tyrosine phosphatase PTPN2 attenuates STAT5 (signal transducer and activator of transcription 5) signaling to regulate T cell lineage commitment and SRC family kinase LCK and STAT5 signaling to regulate αß TCR versus γδ TCR T cell development. Our findings identify PTPN2 as an important regulator of critical checkpoints that dictate the commitment of multipotent precursors to the T cell lineage and their subsequent maturation into αß TCR or γδ TCR T cells.


Asunto(s)
Linaje de la Célula/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/fisiología , Receptores de Antígenos de Linfocitos T gamma-delta/fisiología , Linfocitos T/fisiología , Animales , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Multipotentes/fisiología , Factor de Transcripción STAT5/fisiología
17.
Blood ; 130(7): 933-942, 2017 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-28607133

RESUMEN

Graft-versus-host disease (GVHD) and posttransplant immunodeficiency are frequently related complications of allogeneic hematopoietic transplantation. Alloreactive donor T cells can damage thymic epithelium, thus limiting new T-cell development. Although the thymus has a remarkable capacity to regenerate after injury, endogenous thymic regeneration is impaired in GVHD. The mechanisms leading to this regenerative failure are largely unknown. Here we demonstrate in experimental mouse models that GVHD results in depletion of intrathymic group 3 innate lymphoid cells (ILC3s) necessary for thymic regeneration. Loss of thymic ILC3s resulted in deficiency of intrathymic interleukin-22 (IL-22) compared with transplant recipients without GVHD, thereby inhibiting IL-22-mediated protection of thymic epithelial cells (TECs) and impairing recovery of thymopoiesis. Conversely, abrogating IL-21 receptor signaling in donor T cells and inhibiting the elimination of thymic ILCs improved thymopoiesis in an IL-22-dependent fashion. We found that the thymopoietic impairment in GVHD associated with loss of ILCs could be improved by restoration of IL-22 signaling. Despite uninhibited alloreactivity, exogenous IL-22 administration posttransplant resulted in increased recovery of thymopoiesis and development of new thymus-derived peripheral T cells. Our study highlights the role of innate immune function in thymic regeneration and restoration of adaptive immunity posttransplant. Manipulation of the ILC-IL-22-TEC axis may be useful for augmenting immune reconstitution after clinical hematopoietic transplantation and other settings of T-cell deficiency.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Inmunidad Innata , Linfocitos/inmunología , Timo/inmunología , Animales , Trasplante de Médula Ósea , Interleucinas/deficiencia , Interleucinas/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Transducción de Señal , Linfocitos T Reguladores/inmunología , Interleucina-22
18.
Sci Transl Med ; 9(386)2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28424327

RESUMEN

The molecular pathways that regulate the tissue repair function of type I interferon (IFN-I) during acute tissue damage are poorly understood. We describe a protective role for IFN-I and the RIG-I/MAVS signaling pathway during acute tissue damage in mice. Mice lacking mitochondrial antiviral-signaling protein (MAVS) were more sensitive to total body irradiation- and chemotherapy-induced intestinal barrier damage. These mice developed worse graft-versus-host disease (GVHD) in a preclinical model of allogeneic hematopoietic stem cell transplantation (allo-HSCT) than did wild-type mice. This phenotype was not associated with changes in the intestinal microbiota but was associated with reduced gut epithelial integrity. Conversely, targeted activation of the RIG-I pathway during tissue injury promoted gut barrier integrity and reduced GVHD. Recombinant IFN-I or IFN-I expression induced by RIG-I promoted growth of intestinal organoids in vitro and production of the antimicrobial peptide regenerating islet-derived protein 3 γ (RegIIIγ). Our findings were not confined to RIG-I/MAVS signaling because targeted engagement of the STING (stimulator of interferon genes) pathway also protected gut barrier function and reduced GVHD. Consistent with this, STING-deficient mice suffered worse GVHD after allo-HSCT than did wild-type mice. Overall, our data suggest that activation of either RIG-I/MAVS or STING pathways during acute intestinal tissue injury in mice resulted in IFN-I signaling that maintained gut epithelial barrier integrity and reduced GVHD severity. Targeting these pathways may help to prevent acute intestinal injury and GVHD during allogeneic transplantation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína 58 DEAD Box/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteína 58 DEAD Box/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Trasplante de Células Madre Hematopoyéticas , Interferón Tipo I/metabolismo , Intestinos/efectos de la radiación , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/fisiología , Organoides/citología , Organoides/metabolismo , Reacción en Cadena de la Polimerasa , Transducción de Señal/fisiología , Trasplante Homólogo
19.
Nat Med ; 23(2): 242-249, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28067900

RESUMEN

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potentially curative therapy for hematological malignancies. However, graft-versus-host disease (GVHD) and relapse after allo-HSCT remain major impediments to the success of allo-HSCT. Chimeric antigen receptors (CARs) direct tumor cell recognition of adoptively transferred T cells. CD19 is an attractive CAR target, which is expressed in most B cell malignancies, as well as in healthy B cells. Clinical trials using autologous CD19-targeted T cells have shown remarkable promise in various B cell malignancies. However, the use of allogeneic CAR T cells poses a concern in that it may increase risk of the occurrence of GVHD, although this has not been reported in selected patients infused with donor-derived CD19 CAR T cells after allo-HSCT. To understand the mechanism whereby allogeneic CD19 CAR T cells may mediate anti-lymphoma activity without causing a significant increase in the incidence of GVHD, we studied donor-derived CD19 CAR T cells in allo-HSCT and lymphoma models in mice. We demonstrate that alloreactive T cells expressing CD28-costimulated CD19 CARs experience enhanced stimulation, resulting in the progressive loss of both their effector function and proliferative potential, clonal deletion, and significantly decreased occurrence of GVHD. Concurrently, the other CAR T cells that were present in bulk donor T cell populations retained their anti-lymphoma activity in accordance with the requirement that both the T cell receptor (TCR) and CAR be engaged to accelerate T cell exhaustion. In contrast, first-generation and 4-1BB-costimulated CAR T cells increased the occurrence of GVHD. These findings could explain the reduced risk of GVHD occurring with cumulative TCR and CAR signaling.


Asunto(s)
Reacción Injerto-Huésped/inmunología , Efecto Injerto vs Tumor/inmunología , Trasplante de Células Madre Hematopoyéticas , Linfoma/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Ligando 4-1BB/inmunología , Traslado Adoptivo , Animales , Antígenos CD19/metabolismo , Linfocitos B/inmunología , Antígenos CD28 , Quimera , Citocinas/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Enfermedad Injerto contra Huésped/inmunología , Ratones , Linfocitos T/metabolismo , Trasplante Homólogo
20.
Nat Commun ; 7: 11492, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27173585

RESUMEN

Parental behavioural traits can be transmitted by non-genetic mechanisms to the offspring. Although trait transmission via sperm has been extensively researched, epidemiological studies indicate the exclusive/prominent maternal transmission of many non-genetic traits. Since maternal conditions impact the offspring during gametogenesis and through fetal/early-postnatal life, the resultant phenotype is likely the aggregate of consecutive germline and somatic effects; a concept that has not been previously studied. Here, we dissected a complex maternally transmitted phenotype, reminiscent of comorbid generalized anxiety/depression, to elementary behaviours/domains and their transmission mechanisms in mice. We show that four anxiety/stress-reactive traits are transmitted via independent iterative-somatic and gametic epigenetic mechanisms across multiple generations. Somatic/gametic transmission alters DNA methylation at enhancers within synaptic genes whose functions can be linked to the behavioural traits. Traits have generation-dependent penetrance and sex specificity resulting in pleiotropy. A transmission-pathway-based concept can refine current inheritance models of psychiatric diseases and facilitate the development of better animal models and new therapeutic approaches.


Asunto(s)
Conducta Animal/fisiología , Epigénesis Genética , Células Germinativas/fisiología , Herencia Materna/fisiología , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Ansiedad/genética , Ansiedad/psicología , Metilación de ADN/genética , Modelos Animales de Enfermedad , Femenino , Gametogénesis/fisiología , Impresión Genómica/fisiología , Hipotermia/inducido químicamente , Hipotermia/genética , Hipotermia/psicología , Masculino , Metabolómica/métodos , Ratones , Ratones Noqueados , Modelos Animales , Penetrancia , Fenotipo , Receptor de Serotonina 5-HT1A/genética , Receptor de Serotonina 5-HT1A/metabolismo , Agonistas de Receptores de Serotonina/farmacología , Estrés Psicológico/genética , Estrés Psicológico/psicología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...