Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Eur J Immunol ; 47(2): 291-304, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27861793

RESUMO

Although forkhead-box n1 (Foxn1) is a critical thymic epithelial cell regulator in thymus organogenesis, its association with epithelial differentiation and homeostasis in the postnatal and aged thymic microenvironment remains conflicting. Consequently, we have generated a Foxn1eGFP/+ knock-in mouse model that allows for refined investigation of the aging thymic epithelium. This reporter line differs from those previously published in that concomitant expression of enhanced green fluorescent protein enables live cell sorting of Foxn1+ cell populations. Our heterozygotes did not exhibit haploinsufficiency, with Foxn1 expression resembling that of wild-type mice. Comparative analysis between Foxn1 and enhanced green fluorescent protein at both the transcriptional and translational levels revealed co-localization, with progressive down-regulation observed predominantly in the aging cortical epithelium. Supplementation with bone morphogenetic protein (Bmp)-4 enhanced Foxn1 expression and colony forming efficiency in both embryonic and adult progenitor 3D cultures. Strikingly, selective maintenance of immature cortical and medullary epithelial cells was observed which is consistent with the higher Bmp receptor 2 expression levels seen in these progenitor populations. This study demonstrates the significance of our mouse model in unraveling the role of this master regulator in thymus development, homeostasis and aging, providing a faithful reporter system for phenotypic and functional investigations.


Assuntos
Envelhecimento/fisiologia , Células Epiteliais/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Células-Tronco/fisiologia , Timo/fisiologia , Animais , Proteína Morfogenética Óssea 4/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/genética , Homeostase , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Timo/citologia
2.
Gerontology ; 61(6): 504-14, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25765703

RESUMO

Thymic involution during aging is a major reason for the decreased production of naive T cells and reduced immunity. Alterations within the thymic microenvironment, characterized by the loss of function of thymic epithelial cells (TECs) and fibro-adipogenetic transformation, seem to underlie this process, mainly through declining communication between thymic stromal cells and developing thymocytes. Specifically, the signaling mediated by cytokines and hormones secreted by TECs declines during aging. Many therapies based on the manipulation of growth factors and hormones have succeeded in partially recovering the lymphoid compartment and promoting thymic function. However, considering that aging-induced thymic involution is multifactorial, the thymic reestablishment achieved with treatments that target isolated pathways is incomplete and transitory. Here, we discuss the development of three novel approaches for potentially sustained thymic recovery: the induction of sustained forkhead box N1 expression, the activation of endogenous thymic epithelial progenitor cells (TEPCs), and the generation of TEPCs from pluripotent stem cells. Combined approaches targeting both TECs and lymphoid cells will provide a potentially more effective strategy for sustained rejuvenation of the thymus.


Assuntos
Envelhecimento/patologia , Células Epiteliais/fisiologia , Timo/patologia , Envelhecimento/fisiologia , Microambiente Celular , Fatores de Transcrição Forkhead , Humanos , Transdução de Sinais/fisiologia
3.
Nature ; 453(7193): 330-7, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18480814

RESUMO

There is much interest in using embryonic stem cells to regenerate tissues and organs. For this approach to succeed, these stem cells or their derivatives must engraft in patients over the long term. Unless a cell transplant is derived from the patient's own cells, however, the cells will be targeted for rejection by the immune system. Although standard methods for suppressing the immune system achieve some success, rejection of the transplant is inevitable. Emerging approaches to address this issue include 're-educating' the immune system to induce tolerance to foreign cells and reducing the immune targeting of the transplant by administering 'self stem cells' instead of foreign cells, but each of these approaches has associated challenges.


Assuntos
Rejeição de Enxerto/prevenção & controle , Tolerância Imunológica/imunologia , Transplante de Células-Tronco/métodos , Animais , Rejeição de Enxerto/imunologia , Humanos , Células-Tronco/imunologia , Timo/imunologia , Doadores de Tecidos
4.
J Am Soc Nephrol ; 24(4): 573-85, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23393320

RESUMO

Loss of tolerance to neutrophil myeloperoxidase (MPO) underlies the development of ANCA-associated vasculitis and GN, but the mechanisms underlying this loss of tolerance are poorly understood. Here, we assessed the role of the thymus in deletion of autoreactive anti-MPO T cells and the importance of peripheral regulatory T cells in maintaining tolerance to MPO and protecting from GN. Thymic expression of MPO mRNA predominantly localized to medullary thymic epithelial cells. To assess the role of MPO in forming the T cell repertoire and the role of the autoimmune regulator Aire in thymic MPO expression, we compared the effects of immunizing Mpo(-/-) mice, Aire(-/-) mice, and control littermates with MPO. Immunized Mpo(-/-) and Aire(-/-) mice developed significantly more proinflammatory cytokine-producing anti-MPO T cells and higher ANCA titers than control mice. When we triggered GN with a subnephritogenic dose of anti-glomerular basement membrane antibody, Aire(-/-) mice had more severe renal disease than Aire(+/+) mice, consistent with a role for Aire-dependent central deletion in establishing tolerance to MPO. Furthermore, depleting peripheral regulatory T cells in wild-type mice also led to more anti-MPO T cells, higher ANCA titers, and more severe GN after immunization with MPO. Taken together, these results suggest that Aire-dependent central deletion and regulatory T cell-mediated peripheral tolerance both play major roles in establishing and maintaining tolerance to MPO, thereby protecting against the development of anti-MPO GN.


Assuntos
Anticorpos Anticitoplasma de Neutrófilos/imunologia , Glomerulonefrite/imunologia , Tolerância Imunológica/imunologia , Glomérulos Renais/imunologia , Peroxidase/imunologia , Linfócitos T Reguladores/imunologia , Timo/metabolismo , Animais , Anticorpos Anticitoplasma de Neutrófilos/metabolismo , Citocinas/metabolismo , Glomerulonefrite/metabolismo , Camundongos , Peroxidase/metabolismo , RNA Mensageiro , Linfócitos T Reguladores/metabolismo , Timo/imunologia
5.
Cell Metab ; 2024 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-38959897

RESUMO

A mechanistic connection between aging and development is largely unexplored. Through profiling age-related chromatin and transcriptional changes across 22 murine cell types, analyzed alongside previous mouse and human organismal maturation datasets, we uncovered a transcription factor binding site (TFBS) signature common to both processes. Early-life candidate cis-regulatory elements (cCREs), progressively losing accessibility during maturation and aging, are enriched for cell-type identity TFBSs. Conversely, cCREs gaining accessibility throughout life have a lower abundance of cell identity TFBSs but elevated activator protein 1 (AP-1) levels. We implicate TF redistribution toward these AP-1 TFBS-rich cCREs, in synergy with mild downregulation of cell identity TFs, as driving early-life cCRE accessibility loss and altering developmental and metabolic gene expression. Such remodeling can be triggered by elevating AP-1 or depleting repressive H3K27me3. We propose that AP-1-linked chromatin opening drives organismal maturation by disrupting cell identity TFBS-rich cCREs, thereby reprogramming transcriptome and cell function, a mechanism hijacked in aging through ongoing chromatin opening.

6.
Trends Immunol ; 31(5): 191-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20356793

RESUMO

Most of the steps of lymphopoiesis have been elucidated but contentious issues remain, particularly regarding the identity and function of the earliest lymphoid progenitors that leave the bone marrow and seed the thymus. Hematopoiesis is effectively continuous throughout life, but there is a profound decline in immune function with increasing age, driven by thymus involution and severely curtailed B cell development. A key question is whether defects in bone marrow progenitors, such as reduced differentiation and repopulation potential, are the common denominator. While thymic involution temporally precedes overt HSC functional decline, a logical supposition is that the latter exacerbates the former. This review explores this possible link, and concludes that improving bone marrow function is fundamental to sustained thymic regeneration.


Assuntos
Medula Óssea/imunologia , Timo/imunologia , Animais , Diferenciação Celular , Linhagem da Célula , Células-Tronco Hematopoéticas/imunologia , Humanos , Timo/citologia
7.
Crit Rev Immunol ; 31(3): 171-87, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21740349

RESUMO

In essence, normal thymus function involves the production of a broad repertoire of αßT cells capable of responding to foreign antigens with low risk of autoreactivity. Thymic epithelial cells are an essential component of the thymic stromal microenvironment, promoting the growth and export of self-tolerant thymocytes. Autoimmune disease, resulting from a loss of self-tolerance, is clinically and genetically complex, and accordingly has many potential etiological origins. However, it is commonly linked to defects in the thymic epithelial microenvironment. The study of autoimmune-linked thymic stromal dysfunction has indisputably advanced our understanding of T cell tolerance; notably, a field-wide paradigm shift occurred when autoimmune regulator (Aire) was found to drive expression of a multitude of peripheral tissue-restricted antigens in medullary thymic epithelial cells. Many other associations with polygenically controlled autoimmune diseases have been reported but are more difficult to definitively dissect. Paradoxically, immunodeficiency and age-related immunosenescence are also linked with increased autoimmunity. Here we discuss the theoretical basis and the evidence gathered thus far to support these associations.


Assuntos
Envelhecimento/imunologia , Doenças Autoimunes/imunologia , Células Epiteliais/imunologia , Tolerância a Antígenos Próprios/imunologia , Células Estromais/imunologia , Timo , Fatores de Transcrição/metabolismo , Envelhecimento/metabolismo , Antígenos/imunologia , Antígenos/metabolismo , Doenças Autoimunes/metabolismo , Autoimunidade/imunologia , Imunodeficiência de Variável Comum/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Células Epiteliais/metabolismo , Humanos , Células Estromais/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Timo/imunologia , Timo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteína AIRE
8.
J Immunol ; 184(11): 6014-24, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20483779

RESUMO

Cytotoxic antineoplastic therapy is used to treat malignant disease but results in long-term immunosuppression in postpubertal and adult individuals, leading to increased incidence and severity of opportunistic infections. We have previously shown that sex steroid ablation (SSA) reverses immunodeficiencies associated with age and hematopoietic stem cell transplantation in both autologous and allogeneic settings. In this study, we have assessed the effects of SSA by surgical castration on T cell recovery of young male mice following cyclophosphamide treatment as a model for the impact of chemotherapy. SSA increased thymic cellularity, involving all of the thymocyte subsets and early T lineage progenitors. It also induced early repair of damage to the thymic stromal microenvironment, which is crucial to the recovery of a fully functional T cell-based immune system. These functional changes in thymic stromal subsets included enhanced production of growth factors and chemokines important for thymopoiesis, which preceded increases in both thymocyte and stromal cellularity. These effects collectively translated to an increase in peripheral and splenic naive T cells. In conclusion, SSA enhances T cell recovery following cyclophosphamide treatment of mice, at the level of the thymocytes and their stromal niches. This provides a new approach to immune reconstitution following antineoplastic therapy.


Assuntos
Antineoplásicos/toxicidade , Ciclofosfamida/toxicidade , Hormônios Esteroides Gonadais/imunologia , Orquiectomia , Linfócitos T/imunologia , Animais , Separação Celular , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Esteroides , Linfócitos T/efeitos dos fármacos , Timo/citologia , Timo/efeitos dos fármacos , Timo/imunologia
9.
Blood ; 113(1): 204-13, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-18824597

RESUMO

Bone marrow transplantation (BMT) potentially represents a novel therapy for the amelioration and even cure for multiple sclerosis (MS). It has important advantages over immunosuppressive drug treatments because, while effecting broad-based ablation of the immune system and autoreactive cells, it provides an important means for overcoming the resultant immunodeficiency, while possibly restoring self-tolerance. However, both of these benefits are predicated on a functional thymus that undergoes profound age-induced atrophy from puberty. Reversal of thymic atrophy has been achieved by several procedures, including removal of sex steroids by surgical or chemical (LHRH agonist) castration. Using a murine model of MS, experimental autoimmune encephalomyelitis (EAE), we combined BMT with androgen depletion to induce immune regeneration, and investigated the kinetics of increased thymic function on immune reconstitution and disease reduction. We show that androgen depletion significantly increased the efficacy of BMT to ameliorate the clinical signs of EAE while concurrently restoring the periphery with increased naive and regulatory lymphocytic populations. Upon rechallenge, mice with a regenerated thymus had a slower onset of clinical symptoms compared with mice undergoing BMT only. These results suggest that thymic regeneration strategies may be used as a complement to conventional BMT protocols for the treatment of MS.


Assuntos
Androgênios/metabolismo , Transplante de Medula Óssea , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/terapia , Orquiectomia , Timo/imunologia , Animais , Atrofia , Axônios/imunologia , Axônios/patologia , Terapia Combinada , Doenças Desmielinizantes/imunologia , Encefalomielite Autoimune Experimental/cirurgia , Tolerância Imunológica/imunologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/terapia , Regeneração/imunologia , Linfócitos T Reguladores/imunologia , Timo/patologia , Timo/fisiologia
10.
Neuroimmunomodulation ; 18(5): 281-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21952680

RESUMO

The decline in immune function with aging represents a major clinical challenge in many disease conditions. It is manifest in many parameters but is essentially linked to the adaptive immune responses. The prediction would be that abnormalities in both T and B lymphocytes underlie the loss of cellular and humoral capacity, respectively. Somewhat surprisingly, this is not reflected in numerical losses but more in alterations at the population and single cell levels. There is a major reduction in naïve T cells with a proportional increase in memory cells, and also a generally reduced function of these cells. While bone marrow function reduces with age, the most obvious reason for the T cell defects is the severe atrophy of the thymus. This is closely aligned with puberty, thereby implicating a major aetiological role for sex steroids in both thymus and immune system deterioration with age. Accordingly surgical or chemical castration (utilizing luteinizing hormone-releasing hormone) blocks sex steroids resulting in profound rejuvenation of the immune system.


Assuntos
Alergia e Imunologia , Sistema Endócrino/fisiologia , Endocrinologia , Sistema Imunitário/fisiologia , Animais , Humanos
11.
J Immunol ; 182(10): 6247-60, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19414778

RESUMO

A significant decline in immune function is characteristic of aging. Along with the involution of the thymus and associated impaired architecture, which contributes to profound loss of naive T cell production, there are also significant declines in B cell development and the progenitors that support lymphopoiesis. These collectively lead to a reduced peripheral immune repertoire, increase in opportunistic infections, and limited recovery following cytoablation through chemo- or radiotherapy. We have previously shown that sex steroid ablation (SSA) causes a major reversal of age-related thymic atrophy and improves recovery from hematopoietic stem cell transplant. This study focused on the impact of SSA on the B cell compartment and their progenitors in middle-aged and cyclophosphamide-treated mice. In both models, SSA enhanced the number of lymphoid progenitors and developing B cells in the bone marrow (BM) as well as reversing age-related defects in the cycling kinetics of these cells. Enhanced BM lymphopoiesis was reflected in the periphery by an increase in recent BM emigrants as well as immature and mature plasma cells, leading to an enhanced humoral response to challenge by hepatitis B vaccine. In conclusion, SSA improves lymphoid progenitor and B cell recovery from age- and chemotherapy-induced immunodepletion, complimenting the effects on T cells. Since SSA has been achieved clinically for over 25 years, this provides a novel, rational basis for approaching the need for immune recovery in many clinical conditions.


Assuntos
Envelhecimento/imunologia , Antineoplásicos/efeitos adversos , Hormônios Esteroides Gonadais/deficiência , Hormônios Esteroides Gonadais/imunologia , Linfopoese/fisiologia , Animais , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Células-Tronco Hematopoéticas , Linfopoese/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
J Immunol ; 183(11): 7084-94, 2009 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-19890044

RESUMO

Cytotoxic antineoplastic therapy is widely used in the clinic as a treatment for malignant diseases. The treatment itself, however, leads to long-term depletion of the adaptive immune system, which is more pronounced in older patients, predominantly due to thymic atrophy. We and others have previously shown that withdrawal of sex steroids is able to regenerate the aged thymus and enhance recovery from autologous and allogeneic hematopoietic stem cell transplant. In this study we have examined the effects of sex steroid ablation (SSA) on the recovery of lymphopoiesis in the bone marrow (BM) and thymus following treatment with the chemotherapeutic agent cyclophosphamide (Cy) in middle-aged and old mice. Furthermore, we have also examined the impact of this regeneration on peripheral immunity. SSA enhanced the recovery of BM resident hematopoietic stem cells and lymphoid progenitors and promoted lymphopoiesis. Interestingly, Cy alone caused a profound increase in the recently described common lymphoid progenitor 2 (CLP-2) population in the BM. In the thymus, SSA caused a profound increase in cellularity as well as all intrathymic T-lineage progenitors including early T-lineage progenitors (ETPs) and non-canonical T cell progenitors such as the CLP-2. We also found that these transferred into numerical increases in the periphery with enhanced B and T cell numbers. Furthermore, these lymphocytes were found to have an enhanced functional capacity with no perturbation of the TCR repertoire. Taken together, these results provide the basis for the use of SSA in the clinic to enhance treatment outcomes from cytotoxic antineoplastic therapy.


Assuntos
Envelhecimento/fisiologia , Antineoplásicos/efeitos adversos , Ciclofosfamida/efeitos adversos , Hormônios Esteroides Gonadais/deficiência , Linfopoese/fisiologia , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Castração , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Linfopoese/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Timo/citologia , Timo/efeitos dos fármacos , Timo/imunologia
13.
J Immunol ; 183(2): 823-31, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19564346

RESUMO

Immunosuppressive drugs and cytotoxic chemotherapy agents are designed to kill or suppress autoreactive, alloaggressive, or hyperinflammatory T cells, or disseminated malignancies. However, they also cause severe immunological side effects ranging from interrupted thymopoiesis and general immunodeficiency to, paradoxically, autoimmunity. Consistent with the cross-talk between thymocytes and stromal cells, we now show that these common therapeutic agents have major effects on murine thymic epithelial cells (TEC), crucially required to rebuild immunity posttreatment. We show that the immunosuppressant cyclosporine A, which has been linked to a thymus-dependent autoimmune syndrome in some patients, causes extensive loss of autoimmune regulator (Aire(+)) tolerance-inducing MHC class II(high) medullary TEC (mTEC(high)). Post-cyclosporine A, Aire expression was restored within 7 days. Full recovery of the mTEC(high) subset occurred within 10 days and was linked to a decrease in a relatively resistant MHC class II(low) mTEC subset (mTEC(low)), consistent with a previously described precursor-product relationship. Cyclophosphamide and dexamethasone caused more extensive ablation of thymocytes and stromal cells but again severely depleted tolerance-inducing mTEC(high). Together, these data show that Aire(+) mTECs are highly sensitive to damage and that mTEC regeneration follows a conserved pattern regardless of the treatment regimen used.


Assuntos
Ciclofosfamida/farmacologia , Ciclosporina/farmacologia , Dexametasona/farmacologia , Células Epiteliais/imunologia , Tolerância Imunológica/efeitos dos fármacos , Timo/citologia , Animais , Anti-Inflamatórios/farmacologia , Autoimunidade/efeitos dos fármacos , Células Epiteliais/fisiologia , Imunossupressores/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Regeneração , Células Estromais , Fatores de Transcrição , Proteína AIRE
14.
J Immunol ; 182(9): 5846-54, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380833

RESUMO

Posttransplant immunodeficiency, specifically a lack of T cell reconstitution, is a major complication of allogeneic bone marrow transplantation. This immunosuppression results in an increase in morbidity and mortality from infections and very likely contributes to relapse. In this study, we demonstrate that sex steroid ablation using leuprolide acetate, a luteinizing hormone-releasing hormone agonist (LHRHa), increases the number of lymphoid and myeloid progenitor cells in the bone marrow and developing thymocytes in the thymus. Although few differences are observed in the peripheral myeloid compartments, the enhanced thymic reconstitution following LHRHa treatment and allogeneic bone marrow transplantation leads to enhanced peripheral T cell recovery, predominantly in the naive T cell compartment. This results in an increase in T cell function in vivo and in vitro. Graft-versus-host-disease is not exacerbated by LHRHa treatment and graft-versus-tumor activity is maintained. Because LHRHa allows for reversible (and temporary) sex steroid ablation, has a strong safety profile, and has been clinically approved for diseases such as prostate and breast cancer, this drug treatment represents a novel therapeutic approach to reversal of thymic atrophy and enhancement of immunity following immunosuppression.


Assuntos
Transplante de Medula Óssea/imunologia , Hormônio Liberador de Gonadotropina/administração & dosagem , Linfócitos T/efeitos dos fármacos , Linfócitos T/transplante , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/imunologia , Transplante de Medula Óssea/patologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/imunologia , Feminino , Hormônio Liberador de Gonadotropina/agonistas , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/patologia , Doença Enxerto-Hospedeiro/terapia , Efeito Enxerto vs Tumor/efeitos dos fármacos , Efeito Enxerto vs Tumor/imunologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Humanos , Isoantígenos/administração & dosagem , Isoantígenos/genética , Leuprolida/administração & dosagem , Linfopenia/imunologia , Linfopenia/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Linfócitos T/patologia , Timo/citologia , Timo/efeitos dos fármacos , Timo/imunologia
15.
ACS Biomater Sci Eng ; 6(4): 2198-2208, 2020 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33455336

RESUMO

T cells are predominantly produced by the thymus and play a significant role in maintaining our adaptive immune system. Physiological involution of the thymus occurs gradually with age, compromising naive T cell output, which can have severe clinical complications. Also, T cells are utilized as therapeutic agents in cancer immunotherapies. Therefore, there is an increasing need for strategies aimed at generating naive T cells. The majority of in vitro T cell generation studies are performed in two-dimensional (2D) cultures, which ignore the physiological thymic microenvironment and are not scalable; therefore, we applied a new three-dimensional (3D) approach. Here, we use a gelatin-based 3D microgel system for T lineage induction by co-culturing OP9-DL4 cells and mouse fetal-liver-derived hematopoietic stem cells (HSCs). Flow cytometric analysis revealed that microgel co-cultures supported T lineage induction similar to 2D cultures while providing a 3D environment. We also encapsulated mouse embryonic thymic epithelial cells (TECs) within the microgels to provide a defined 3D culture platform. The microgel system supported TEC maintenance and retained their phenotype. Together, these data show that our microgel system has the capacity for TEC maintenance and induction of in vitro T lineage differentiation with potential for scalability.


Assuntos
Diferenciação Celular , Gelatina , Microgéis , Linfócitos T , Animais , Células Epiteliais , Camundongos
16.
Front Immunol ; 11: 302, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194555

RESUMO

One of the main consequences of thymus aging is the decrease in naïve T cell output. This condition accelerates at the onset of puberty, and presents as a major clinical complication for cancer patients who require cytoablative therapy. Specifically, the extensive use of chemotherapeutics, such as cyclophosphamide, in such treatments damage thymic structure and eliminate the existing naïve T cell repertoire. The resulting immunodeficiency can lead to increased incidence of opportunistic infections, tumor growth relapse and/or autoimmune diseases, particularly in older patients. Thus, strategies aimed at rejuvenating the aged thymus following chemotherapeutic damage are required. Previous studies have revealed that sex hormone deprivation in male mice is capable of regenerating the thymic microenvironment following chemotherapy treatment, however, further investigation is crucial to identify gender-based differences, and the molecular mechanisms involved during thymus regeneration. Through phenotypic analyzes, we identified gender-specific alterations in thymocytes and thymic epithelial cell (TEC) subsets from the onset of puberty. By middle-age, females presented with a higher number of thymocytes in comparison to males, yet a decrease in their Aire+ medullary TEC/thymocyte ratio was observed. This reduction could be associated with an increased risk of autoimmune disease in middle-aged women. Given the concurrent increase in female Aire+ cTEC/thymocyte ratio, we proposed that there may be an impediment in Aire+ mTEChi differentiation, and Aire+ cTEChi as its upstream precursor. The regenerative effects of LHRH receptor antagonist, degarelix, on TEC subsets was also less pronounced in middle-aged females compared to males, possibly due to slower progression of thymic involution in the former, which presented with greater TEChi proportions. Furthermore, following cyclophosphamide treatment, degarelix enhanced thymocyte and mature TEC subset recovery, with faster recovery kinetics observed in females. These events were found to involve both reactivation and proliferation of thymic epithelial progenitor cells. Taken together, the findings from this study portray a relationship between gender disparity and thymus aging, and highlight the potential benefits of LHRH receptor antagonist treatment for thymic regeneration. Further research is required, however, to determine how gender may impact on the mechanisms underpinning these events.


Assuntos
Envelhecimento/imunologia , Antineoplásicos Alquilantes/toxicidade , Ciclofosfamida/toxicidade , Células Epiteliais/efeitos dos fármacos , Oligopeptídeos/uso terapêutico , Receptores LHRH/antagonistas & inibidores , Caracteres Sexuais , Timócitos/efeitos dos fármacos , Timo/efeitos dos fármacos , Animais , Atrofia , Contagem de Células , Células Cultivadas , Feminino , Hormônio Foliculoestimulante/sangue , Hormônios Esteroides Gonadais/fisiologia , Hormônio Luteinizante/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oligopeptídeos/farmacologia , Tolerância a Antígenos Próprios , Maturidade Sexual , Células Estromais , Timo/crescimento & desenvolvimento , Timo/patologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Proteína AIRE
17.
Nat Cell Biol ; 22(1): 60-73, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31907413

RESUMO

Defining the ontogeny of the human adaptive immune system during embryogenesis has implications for understanding childhood diseases including leukaemias and autoimmune conditions. Using RAG1:GFP human pluripotent stem cell reporter lines, we examined human T-cell genesis from pluripotent-stem-cell-derived haematopoietic organoids. Under conditions favouring T-cell development, RAG1+ cells progressively upregulated a cohort of recognized T-cell-associated genes, arresting development at the CD4+CD8+ stage. Sort and re-culture experiments showed that early RAG1+ cells also possessed B-cell, myeloid and erythroid potential. Flow cytometry and single-cell-RNA-sequencing data showed that early RAG1+ cells co-expressed the endothelial/haematopoietic progenitor markers CD34, VECAD and CD90, whereas imaging studies identified RAG1+ cells within CD31+ endothelial structures that co-expressed SOX17+ or the endothelial marker CAV1. Collectively, these observations provide evidence for a wave of human T-cell development that originates directly from haemogenic endothelium via a RAG1+ intermediate with multilineage potential.


Assuntos
Endotélio/citologia , Hemangioblastos/citologia , Células-Tronco Hematopoéticas/citologia , Proteínas de Homeodomínio/metabolismo , Células-Tronco Pluripotentes/citologia , Diferenciação Celular/fisiologia , Linhagem Celular , Desenvolvimento Embrionário/fisiologia , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Organoides/citologia
18.
Clin Cancer Res ; 14(4): 1138-49, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18281548

RESUMO

PURPOSE: To determine if temporarily blocking sex steroids prior to stem cell transplantation can increase thymus function and thus enhance the rate of T cell regeneration. EXPERIMENTAL DESIGN: This was a pilot study of luteinizing hormone-releasing hormone agonist (LHRH-A) goserelin given 3 weeks prior to allogeneic or autologous hemopoietic stem cell transplantation and administered up to 3 months posttransplantation. Patients (with or without LHRH-A administration) were assessed from 1 week to 12 months posttransplantation for multiple immunologic variables by flow cytometry (particularly naïve T cells), quantitative PCR to assess T-cell receptor excision circle levels (as a correlate of thymus function), CDR3 length analysis to determine the variability of the TCR repertoire, and in vitro assays to determine functional T cell responses. RESULTS: LHRH-A administration prior to stem cell transplantation significantly increased neutrophil and lymphocyte numbers within the first month of posttransplantation. Most importantly, total and naïve CD4(+) T cell regeneration together with T-cell receptor excision circle production, T cell repertoire regeneration, and peripheral T cell function were also significantly enhanced at multiple time points posttransplant. In addition, an increase in disease-free survival (P = 0.04) was seen in the autologous setting. Although LHRH-A administration increased T cell responses in vitro, it did not exacerbate graft-versus-host disease in the allogeneic setting. CONCLUSIONS: This study provides an important new approach to the improvement of immune reconstitution in patients undergoing hemopoietic stem cell transplantation and may have generic applications in many T cell-based disorders.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Gosserrelina/uso terapêutico , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Linfócitos T/efeitos dos fármacos , Condicionamento Pré-Transplante/métodos , Adolescente , Adulto , Terapia Combinada , Feminino , Citometria de Fluxo , Doença Enxerto-Hospedeiro , Neoplasias Hematológicas/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Autólogo , Transplante Homólogo
19.
Cell Rep ; 27(13): 3887-3901.e4, 2019 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-31242421

RESUMO

A key feature of immune functional impairment with age is the progressive involution of thymic tissue responsible for naive T cell production. In this study, we identify two major phases of thymic epithelial cell (TEC) loss during aging: a block in mature TEC differentiation from the pool of immature precursors, occurring at the onset of puberty, followed by impaired bipotent TEC progenitor differentiation and depletion of Sca-1lo cTEC and mTEC lineage-specific precursors. We reveal that an increase in follistatin production by aging TECs contributes to their own demise. TEC loss occurs primarily through the antagonism of activin A signaling, which we show is required for TEC maturation and acts in dissonance to BMP4, which promotes the maintenance of TEC progenitors. These results support a model in which an imbalance of activin A and BMP4 signaling underpins the degeneration of postnatal TEC maintenance during aging, and its reversal enables the transient replenishment of mature TECs.


Assuntos
Ativinas/metabolismo , Envelhecimento/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Células Epiteliais/metabolismo , Folistatina/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Timo/metabolismo , Animais , Células Epiteliais/citologia , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Células-Tronco/citologia , Timo/citologia
20.
J Immunol Methods ; 329(1-2): 56-66, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17988680

RESUMO

The microenvironment of the thymus consists of functionally discrete niches composed of distinct stromal cell subsets. Clinically relevant changes affecting T-cell differentiation occur within these niches with age and injury caused by irradiation and chemotherapy treatments. The study of thymic stromal cells has been hampered by the technical difficulty in isolating significant numbers of this important population. Here we present an improved protocol for enzymatic isolation of stromal cells that enables comparative flow cytometric analyses and their purification for downstream cellular or molecular analysis. Fractions analyzed throughout enzymatic digestion of the thymus revealed that various stromal subsets are isolated at characteristic intervals. This highlights the importance of pooling all cells isolated from the thymus for numerical and phenotypic analysis to avoid biased representation of subpopulations. We also describe refined magnetic bead separation techniques that yield almost pure preparations of CD45(-) stroma. Sorting of these suspensions using defined markers enabled purification of the major epithelial subsets, confirmed by keratin staining and PCR analysis. This three-step procedure represents a rapid, reproducible method for the unbiased purification of the stromal cells that direct thymic T-cell differentiation.


Assuntos
Separação Celular , Células Epiteliais , Citometria de Fluxo , Células Estromais , Timo/citologia , Animais , Células Epiteliais/química , Células Epiteliais/imunologia , Fatores de Transcrição Forkhead/análise , Fatores de Transcrição Forkhead/genética , Separação Imunomagnética , Queratinas/análise , Antígenos Comuns de Leucócito/análise , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/química , Células Estromais/imunologia , Temperatura , Timo/química , Timo/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA