Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Development ; 149(10)2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35587733

RESUMO

The thymus stroma constitutes a fundamental microenvironment for T-cell generation. Despite the chief contribution of thymic epithelial cells, recent studies emphasize the regulatory role of mesenchymal cells in thymic function. Mesenchymal progenitors are suggested to exist in the postnatal thymus; nonetheless, an understanding of their nature and the mechanism controlling their homeostasis in vivo remains elusive. We resolved two new thymic fibroblast subsets with distinct developmental features. Whereas CD140αß+GP38+SCA-1- cells prevailed in the embryonic thymus and declined thereafter, CD140αß+GP38+SCA-1+ cells emerged in the late embryonic period and predominated in postnatal life. The fibroblastic-associated transcriptional programme was upregulated in CD140αß+GP38+SCA-1+ cells, suggesting that they represent a mature subset. Lineage analysis showed that CD140αß+GP38+SCA-1+ maintained their phenotype in thymic organoids. Strikingly, CD140αß+GP38+SCA-1- generated CD140αß+GP38+SCA-1+, inferring that this subset harboured progenitor cell activity. Moreover, the abundance of CD140αß+GP38+SCA-1+ fibroblasts was gradually reduced in Rag2-/- and Rag2-/-Il2rg-/- thymi, indicating that fibroblast maturation depends on thymic crosstalk. Our findings identify CD140αß+GP38+SCA-1- as a source of fibroblast progenitors and define SCA-1 as a marker for developmental stages of thymic fibroblast differentiation.


Assuntos
Células-Tronco , Linfócitos T , Animais , Diferenciação Celular , Células Epiteliais , Fibroblastos , Camundongos , Timo
2.
Eur J Immunol ; 53(3): e2250202, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36642953

RESUMO

Within the thymus, thymic epithelial cells (TECs) provide a dedicated niche for the selection of functional T cells expressing a highly variable and self-tolerant T-cell receptor (TCR) repertoire. In this minireview, we start by summarizing recent studies that have improved our understanding on the composition of cortical TEC and medullary TEC microenvironments. Next, we focus on the molecular processes that control the function of TECs in T-cell selection. In particular, we discuss the role of cortical TECs in positive selection and the pathways employed by these cells to generate and present selecting self-peptides:MHC II complexes. Several studies have underscored the role of the ß5t-containing thymoproteasome in the production of unique MHC I-bound peptides critical for CD8 T-cell selection. Contrarily, the identity of the molecular determinants that regulate the generation of MHC II-bound self-peptides capable of positive selecting CD4 T cells is far more uncertain. We highlight recent advances that interconnect the autophagy-lysosomal pathway, the presentation of specific sets of self-peptide:MHC II complexes, and the diversification of CD4 TCR repertoire. Lastly, we discuss how these findings may open up new avenues for deciphering the identity of the MHC I and MHC II ligandome in the thymus.


Assuntos
Células Epiteliais , Timo , Peptídeos/metabolismo , Linfócitos T CD4-Positivos , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo
3.
Eur J Immunol ; 51(2): 311-318, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32845012

RESUMO

Autoimmune regulator+ (Aire) medullary thymic epithelial cells (mTECs) play a critical role in tolerance induction. Several studies demonstrated that Aire+ mTECs differentiate further into Post-Aire cells. Yet, the identification of terminal stages of mTEC maturation depends on unique fate-mapping mouse models. Herein, we resolve this limitation by segmenting the mTEChi (MHCIIhi CD80hi ) compartment into mTECA/hi (CD24- Sca1- ), mTECB/hi (CD24+ Sca1- ), and mTECC/hi (CD24+ Sca1+ ). While mTECA/hi included mostly Aire-expressing cells, mTECB/hi contained Aire+ and Aire- cells and mTECC/hi were mainly composed of cells lacking Aire. The differential expression pattern of Aire led us to investigate the precursor-product relationship between these subsets. Strikingly, transcriptomic analysis of mTECA/hi , mTECB/hi , and mTECC/hi sequentially mirrored the specific genetic program of Early-, Late- and Post-Aire mTECs. Corroborating their Post-Aire nature, mTECC/hi downregulated the expression of tissue-restricted antigens, acquired traits of differentiated keratinocytes, and were absent in Aire-deficient mice. Collectively, our findings reveal a new and simple blueprint to survey late stages of mTEC differentiation.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Queratinócitos/imunologia , Timo/imunologia , Fatores de Transcrição/genética , Animais , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/imunologia , Proteína AIRE
4.
Trends Immunol ; 39(1): 2-5, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29236672

RESUMO

Medullary thymic epithelial cells (mTECs) play a central role in T cell tolerance. However, how the mTEC compartment is maintained remains elusive. We review recent discoveries on new transcription factors involved in mTEC homeostasis and discuss the possibility that their actions might be facilitated by the unique biology of mTECs.


Assuntos
Células Epiteliais/fisiologia , Tolerância Imunológica , Linfócitos T/fisiologia , Timo/fisiologia , Animais , Diferenciação Celular , Hematopoese , Humanos , Camundongos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Biomacromolecules ; 21(12): 4771-4780, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-33238090

RESUMO

Thymic epithelial cells (TECs) are the main regulators of T lymphocyte development and selection, requiring a three-dimensional (3D) environment to properly perform these biological functions. The aim of this work was to develop a 3D culture substrate that allows the survival and proliferation of TECs. Thus, electrospun fibrous meshes (eFMs) were functionalized with fibronectin, one of the major extracellular matrix (ECM) proteins of the thymus. For that, highly porous eFMs were activated using oxygen plasma treatment followed by amine insertion, which allows the immobilization of fibronectin through EDC/NHS chemistry. The medullary TECs presented increased proliferation, viability, and protein synthesis when cultured on fibronectin-functionalized eFMs (FN-eFMs). These cells showed a spread morphology, with increased migration toward the inner layers of FN-eFMs and the production of thymic ECM proteins, such as collagen type IV and laminin. These results suggest that FN-eFMs are an effective substrate for supporting thymic cell cultures.


Assuntos
Células Epiteliais , Fibronectinas , Animais , Diferenciação Celular , Células Cultivadas , Matriz Extracelular , Proteínas da Matriz Extracelular , Laminina , Camundongos
6.
J Immunol ; 200(4): 1389-1398, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29321277

RESUMO

Despite the well-documented effect of castration in thymic regeneration, the singular contribution of the bone marrow (BM) versus the thymus to this process remains unclear. The chief role of IL-7 in pre- and intrathymic stages of T lymphopoiesis led us to investigate the impact of disrupting this cytokine during thymic rebound induced by androgen blockade. We found that castration promoted thymopoiesis in young and aged wild-type mice. In contrast, only young germline IL-7-deficient (Il7-/- ) mice consistently augmented thymopoiesis after castration. The increase in T cell production was accompanied by the expansion of the sparse medullary thymic epithelial cell and the peripheral T cell compartment in young Il7-/- mice. In contrast to young Il7-/- and wild-type mice, the poor thymic response of aged Il7-/- mice after castration was associated with a defect in the expansion of BM hematopoietic progenitors. These findings suggest that BM-derived T cell precursors contribute to thymic rebound driven by androgen blockade. To assess the role of IL-7 within the thymus, we generated mice with conditional deletion of IL-7 (Il7 conditional knockout [cKO]) in thymic epithelial cells. As expected, Il7cKO mice presented a profound defect in T cell development while maintaining an intact BM hematopoietic compartment across life. Unlike Il7-/- mice, castration promoted the expansion of BM precursors and enhanced thymic activity in Il7cKO mice independently of age. Our findings suggest that the mobilization of BM precursors acts as a prime catalyst of castration-driven thymopoiesis.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Linfopoese/fisiologia , Timo/imunologia , Androgênios/metabolismo , Animais , Células da Medula Óssea/imunologia , Castração , Diferenciação Celular/fisiologia , Interleucina-7/deficiência , Interleucina-7/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Timo/citologia
7.
Blood ; 130(4): 478-488, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28559356

RESUMO

Thymic epithelial cells (TECs) provide crucial microenvironments for T-cell development and tolerance induction. As the regular function of the thymus declines with age, it is of fundamental and clinical relevance to decipher new determinants that control TEC homeostasis in vivo. Beyond its recognized tumor suppressive function, p53 controls several immunoregulatory pathways. To study the cell-autonomous role of p53 in thymic epithelium functioning, we developed and analyzed mice with conditional inactivation of Trp53 in TECs (p53cKO). We report that loss of p53 primarily disrupts the integrity of medullary TEC (mTEC) niche, a defect that spreads to the adult cortical TEC compartment. Mechanistically, we found that p53 controls specific and broad programs of mTEC differentiation. Apart from restraining the expression and responsiveness of the receptor activator of NF-κB (RANK), which is central for mTEC differentiation, deficiency of p53 in TECs altered multiple functional modules of the mTEC transcriptome, including tissue-restricted antigen expression. As a result, p53cKO mice presented premature defects in mTEC-dependent regulatory T-cell differentiation and thymocyte maturation, which progressed to a failure in regular and regenerative thymopoiesis and peripheral T-cell homeostasis in the adulthood. Lastly, peripheral signs of altered immunological tolerance unfold in mutant mice and in immunodeficient mice that received p53cKO-derived thymocytes. Our findings position p53 as a novel molecular determinant of thymic epithelium function throughout life.


Assuntos
Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Linfócitos T Reguladores/imunologia , Timócitos/imunologia , Proteína Supressora de Tumor p53/imunologia , Animais , Diferenciação Celular/genética , Células Epiteliais/citologia , Camundongos , Camundongos Knockout , Linfócitos T Reguladores/citologia , Timócitos/citologia , Timo , Proteína Supressora de Tumor p53/genética
8.
J Immunol ; 199(4): 1429-1439, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28687660

RESUMO

IFN-γ is known to be predominantly produced by lymphoid cells such as certain subsets of T cells, NK cells, and other group 1 innate lymphoid cells. In this study, we used IFN-γ reporter mouse models to search for additional cells capable of secreting this cytokine. We identified a novel and rare population of nonconventional IFN-γ-producing cells of hematopoietic origin that were characterized by the expression of Thy1.2 and the lack of lymphoid, myeloid, and NK lineage markers. The expression of IFN-γ by this population was higher in the liver and lower in the spleen. Furthermore, these cells were present in mice lacking both the Rag2 and the common γ-chain (γc) genes (Rag2-/-γc-/-), indicating their innate nature and their γc cytokine independence. Rag2-/-γc-/- mice are as resistant to Mycobacterium avium as Rag2-/- mice, whereas Rag2-/- mice lacking IFN-γ are more susceptible than either Rag2-/- or Rag2-/-γc-/- These lineage-negative CD45+/Thy1.2+ cells are found within the mycobacterially induced granulomatous structure in the livers of infected Rag2-/-γc-/- animals and are adjacent to macrophages that expressed inducible NO synthase, suggesting a potential protective role for these IFN-γ-producing cells. Accordingly, Thy1.2-specific mAb administration to infected Rag2-/-γc-/- animals increased M. avium growth in the liver. Overall, our results demonstrate that a population of Thy1.2+ non-NK innate-like cells present in the liver expresses IFN-γ and can confer protection against M. avium infection in immunocompromised mice.


Assuntos
Células-Tronco Hematopoéticas/imunologia , Imunidade Inata , Interferon gama/biossíntese , Interferon gama/genética , Subunidade gama Comum de Receptores de Interleucina/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Granuloma/imunologia , Granuloma/microbiologia , Hospedeiro Imunocomprometido/imunologia , Interferon gama/imunologia , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Células Matadoras Naturais/imunologia , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Fígado/citologia , Fígado/imunologia , Fígado/microbiologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium avium/crescimento & desenvolvimento , Mycobacterium avium/imunologia , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Baço/citologia , Baço/imunologia , Antígenos Thy-1/genética , Antígenos Thy-1/imunologia
9.
Carcinogenesis ; 39(12): 1463-1476, 2018 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-30256907

RESUMO

T-cell acute lymphoblastic leukemia (T-ALL) and T-lymphoblastic lymphomas (T-LBL) are aggressive malignancies of thymocytes. The role of thymic microenvironmental cells and stromal factors in thymocyte malignant transformation and T-ALL development remains little explored. Here, using the TEL-JAK2 transgenic (TJ2-Tg) mouse model of T-ALL/LBL, which is driven by constitutive JAK/STAT signaling and characterized by the acquisition of Notch1 mutations, we sought to identify stromal cell alterations associated with thymic leukemogenesis. Immunofluorescence analyses showed that thymic lymphomas presented epithelial areas characterized by keratin (Krt) 5 and Krt8 expression, adjacently to epithelial-free areas negative for Krt expression. Both areas contained abundant laminin (extracellular matrix) and ER-TR7+ (fibroblasts) CD31+ (endothelial) and CD11c+ (dendritic) cells. Besides Krt5, Krt-positive areas harbored medullary thymic epithelial cells (TECs) labeled by Ulex europaeus agglutinin-1. By performing flow cytometry and RNA sequencing analyses of thymic lymphomas, we observed an enrichment in medullary TEC markers in detriment of cortical TEC markers. To assess whether TECs are important for T-ALL/LBL development, we generated TJ2-Tg mice heterozygous for the FoxN1 transcription factor nude null mutation (Foxn1+/nu). Strikingly, in TJ2-Tg;Foxn1+/nu compound mice, both emergence of malignant cells in preleukemic thymi and overt T-ALL onset were significantly delayed. Moreover, in transplantation assays, leukemic cell expansion within the thymus of recipient Foxn1+/nu mice was reduced as compared with control littermates. Since thymopoesis is largely normal in Foxn1+/nu mice, these results indicate that FoxN1 haploinsufficiency in TECs has a more profound impact in thymic leukemogenesis.


Assuntos
Carcinogênese/patologia , Células Epiteliais/patologia , Fatores de Transcrição Forkhead/genética , Leucemia de Células T/genética , Leucemia de Células T/patologia , Timo/patologia , Animais , Biomarcadores Tumorais , Diferenciação Celular/genética , Modelos Animais de Doenças , Epitélio/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos/genética , Mutação/genética , Análise de Sequência de RNA/métodos , Transdução de Sinais/genética , Células Estromais/patologia
10.
Eur J Immunol ; 47(6): 958-969, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28318017

RESUMO

Cortical (cTEC) and medullary (mTEC) thymic epithelial cells establish key microenvironments for T-cell differentiation and arise from thymic epithelial cell progenitors (TEP). However, the nature of TEPs and the mechanism controlling their stemness in the postnatal thymus remain poorly defined. Using TEC clonogenic assays as a surrogate to survey TEP activity, we found that a fraction of cTECs generates specialized clonal-derived colonies, which contain cells with sustained colony-forming capacity (ClonoTECs). These ClonoTECs are EpCAM+MHCII-Foxn1lo cells that lack traits of mature cTECs or mTECs but co-express stem-cell markers, including CD24 and Sca-1. Supportive of their progenitor identity, ClonoTECs reintegrate within native thymic microenvironments and generate cTECs or mTECs in vivo. Strikingly, the frequency of cTECs with the potential to generate ClonoTECs wanes between the postnatal and young adult immunocompetent thymus, but it is sustained in alymphoid Rag2-/-Il2rg-/- counterparts. Conversely, transplantation of wild-type bone marrow hematopoietic progenitors into Rag2-/-Il2rg-/- mice and consequent restoration of thymocyte-mediated TEC differentiation diminishes the frequency of colony-forming units within cTECs. Our findings provide evidence that the cortical epithelium contains a reservoir of epithelial progenitors whose abundance is dynamically controlled by continual interactions with developing thymocytes across lifespan.


Assuntos
Células Epiteliais/citologia , Células-Tronco/fisiologia , Timócitos/fisiologia , Timo/citologia , Animais , Diferenciação Celular , Células Clonais , Células Epiteliais/fisiologia , Humanos , Ativação Linfocitária , Camundongos , Timócitos/imunologia , Timo/metabolismo
11.
Eur J Immunol ; 46(4): 829-33, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26947141

RESUMO

The development of effective T-cell-based immunotherapies to treat infection, cancer, and autoimmunity should incorporate the ground rules that control differentiation of T cells in the thymus. Within the thymus, thymic epithelial cells (TECs) provide microenvironments supportive of the generation and selection of T cells that are responsive to pathogen-derived antigens, and yet tolerant to self-determinants. Defects in TEC differentiation cause syndromes that range from immunodeficiency to autoimmunity, which makes the study of TECs of fundamental and clinical importance to comprehend how immunity and tolerance are balanced. Critical to tolerance induction are medullary thymic epithelial cells (mTECs), which purge autoreactive T cells, or redirect them to a regulatory T-cell lineage. In this issue of the European Journal of Immunology, studies by Baik et al. and Mayer et al. [Eur. J. Immunol. 2016. 46: XXXX-XXXX and 46: XXXX-XXXX]) document novel spatial-temporal singularities in the lineage specification and maintenance of mTECs. While Baik et al. define a developmental checkpoint during mTEC specification in the embryo, Mayer et al. reveal that the generation and maintenance of the adult mTEC compartment is temporally controlled in vivo. The two reports described new developmentally related, but temporally distinct principles that underlie the homeostasis of the thymic medulla across life.


Assuntos
Diferenciação Celular/imunologia , Células Epiteliais/imunologia , Autoimunidade , Humanos , Linfócitos T Reguladores , Timo
13.
J Virol ; 89(1): 568-80, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25339770

RESUMO

UNLABELLED: Human cytomegalovirus (hCMV) infection is characterized by a vast expansion of resting effector-type virus-specific T cells in the circulation. In mice, interleukin-7 receptor α (IL-7Rα)-expressing cells contain the precursors for long-lived antigen-experienced CD8(+) T cells, but it is unclear if similar mechanisms operate to maintain these pools in humans. Here, we studied whether IL-7Rα-expressing cells obtained from peripheral blood (PB) or lymph nodes (LNs) sustain the circulating effector-type hCMV-specific pool. Using flow cytometry and functional assays, we found that the IL-7Rα(+) hCMV-specific T cell population comprises cells that have a memory phenotype and lack effector features. We used next-generation sequencing of the T cell receptor to compare the clonal repertoires of IL-7Rα(+) and IL-7Rα(-) subsets. We observed limited overlap of clones between these subsets during acute infection and after 1 year. When we compared the hCMV-specific repertoire between PB and paired LNs, we found many identical clones but also clones that were exclusively found in either compartment. New clones that were found in PB during antigenic recall were only rarely identical to the unique LN clones. Thus, although PB IL-7Rα-expressing and LN hCMV-specific CD8(+) T cells show typical traits of memory-type cells, these populations do not seem to contain the precursors for the novel hCMV-specific CD8(+) T cell pool during latency or upon antigen recall. IL-7Rα(+) PB and LN hCMV-specific memory cells form separate virus-specific compartments, and precursors for these novel PB hCMV-specific CD8(+) effector-type T cells are possibly located in other secondary lymphoid tissues or are being recruited from the naive CD8(+) T cell pool. IMPORTANCE: Insight into the self-renewal properties of long-lived memory CD8(+) T cells and their location is crucial for the development of both passive and active vaccination strategies. Human CMV infection is characterized by a vast expansion of resting effector-type cells. It is, however, not known how this population is maintained. We here investigated two possible compartments for effector-type cell precursors: circulating acute-phase IL-7Rα-expressing hCMV-specific CD8(+) T cells and lymph node (LN)-residing hCMV-specific (central) memory cells. We show that new clones that appear after primary hCMV infection or during hCMV reactivation seldom originate from either compartment. Thus, although identical clones may be maintained by either memory population, the precursors of the novel clones are probably located in other (secondary) lymphoid tissues or are recruited from the naive CD8(+) T cell pool.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Evolução Clonal , Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Subpopulações de Linfócitos T/imunologia , Latência Viral , Adolescente , Adulto , Idoso , Animais , Linfócitos T CD8-Positivos/química , Linfócitos T CD8-Positivos/classificação , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Receptores de Interleucina-7/análise , Subpopulações de Linfócitos T/química , Subpopulações de Linfócitos T/classificação , Adulto Jovem
14.
Br J Haematol ; 171(5): 736-51, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26456771

RESUMO

Lymphotoxin-mediated activation of the lymphotoxin-ß receptor (LTßR; LTBR) has been implicated in cancer, but its role in T-cell acute lymphoblastic leukaemia (T-ALL) has remained elusive. Here we show that the genes encoding lymphotoxin (LT)-α and LTß (LTA, LTB) are expressed in T-ALL patient samples, mostly of the TAL/LMO molecular subtype, and in the TEL-JAK2 transgenic mouse model of cortical/mature T-ALL (Lta, Ltb). In these mice, expression of Lta and Ltb is elevated in early stage T-ALL. Surface LTα1 ß2 protein is expressed in primary mouse T-ALL cells, but only in the absence of microenvironmental LTßR interaction. Indeed, surface LT expression is suppressed in leukaemic cells contacting Ltbr-expressing but not Ltbr-deficient stromal cells, both in vitro and in vivo, thus indicating that dynamic surface LT expression in leukaemic cells depends on interaction with its receptor. Supporting the notion that LT signalling plays a role in T-ALL, inactivation of Ltbr results in a significant delay in TEL-JAK2-induced leukaemia onset. Moreover, young asymptomatic TEL-JAK2;Ltbr(-/-) mice present markedly less leukaemic thymocytes than age-matched TEL-JAK2;Ltbr(+/+) mice and interference with LTßR function at this early stage delayed T-ALL development. We conclude that LT expression by T-ALL cells activates LTßR signalling in thymic stromal cells, thus promoting leukaemogenesis.


Assuntos
Receptor beta de Linfotoxina/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Linhagem da Célula/genética , Expressão Gênica/genética , Humanos , Imunofenotipagem , Janus Quinase 2/genética , Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Transdução de Sinais , Microambiente Tumoral/genética
15.
Eur J Immunol ; 44(10): 2918-24, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25070355

RESUMO

Cortical and medullary thymic epithelial cells (cTECs and mTECs, respectively) provide inductive microenvironments for T-cell development and selection. The differentiation pathway of cTEC/mTEC lineages downstream of common bipotent progenitors at discrete stages of development remains unresolved. Using IL-7/CCRL1 dual reporter mice that identify specialized TEC subsets, we show that the stepwise acquisition of chemokine (C-C motif) receptor-like 1 (CCRL1) is a late determinant of cTEC differentiation. Although cTECs expressing high CCRL1 levels (CCRL1(hi) ) develop normally in immunocompetent and Rag2(-/-) thymi, their differentiation is partially blocked in Rag2(-/-) Il2rg(-/-) counterparts. These results unravel a novel checkpoint in cTEC maturation that is regulated by the cross-talk between TECs and immature thymocytes. Additionally, we identify new Ulex europaeus agglutinin 1 (UEA)(+) mTEC subtypes expressing intermediate CCRL1 levels (CCRL1(int) ) that conspicuously emerge in the postnatal thymus and differentially express Tnfrsf11a, Ccl21, and Aire. While rare in fetal and in Rag2(-/-) thymi, CCRL1(int) mTECs are restored in Rag2(-/-) Marilyn TCR-Tg mice, indicating that the appearance of postnatal-restricted mTECs is closely linked with T-cell selection. Our findings suggest that alternative temporally restricted routes of new mTEC differentiation contribute to the establishment of the medullary niche in the postnatal thymus.


Assuntos
Diferenciação Celular/imunologia , Células Epiteliais/citologia , Receptores CCR/biossíntese , Timo/citologia , Timo/crescimento & desenvolvimento , Animais , Biomarcadores/análise , Citometria de Fluxo , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Receptores CCR/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Timo/imunologia , Transcriptoma
16.
Eur J Immunol ; 44(1): 16-22, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24214487

RESUMO

Thymic epithelial cells (TECs) provide key instructive signals for T-cell differentiation. Thymic cortical (cTECs) and medullary (mTECs) epithelial cells constitute two functionally distinct microenvironments for T-cell development, which derive from a common bipotent TEC progenitor. While seminal studies have partially elucidated events downstream of bipotent TECs in relation to the emergence of mTECs and their progenitors, the control and timing of the emergence of the cTEC lineage, particularly in relation to that of mTEC progenitors, has remained elusive. In this review, we describe distinct models that explain cTEC/mTEC lineage divergence from common bipotent progenitors. In particular, we summarize recent studies in mice providing evidence that mTECs, including the auto-immune regulator(+) subset, derive from progenitors initially endowed with phenotypic properties typically associated with the cTEC lineage. These observations support a novel "serial progression" model of TEC development, in which progenitors serially acquire cTEC lineage markers, prior to their commitment to the mTEC differentiation pathway. Gaining a better understanding of the phenotypic properties of early stages in TEC progenitor development should help in determining the mechanisms regulating cTEC/mTEC lineage development, and in strategies aimed at thymus reconstitution involving TEC therapy.


Assuntos
Células Epiteliais/imunologia , Imunoterapia/métodos , Células Progenitoras Linfoides/imunologia , Modelos Imunológicos , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Timo/imunologia , Animais , Autoimunidade , Comunicação Celular/imunologia , Diferenciação Celular , Linhagem da Célula , Humanos , Imunoterapia/tendências , Camundongos
17.
J Immunol ; 191(3): 1200-9, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23794633

RESUMO

Thymic epithelial cells (TECs) help orchestrate thymopoiesis, and TEC differentiation relies on bidirectional interactions with thymocytes. Although the molecular mediators that stimulate medullary thymic epithelial cell (mTEC) maturation are partially elucidated, the signals that regulate cortical thymic epithelial cell (cTEC) homeostasis remain elusive. Using IL-7 reporter mice, we show that TECs coexpressing high levels of IL-7 (Il7(YFP+) TECs) reside within a subset of CD205(+)Ly51(+)CD40(low) cTECs that coexpresses Dll4, Ccl25, Ccrl1, Ctsl, Psmb11, and Prss16 and segregates from CD80(+)CD40(high) mTECs expressing Tnfrsf11a, Ctss, and Aire. As the frequency of Il7(YFP+) TECs gradually declines as mTEC development unfolds, we explored the relationship between Il7(YFP+) TECs and mTECs. In thymic organotypic cultures, the thymocyte-induced reduction in Il7(YFP+) TECs dissociates from the receptor activator of NF-κB-mediated differentiation of CD80(+) mTECs. Still, Il7(YFP+) TECs can generate some CD80(+) mTECs in a stepwise differentiation process via YFP(-)Ly51(low)CD80(low) intermediates. Il7(YFP+) TECs are sustained in Rag2(-/-) mice, even following in vivo anti-CD3ε treatment that mimics the process of pre-TCR ß-selection of thymocytes to the double positive (DP) stage. Using Marilyn-Rag2(-/-) TCR transgenic, we find that positive selection into the CD4 lineage moderately reduces the frequency of Il7(YFP+) TECs, whereas negative selection provokes a striking loss of Il7(YFP+) TECs. These results imply that the strength of MHC/peptide-TCR interactions between TECs and thymocytes during selection constitutes a novel rheostat that controls the maintenance of IL-7-expressing cTECs.


Assuntos
Células Epiteliais/imunologia , Interleucina-7/biossíntese , Timo/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos CD/metabolismo , Antígeno B7-1/metabolismo , Complexo CD3/imunologia , Antígenos CD40/metabolismo , Proteínas de Ligação ao Cálcio , Catepsina L/biossíntese , Catepsinas/biossíntese , Diferenciação Celular/imunologia , Movimento Celular , Células Cultivadas , Quimiocinas CC/biossíntese , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Células Epiteliais/metabolismo , Homeostase , Interleucina-7/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Lectinas Tipo C/metabolismo , Ativação Linfocitária , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Antígenos de Histocompatibilidade Menor , Técnicas de Cultura de Órgãos , Receptor Ativador de Fator Nuclear kappa-B/biossíntese , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptores CCR/biossíntese , Receptores de Superfície Celular/metabolismo , Serina Endopeptidases/biossíntese , Timócitos/metabolismo , Timo/metabolismo , Fatores de Transcrição/biossíntese , Proteína AIRE
18.
Proc Natl Acad Sci U S A ; 108(15): 6217-22, 2011 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-21444793

RESUMO

Cytokine immunotherapies targeting T lymphocytes are attractive clinical interventions against viruses and tumors. In the mouse, the homeostasis of memory α/ß CD8(+) T cells and natural killer (NK) cells is significantly improved with increased IL-15 bioavailability. In contrast, the role of "transpresented" IL-15 on human T-cell development and homeostasis in vivo is unknown. We found that both CD8 and CD4 T cells in human immune system (HIS) mice are highly sensitive to transpresented IL-15 in vivo, with both naïve (CD62L(+)CD45RA(+)) and memory phenotype (CD62L(-)CD45RO(+)) subsets being significantly increased following IL-15 "boosting." The unexpected global improvement in human T-cell homeostasis involved enhanced proliferation and survival of both naïve and memory phenotype peripheral T cells, which potentiated B-cell responses by increasing the frequency of antigen-specific responses following immunization. Transpresented IL-15 did not modify T-cell activation patterns or alter the global T-cell receptor (TCR) repertoire diversity. Our results indicate an unexpected effect of IL-15 on human T cells in vivo, in particular on CD4(+) T cells. As IL-15 promotes human peripheral T-cell homeostasis and increases the frequency of neutralizing antibody responses in HIS mice, IL-15 immunotherapy could be envisaged as a unique approach to improve vaccine responses in the clinical setting.


Assuntos
Formação de Anticorpos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Interleucina-15/imunologia , Animais , Proliferação de Células , Humanos , Camundongos , Camundongos Mutantes , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Receptores de Interleucina-15/antagonistas & inibidores
19.
Cell Death Dis ; 15(5): 352, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773063

RESUMO

Within the thymus, thymic epithelial cells (TECs) create dedicated microenvironments for T cell development and selection. Considering that TECs are sensitive to distinct pathophysiological conditions, uncovering the molecular elements that coordinate their thymopoietic role has important fundamental and clinical implications. Particularly, medullary thymic epithelial cells (mTECs) play a crucial role in central tolerance. Our previous studies, along with others, suggest that mTECs depend on molecular factors linked to genome-protecting pathways, but the precise mechanisms underlying their function remain unknown. These observations led us to examine the role of Foxo3, as it is expressed in TECs and involved in DNA damage response. Our findings show that mice with TEC-specific deletion of Foxo3 (Foxo3cKO) displayed a disrupted mTEC compartment, with a more profound impact on the numbers of CCL21+ and thymic tuft mTEClo subsets. At the molecular level, Foxo3 controls distinct functional modules in the transcriptome of cTECs and mTECs under normal conditions, which includes the regulation of ribosomal biogenesis and DNA damage response, respectively. These changes in the TEC compartment resulted in a reduced total thymocyte cellularity and specific changes in regulatory T cell and iNKT cell development in the Foxo3cKO thymus. Lastly, the thymic defects observed in adulthood correlated with mild signs of altered peripheral immunotolerance in aged Foxo3cKO mice. Moreover, the deficiency in Foxo3 moderately aggravated the autoimmune predisposition observed in Aire-deficient mice. Our findings highlight the importance of Foxo3 in preserving the homeostasis of TECs and in supporting their role in T cell development and tolerance.


Assuntos
Células Epiteliais , Proteína Forkhead Box O3 , Homeostase , Timo , Animais , Timo/metabolismo , Timo/citologia , Proteína Forkhead Box O3/metabolismo , Proteína Forkhead Box O3/genética , Células Epiteliais/metabolismo , Camundongos , Camundongos Knockout , Diferenciação Celular , Linfócitos T/metabolismo , Linfócitos T/imunologia , Camundongos Endogâmicos C57BL
20.
ACS Biomater Sci Eng ; 9(5): 2514-2523, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-37074315

RESUMO

The thymus is responsible for the selection and development of T cells, having an essential role in the establishment of adaptive immunity. Thymic epithelial cells (TECs) are key players in T cell development interacting with thymocytes in the thymic 3D environment. Feeder-layer cells have been frequently used as platforms for the successful establishment of TEC cultures. Nevertheless, the role of the feeder cell-derived extracellular matrix (ECM) on TEC cultures was not previously reported. Therefore, this work aimed at assessing the effect of the ECM produced by feeder cells cultured at two different densities on the establishment of TEC culture. Due to the high surface area and porosity, electrospun fibrous meshes were used to support ECM deposition. The feeder cell-derived ECM was efficiently recovered after decellularization, maintaining the composition of major proteins. All the decellularized matrices were permeable and showed an increase in surface mechanical properties after decellularization. TEC cultures confirmed that the ECM density impacts cellular performance, with higher densities showing a decreased cellular activity. Our findings provide evidence that feeder cell-derived ECM is a suitable substrate for TEC culture and can potentially be applied in thymus bioengineering.


Assuntos
Células Epiteliais , Matriz Extracelular , Células Alimentadoras , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Linfócitos T/metabolismo , Timo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA