Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int Heart J ; 65(3): 444-451, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38749743

RESUMO

The elevated risk of cardiovascular disease (CVD) in cancer patients and survivors is likely the result of normal age-related pathologies coupled with the direct and indirect effects of cancer therapy that extend across multiple systems. The purpose of this study was to investigate the impact of cardiac rehabilitation (CR) on CVD patients with a history of cancer.In this study, patients who had participated in the outpatient CR program were enrolled and were divided into 2 groups (cancer survivor group and no-cancer group) based on their history of cancer. The cardiopulmonary exercise test (CPET) was performed at the beginning (baseline) and at the end of the CR program (follow-up). The results of CPET at baseline and those at follow-up were analyzed retrospectively.A total of 105 patients were analyzed in this study. The cancer survivor group had 25 patients, and the non-cancer group 80. At baseline, peak oxygen uptake (peak VO2) (14.7 [11.9 to 17.6] mL/kg/minute versus 11.3 [9.7 to 14.7] mL/kg/minute; P = 0.003) was significantly lower in cancer survivors. The percent changes in peak VO2 between baseline and follow-up were not significantly different between the 2 groups (7.9 % [-11.5 to 24.5] versus 9.4 % [-7.5 to 27.3] P = 0.520).The percent changes in peak VO2 of CR participants were not significantly different despite their cancer history.


Assuntos
Sobreviventes de Câncer , Reabilitação Cardíaca , Doenças Cardiovasculares , Teste de Esforço , Neoplasias , Consumo de Oxigênio , Humanos , Masculino , Feminino , Teste de Esforço/métodos , Pessoa de Meia-Idade , Doenças Cardiovasculares/fisiopatologia , Estudos Retrospectivos , Neoplasias/complicações , Neoplasias/fisiopatologia , Reabilitação Cardíaca/métodos , Idoso , Consumo de Oxigênio/fisiologia
2.
J Immunol ; 210(11): 1653-1666, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37067332

RESUMO

Regulatory T cells (Tregs) are produced in the thymus to establish self-tolerance, and agonistic stimuli by self-Ags play a pivotal role in this process. Although two types of APCs, medullary thymic epithelial cells (mTECs) and dendritic cells (DCs), are responsible for presenting self-Ags together with costimulatory/cytokine signals, the distinct role of each APC in producing Tregs remains enigmatic. We have approached this issue by depleting the mTECs and DCs using mice expressing diphtheria toxin receptors driven by Aire and CD11c promoters, respectively. Depletion of mTECs showed an effect on Treg production quantitatively and qualitatively more profound than that of DCs followed by the development of distinct organ-specific autoimmune lesions in the hosts. Because self-Ags produced by mTECs are transferable to DCs through a process known as Ag transfer, we monitored the process of Ag transfer using mice expressing GFP from TECs. Although GFP expressed from total TECs was effectively transferred to DCs, GFP expressed from cortical TECs was not, suggesting that mTECs are the predominant source of self-Ags. We also found that GFP expressed not only from mature mTECs but also from immature mTECs was transferred to DCs, suggesting that a broad spectrum of molecules were subjected to Ag transfer during mTEC development. Interestingly, the numbers of recirculating non-Tregs producing IL-2, an important source for Treg expansion in the thymus, were reduced only in the mTEC-depleted mice. These results suggested the cooperative but distinct role of mTECs and DCs in the production of Tregs to avoid autoimmunity.


Assuntos
Linfócitos T Reguladores , Timo , Camundongos , Animais , Camundongos Endogâmicos C57BL , Células Epiteliais , Células Dendríticas , Diferenciação Celular
3.
Cancer Med ; 12(8): 9843-9848, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36912123

RESUMO

Despite the clear distinction between cortical (cTECs) and medullary thymic epithelial cells (mTECs) in physiology, the cell of origin of thymic carcinomas (TCs) and other thymic epithelial tumors remained enigmatic. We addressed this issue by focusing on AIRE, an mTEC-specific transcriptional regulator that is required for immunological self-tolerance. We found that a large proportion of TCs expressed AIRE with typical nuclear dot morphology by immunohistochemistry. AIRE expression in TCs was supported by the RNA-seq data in the TCGA-THYM database. Furthermore, our bioinformatics approach to the recent single-cell RNA-seq data on human thymi has revealed that TCs hold molecular characteristics of multiple mTEC subpopulations. In contrast, TCs lacked the gene signatures for cTECs. We propose that TCs are tumors derived from mTECs.


Assuntos
Timoma , Neoplasias do Timo , Humanos , Diferenciação Celular/genética , Células Epiteliais/metabolismo , Timoma/patologia , Timo , Neoplasias do Timo/genética , Neoplasias do Timo/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína AIRE
4.
Immunohorizons ; 6(3): 202-210, 2022 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-35264373

RESUMO

Ly6C comprises two homologous components of Ly6C1 and Ly6C2, and the expression of either of the Ly6C molecules defines unique functional subsets of monocytes. Ly6C is also expressed by other immune cell types, including Aire-expressing medullary thymic epithelial cells. Because the role of Ly6C expression in determining the functional subsets remains unclear, we generated mice deficient for both Ly6C1 and Ly6C2 with CRISPR-Cas9-mediated deletion. Mice deficient for Ly6C1/Ly6C2 showed no major alterations in the subsets and function of monocyte and other immune cells, including the cells involved in the dextran sulfate sodium salt-induced colitis model. By generating the mice deficient for Ly6C1 alone, we have also investigated the expression pattern of Ly6C1 and Ly6C2 in immune cells. Except for medullary thymic epithelial cells and CD4 single-positive T cells, immune cells predominantly expressed Ly6C2. Thus, despite the importance as a marker with a unique differential expression pattern, the Ly6C molecules have no major impact on determining the functional subsets and maintaining immune homeostasis.


Assuntos
Colite , Monócitos , Animais , Linfócitos T CD4-Positivos , Colite/induzido quimicamente , Colite/metabolismo , Células Epiteliais , Homeostase , Camundongos
5.
Cell Rep ; 38(7): 110384, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35172142

RESUMO

Impaired production of thymic regulatory T cells (Tregs) is implicated in the development of Aire-dependent autoimmunity. Because Tregs require agonistic T cell receptor stimuli by self-antigens to develop, reduced expression of self-antigens from medullary thymic epithelial cells (mTECs) has been considered to play a major role in the reduced Treg production in Aire deficiency. Here, we show that mTECs abnormally express co-inhibitory receptor CTLA-4 if Aire is non-functional. Upon binding with CD80/CD86 ligands expressed on thymic dendritic cells (DCs), the ectopically expressed CTLA-4 from Aire-deficient mTECs removes the CD80/CD86 ligands from the DCs. This attenuates the ability of DCs to provide co-stimulatory signals and to present self-antigens transferred from mTECs, both of which are required for Treg production. Accordingly, impaired production of Tregs and organ-specific autoimmunity in Aire-deficient mice are rescued by the depletion of CTLA-4 expression from mTECs. Our studies illuminate the significance of mTEC-DC interaction coordinated by Aire for the establishment of thymic tolerance.


Assuntos
Autoimunidade , Antígeno CTLA-4/metabolismo , Timo/citologia , Animais , Antígenos CD/metabolismo , Autoantígenos/metabolismo , Células Epiteliais/metabolismo , Ligantes , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Células Estromais/metabolismo , Linfócitos T Reguladores/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo , Proteína AIRE
6.
J Immunol ; 208(2): 303-320, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34930780

RESUMO

The deficiency of Aire, a transcriptional regulator whose defect results in the development of autoimmunity, is associated with reduced expression of tissue-restricted self-Ags (TRAs) in medullary thymic epithelial cells (mTECs). Although the mechanisms underlying Aire-dependent expression of TRAs need to be explored, the physical identification of the target(s) of Aire has been hampered by the low and promiscuous expression of TRAs. We have tackled this issue by engineering mice with augmented Aire expression. Integration of the transcriptomic data from Aire-augmented and Aire-deficient mTECs revealed that a large proportion of so-called Aire-dependent genes, including those of TRAs, may not be direct transcriptional targets downstream of Aire. Rather, Aire induces TRA expression indirectly through controlling the heterogeneity of mTECs, as revealed by single-cell analyses. In contrast, Ccl25 emerged as a canonical target of Aire, and we verified this both in vitro and in vivo. Our approach has illuminated the Aire's primary targets while distinguishing them from the secondary targets.


Assuntos
Autoantígenos/imunologia , Autoimunidade/imunologia , Quimiocinas CC/metabolismo , Timo/imunologia , Fatores de Transcrição/metabolismo , Animais , Autoimunidade/genética , Quimiocinas CC/genética , Células Epiteliais/imunologia , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Timo/citologia , Fatores de Transcrição/genética , Transcrição Gênica/genética , Proteína AIRE
7.
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
8.
J Immunol ; 201(11): 3244-3257, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30389776

RESUMO

Medullary thymic epithelial cells (mTECs), which express a wide range of tissue-restricted Ags (TRAs), contribute to the establishment of self-tolerance by eliminating autoreactive T cells and/or inducing regulatory T cells. Aire controls a diverse set of TRAs within Aire-expressing cells by employing various transcriptional pathways. As Aire has a profound effect on transcriptomes of mTECs, including TRAs not only at the single-cell but also the population level, we suspected that Aire (Aire+ mTECs) might control the cellular composition of the thymic microenvironment. In this study, we confirmed that this is indeed the case by identifying a novel mTEC subset expressing Ly-6 family protein whose production was defective in Aire-deficient thymi. Reaggregated thymic organ culture experiments demonstrated that Aire did not induce the expression of Ly-6C/Ly-6G molecules from mTECs as Aire-dependent TRAs in a cell-intrinsic manner. Instead, Aire+ mTECs functioned in trans to maintain Ly-6C/Ly-6G+ mTECs. Thus, Aire not only controls TRA expression transcriptionally within the cell but also controls the overall composition of mTECs in a cell-extrinsic manner, thereby regulating the transcriptome from mTECs on a global scale.


Assuntos
Células Epiteliais/patologia , Timo/fisiologia , Fatores de Transcrição/metabolismo , Animais , Antígenos Ly/metabolismo , Células Cultivadas , Microambiente Celular , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Cultura de Órgãos , Fatores de Transcrição/genética , Ativação Transcricional , Proteína AIRE
9.
J Immunol ; 195(11): 5149-58, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26503950

RESUMO

Aire in medullary thymic epithelial cells (mTECs) plays an important role in the establishment of self-tolerance. Because Aire(+) mTECs appear to be a limited subset, they may constitute a unique lineage(s) among mTECs. An alternative possibility is that all mTECs are committed to express Aire in principle, but Aire expression by individual mTECs is conditional. To investigate this issue, we established a novel Aire reporter strain in which endogenous Aire is replaced by the human AIRE-GFP-Flag tag (Aire/hAGF-knockin) fusion gene. The hAGF reporter protein was produced and retained very efficiently within mTECs as authentic Aire nuclear dot protein. Remarkably, snapshot analysis revealed that mTECs expressing hAGF accounted for >95% of mature mTECs, suggesting that Aire expression does not represent a particular mTEC lineage(s). We confirmed this by generating Aire/diphtheria toxin receptor-knockin mice in which long-term ablation of Aire(+) mTECs by diphtheria toxin treatment resulted in the loss of most mature mTECs beyond the proportion of those apparently expressing Aire. These results suggest that Aire expression is inherent to all mTECs but may occur at particular stage(s) and/or cellular states during their differentiation, thus accounting for the broad impact of Aire on the promiscuous gene expression of mTECs.


Assuntos
Células Epiteliais/metabolismo , Timo/metabolismo , Fatores de Transcrição/biossíntese , Animais , Diferenciação Celular , Toxina Diftérica/farmacologia , Células Epiteliais/citologia , Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Introdução de Genes , Proteínas de Fluorescência Verde/genética , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Timo/citologia , Fatores de Transcrição/genética , Proteína AIRE
10.
J Immunol ; 195(10): 4641-9, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26453754

RESUMO

Cortical thymic epithelial cells (cTECs) and medullary thymic epithelial cells (mTECs) play essential roles in the positive and negative selection of developing thymocytes, respectively. Aire in mTECs plays an essential role in the latter process through expression of broad arrays of tissue-restricted Ags. To determine whether the location of Aire within the medulla is absolutely essential or whether Aire could also function within the cortex for establishment of self-tolerance, we used bacterial artificial chromosome technology to establish a semiknockin strain of NOD-background (ß5t/Aire-transgenic) mice expressing Aire under control of the promoter of ß5t, a thymoproteasome expressed exclusively in the cortex. Although Aire was expressed in cTECs as typical nuclear dot protein in ß5t/Aire-Tg mice, cTECs expressing Aire ectopically did not confer transcriptional expression of either Aire-dependent or Aire-independent tissue-restricted Ag genes. We then crossed ß5t/Aire-Tg mice with Aire-deficient NOD mice, generating a strain in which Aire expression was confined to cTECs. Despite the presence of Aire(+) cTECs, these mice succumbed to autoimmunity, as did Aire-deficient NOD mice. The thymic microenvironment harboring Aire(+) cTECs, within which many Aire-activated genes were present, also showed no obvious alteration of positive selection, suggesting that Aire's unique property of generating a self-tolerant T cell repertoire is functional only in mTECs.


Assuntos
Autoimunidade/genética , Tolerância a Antígenos Próprios/genética , Timócitos/imunologia , Timo/imunologia , Fatores de Transcrição/genética , Animais , Autoimunidade/imunologia , Diferenciação Celular/imunologia , Cromossomos Artificiais Bacterianos/genética , Células Epiteliais/citologia , Células Epiteliais/imunologia , Técnicas de Introdução de Genes , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Tolerância a Antígenos Próprios/imunologia , Linfócitos T/imunologia , Timócitos/citologia , Timo/citologia , Fatores de Transcrição/metabolismo , Proteína AIRE
11.
Arthritis Rheumatol ; 67(9): 2512-22, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25989265

RESUMO

OBJECTIVE: Syndecan 4 has been implicated as a critical mediator of inflammatory responses because of its functions as a coreceptor and reservoir for growth factors and chemokines. Although syndecan 4 is known to be expressed on B cells, its role in immune responses remains unclear. The purpose of this study was to investigate the contribution of syndecan 4 to the development of immune arthritis in murine models. METHODS: The clinical severity of 3 immunopathologically distinct models, collagen-induced arthritis (CIA), antigen-induced arthritis (AIA), and collagen antibody-induced arthritis (CAIA), was evaluated in wild-type (WT) mice and in syndecan 4-deficient (Sdc4(-/-) ) mice. Germinal center (GC) formation, B cell profiles, humoral immune responses, and B cell migration were analyzed during the course of CIA. RESULTS: Sdc4(-/-) mice were resistant to the induction of CIA, which is T cell and B cell dependent, but AIA and CAIA, which are T cell dependent and T cell independent, respectively, occurred with equal frequency in WT mice and Sdc4(-/-) mice. Furthermore, Sdc4(-/-) mice had reduced numbers of B cells and deficient GC formation in draining lymph nodes (DLNs) during the course of CIA, resulting in reduced production of collagen-specific autoantibodies. Compared with B cells from WT mice, B cells from Sdc4(-/-) mice showed reduced chemotactic migration toward stromal cell-derived factor 1 (SDF-1) and reduced SDF-1-mediated Akt phosphorylation. Consistent with these findings, in vivo transfer experiments showed that fewer Sdc4(-/-) B cells than WT B cells were found in and around the follicles in the DLNs. CONCLUSION: Our findings suggest that syndecan 4 contributes to the development of CIA by promoting GC formation and autoantibody production through its regulation of SDF-1-mediated B cell migration.


Assuntos
Artrite Experimental/genética , Linfócitos B/imunologia , Quimiocina CXCL12/imunologia , Centro Germinativo/imunologia , Imunidade Humoral/genética , Sindecana-4/genética , Adjuvantes Imunológicos/toxicidade , Animais , Artrite Experimental/imunologia , Movimento Celular/genética , Movimento Celular/imunologia , Colágeno Tipo II/toxicidade , Imunidade Humoral/imunologia , Camundongos , Camundongos Knockout , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sindecana-4/imunologia , Linfócitos T/imunologia
12.
J Immunol ; 193(9): 4356-67, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25261487

RESUMO

Essential roles of NF-κB-inducing kinase (NIK) for the development of medullary thymic epithelial cells (mTECs) and regulatory T cells have been highlighted by studies using a strain of mouse bearing a natural mutation of the NIK gene (aly mice). However, the exact mechanisms underlying the defect in thymic cross-talk leading to the breakdown of self-tolerance in aly mice remain elusive. In this study, we demonstrated that production of regulatory T cells and the final maturation process of positively selected conventional αß T cells are impaired in aly mice, partly because of a lack of mature mTECs. Of note, numbers of thymic dendritic cells and their expression of costimulatory molecules were also affected in aly mice in a thymic stroma-dependent manner. The results suggest a pivotal role of NIK in the thymic stroma in establishing self-tolerance by orchestrating cross-talk between mTECs and dendritic cells as well as thymocytes. In addition, we showed that negative selection was impaired in aly mice as a result of the stromal defect, which accounts for the development of organ-specific autoimmunity through a lack of normal NIK.


Assuntos
Comunicação Celular/imunologia , Células Dendríticas/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Tolerância a Antígenos Próprios/imunologia , Timócitos/imunologia , Animais , Antígeno B7-1/metabolismo , Diferenciação Celular , Células Dendríticas/metabolismo , Células Epiteliais/metabolismo , Expressão Gênica , Imunofenotipagem , Masculino , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Mutação , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Células Estromais/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Timócitos/metabolismo , Timo/imunologia , Timo/metabolismo , Quinase Induzida por NF-kappaB
13.
J Mol Med (Berl) ; 92(12): 1271-81, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25099519

RESUMO

UNLABELLED: Tumor-derived matricellular proteins such as osteopontin (OPN) and tenascin-C (TN-C) have been implicated in tumor growth and metastasis. However, the molecular basis of how these proteins contribute to tumor progression remains to be elucidated. Importantly, these matricellular proteins are known to interact with α9ß1 integrin. Therefore, we hypothesized that tumor-derived α9ß1 integrin may contribute to tumor progression. To clarify the roles of α9ß1 integrin in tumor growth and lymphatic metastasis, we used an inhibitory anti-human α9ß1 integrin antibody (anti-hα9ß1 antibody) and a α9ß1 integrin-positive human breast cancer cell line, MDA-MB-231 luc-D3H2LN (D3H2LN), in vitro functional assays, and an in vivo orthotopic xenotransplantation model. In this study, we demonstrated that tumor, but not host α9ß1 integrin, contributes to tumor growth, lymphatic metastasis, recruitment of cancer-associated fibroblasts (CAFs), and host-derived OPN production. We also found that CAFs contributed to tumor growth, lymphatic metastasis, and host-derived OPN levels. Consistent with those findings, tumor volume was well-correlated with numbers of CAFs and levels of host-derived OPN. Furthermore, it was shown that the inoculation of D3H2LN cells into mammary fat pads with mouse embryonic fibroblasts (MEFs), obtained from wild type, but not OPN knock-out mice, resulted in enhancement of tumor growth, thus indicating that CAF-derived OPN enhanced tumor growth. These results suggested that tumor α9ß1-mediated signaling plays a pivotal role in generating unique primary tumor tissue microenvironments, which favor lymphatic metastasis and tumor growth. KEY MESSAGES: Tumor α9ß1 integrin promotes lymphatic metastasis through enhancing invasion. Tumor α9ß1 integrin promotes tumor growth through CAFs. Tumor α9ß1 integrin enhances the recruitment of CAFs into the primary tumor. Tumor cells induce the production of OPN by CAFs in the primary tumor. CAF-derived OPN promotes tumor growth.


Assuntos
Neoplasias da Mama/patologia , Fibroblastos/patologia , Integrinas/metabolismo , Metástase Linfática/patologia , Transdução de Sinais , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Fibroblastos/metabolismo , Humanos , Integrinas/análise , Metástase Linfática/genética , Camundongos , Camundongos Knockout , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Osteopontina/análise , Osteopontina/genética , Osteopontina/metabolismo
14.
J Immunol ; 192(6): 2585-92, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24516201

RESUMO

Understanding the cellular dynamics of Aire-expressing lineage(s) among medullary thymic epithelial cells (AEL-mTECs) is essential for gaining insight into the roles of Aire in establishment of self-tolerance. In this study, we monitored the maturation program of AEL-mTECs by temporal lineage tracing, in which bacterial artificial chromosome transgenic mice expressing tamoxifen-inducible Cre recombinase under control of the Aire regulatory element were crossed with reporter strains. We estimated that the half-life of AEL-mTECs subsequent to Aire expression was ∼7-8 d, which was much longer than that reported previously, owing to the existence of a post-Aire stage. We found that loss of Aire did not alter the overall lifespan of AEL-mTECs, inconsistent with the previous notion that Aire expression in medullary thymic epithelial cells (mTECs) might result in their apoptosis for efficient cross-presentation of self-antigens expressed by AEL-mTECs. In contrast, Aire was required for the full maturation program of AEL-mTECs, as exemplified by the lack of physiological downregulation of CD80 during the post-Aire stage in Aire-deficient mice, thus accounting for the abnormally increased CD80(high) mTECs seen in such mice. Of interest, increased CD80(high) mTECs in Aire-deficient mice were not mTEC autonomous and were dependent on cross-talk with thymocytes. These results further support the roles of Aire in the differentiation program of AEL-mTECs.


Assuntos
Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Células Epiteliais/imunologia , Fatores de Transcrição/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Autoantígenos/imunologia , Autoantígenos/metabolismo , Antígeno B7-1/imunologia , Antígeno B7-1/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Cultivadas , Apresentação Cruzada/genética , Apresentação Cruzada/imunologia , Células Epiteliais/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Cinética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Timócitos/citologia , Timócitos/imunologia , Timócitos/metabolismo , Timo/citologia , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína AIRE
15.
J Cell Biochem ; 115(3): 585-95, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24129963

RESUMO

The osteoclast (OC) is a major player in the pathogenic bone destruction of inflammatory bone diseases such as rheumatoid arthritis and Langerhans cell histiocytosis. Recently, it was shown that immature dendritic cells (iDC) fuse faster and more efficiently than monocytes in forming OC-like multinucleated giant cells (MGCs), and that osteopontin (OPN) is involved in the pathogenesis of inflammatory bone diseases. In this study, we hypothesized that OPN is a key factor for generation of OC-like MGCs from iDCs. We used an in vitro culture system to differentiate iDCs, derived from monocytes obtained from the blood of healthy donors, into OC-like MGCs. We evaluated OPN levels and expression of OPN receptors during the course of differentiation. OPN has an arginine-glycine-aspartic acid (RGD) motif, and protease cleavage reveals a SVVYGLR motif. The concentrations of both full-length and cleaved forms of OPN increased during the course of OC-like MGC formation. Expression of OPN RGD- and SVVYGLR-recognizing receptors also increased at later stages. We analyzed whether blocking OPN binding to its receptors affected OC-like MGC formation. Monocytes treated with OPN siRNA were able to differentiate into iDCs effectively; however, differentiation of these iDCs into OC-like MGCs was significantly reduced. The formation of OC-like MGCs was not significantly reduced by RGD synthetic peptide. By contrast, SVVYGLR synthetic peptide caused a significant reduction. These data suggest that the cleaved form of OPN plays a critical role in driving iDC differentiation into OC-like MGCs in the early phase of differentiation, in an autocrine and/or paracrine fashion.


Assuntos
Artrite Reumatoide/genética , Histiocitose de Células de Langerhans/genética , Osteoclastos/metabolismo , Osteopontina/metabolismo , Artrite Reumatoide/patologia , Diferenciação Celular/genética , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Células Gigantes/efeitos dos fármacos , Histiocitose de Células de Langerhans/metabolismo , Humanos , Oligopeptídeos , Osteoclastos/citologia , Osteopontina/genética , RNA Interferente Pequeno
16.
Cytokine ; 60(1): 129-37, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22818182

RESUMO

The overexpression of osteopontin is associated with various inflammatory liver diseases. Interestingly, each of these diseases is also associated with IL-17 expression. Therefore, we sought to determine whether there is any mechanistic link between osteopontin and IL-17. Herein we show that IL-17 and osteopontin levels were significantly increased in patients with chronic hepatitis B. We found that IL-17 and osteopontin levels increased similarly in mice with concanavalin A-induced hepatitis. Both osteopontin- and IL-17-deficient mice were resistant to concanavalin A-induced hepatic injury. In addition, osteopontin markedly induced IL-17 expression by leukocytes (from humans and mice). This effect could be blocked by a specific antibody against osteopontin. ß3 integrin (one of the osteopontin receptors) was critically involved in the induction of IL-17 production by osteopontin. Osteopontin-induced IL-17 expression was mediated through the p38, JNK, and NF-κB pathways. These findings suggest that osteopontin regulates IL-17 production during the pathogenesis of hepatitis and provide new evidence for the critical roles of osteopontin and IL-17 in hepatitis.


Assuntos
Hepatite B Crônica/sangue , Interleucina-17/sangue , Leucócitos Mononucleares/metabolismo , Osteopontina/sangue , Adulto , Animais , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/genética , Concanavalina A/toxicidade , Resistência a Medicamentos/genética , Inibidores Enzimáticos/farmacologia , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Interleucina-17/genética , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leucócitos Mononucleares/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Osteopontina/genética , Peptídeos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Adulto Jovem , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Blood ; 119(23): 5429-37, 2012 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-22553313

RESUMO

The BM microenvironment is required for the maintenance, proliferation, and mobilization of hematopoietic stem and progenitor cells (HSPCs), both during steady-state conditions and hematopoietic recovery after myeloablation. The ECM meshwork has long been recognized as a major anatomical component of the BM microenvironment; however, the molecular signatures and functions of the ECM to support HSPCs are poorly understood. Of the many ECM proteins, the expression of tenascin-C (TN-C) was found to be dramatically up-regulated during hematopoietic recovery after myeloablation. The TN-C gene was predominantly expressed in stromal cells and endothelial cells, known as BM niche cells, supporting the function of HSPCs. Mice lacking TN-C (TN-C(-/-)) mice showed normal steady-state hematopoiesis; however, they failed to reconstitute hematopoiesis after BM ablation and showed high lethality. The capacity to support transplanted wild-type hematopoietic cells to regenerate hematopoiesis was reduced in TN-C(-/-) recipient mice. In vitro culture on a TN-C substratum promoted the proliferation of HSPCs in an integrin α9-dependent manner and up-regulated the expression of the cyclins (cyclinD1 and cyclinE1) and down-regulated the expression of the cyclin-dependent kinase inhibitors (p57(Kip2), p21(Cip1), p16(Ink4a)). These results identify TN-C as a critical component of the BM microenvironment that is required for hematopoietic regeneration.


Assuntos
Células da Medula Óssea/citologia , Medula Óssea/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , Tenascina/metabolismo , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/efeitos da radiação , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Proliferação de Células , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fluoruracila/administração & dosagem , Fluoruracila/efeitos adversos , Deleção de Genes , Células-Tronco Hematopoéticas/metabolismo , Cadeias alfa de Integrinas/genética , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Células Estromais/citologia , Células Estromais/metabolismo , Tenascina/análise , Tenascina/genética , Regulação para Cima , Irradiação Corporal Total
18.
J Immunol ; 187(11): 5851-64, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22039306

RESUMO

The interaction between matricellular proteins such as tenascin-C (TN-C) and osteopontin (OPN) and integrins has been implicated in the pathology of rheumatoid arthritis in which Th17 cells are recognized as primary pathogenic cells. The differentiation of Th17 cells is tightly regulated by cytokines derived from APCs, receiving various signals including TLR stimuli. In this study, we used a collagen-induced arthritis model and found that increased numbers of α(9) integrin-positive conventional dendritic cells and macrophage were detectable in the draining lymph node (dLN) shortly following first immunization, and these cells produced both TN-C and OPN, ligands for α(9) integrin. α(9) integrin-mediated signaling, induced by TN-C and OPN, promoted the production of Th17-related cytokines by conventional dendritic cells and macrophages in synergy with TLR2 and 4 signaling. This led to the Th17 cell differentiation and arthritis development. Moreover, Th17 cells generated under blocking of α(9) integrin-mediated signaling showed low level of CCR6 expression and impaired migration ability toward CCL20. Thus, we have identified α(9) integrin-mediated signaling by TN-C and OPN as a novel intrinsic regulator of pathogenic Th17 cell generation that contributes to the development of rheumatoid arthritis.


Assuntos
Artrite Experimental/imunologia , Glicoproteínas/imunologia , Integrinas/imunologia , Transdução de Sinais/imunologia , Tenascina/imunologia , Células Th17/citologia , Animais , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Western Blotting , Diferenciação Celular/imunologia , Separação Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Citometria de Fluxo , Imunofluorescência , Glicoproteínas/biossíntese , Humanos , Integrinas/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Reação em Cadeia da Polimerase em Tempo Real , Tenascina/biossíntese , Células Th17/imunologia
19.
J Immunol ; 187(11): 5671-83, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22021613

RESUMO

The adaptive immune system generates memory cells, which induce a rapid and robust immune response following secondary Ag encounter. Memory CD8(+) T cells are a critical component of protective immunity against infections and cancers. Therefore, understanding the mechanism whereby memory CD8(+) T cells are generated and maintained is important for inducing effective memory CD8(+) T cell response. Recent studies have demonstrated that the inflammatory cytokine IL-12 favors the generation of terminal effector CD8(+) T cells rather than memory precursor effector CD8(+) T cells by regulating the expression of the transcription factor T-bet. In this study, we report that the inflammatory cytokine osteopontin (Opn) modulates memory CD8(+) T cell generation during influenza virus infection. Although Opn wild-type and Opn knockout (KO) mice had similar numbers of virus-specific effector CD8(+) T cells, virus-specific effector CD8(+) T cells generated in Opn KO mice showed low levels of T-bet expression and an increased memory precursor cell population compared with cells generated in Opn wild-type mice. This resulted in the persistently increased number of memory CD8(+) T cells in Opn KO mice. Studies with bone marrow-derived dendritic cells demonstrated that Opn deficiency in bone marrow-derived dendritic cells results in low levels of IL-12 production in response to the stimulation with influenza virus. Thus, we hypothesize that Opn modulates the generation of memory precursor effector CD8(+) T cells by regulating cytokine milieu during the acute phase of virus infection. This finding may provide new insight into the role of Opn in adaptive immune response.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Infecções por Orthomyxoviridae/imunologia , Osteopontina/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Separação Celular , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteopontina/metabolismo , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/virologia , Reação em Cadeia da Polimerase em Tempo Real
20.
Arterioscler Thromb Vasc Biol ; 31(5): 1066-74, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21330609

RESUMO

OBJECTIVE: Syndecan-4 (Syn4) is a heparan sulfate proteoglycan and works as a coreceptor for various growth factors. We examined whether Syn4 could be involved in the development of neointimal formation in vivo. METHODS AND RESULTS: Wild-type (WT) and Syn4-deficient (Syn4-/-) mice were subjected to wire-induced femoral artery injury. Syn4 mRNA was upregulated after vascular injury in WT mice. Neointimal formation was attenuated in Syn4-/- mice, concomitantly with the reduction of Ki67-positive vascular smooth muscle cells (VSMCs). Basic-fibroblast growth factor- or platelet-derived growth factor-BB-induced proliferation, extracellular signal-regulated kinase activation, and expression of cyclin D1 and Bcl-2 were impaired in VSMCs from Syn4-/- mice. To examine the role of Syn4 in bone marrow (BM)-derived vascular progenitor cells (VPCs) and vascular walls, we generated chimeric mice by replacing the BM cells of WT and Syn4-/- mice with those of WT or Syn4-/- mice. Syn4 expressed by both vascular walls and VPCs contributed to the neointimal formation after vascular injury. Although the numbers of VPCs were compatible between WT and Syn4-/- mice, mobilization of VPCs from BM after vascular injury was defective in Syn4-/- mice. CONCLUSIONS: Syn4 deficiency limits neointimal formation after vascular injury by regulating VSMC proliferation and VPC mobilization. Therefore, Syn4 may be a novel therapeutic target for preventing arterial restenosis after angioplasty.


Assuntos
Movimento Celular , Proliferação de Células , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Células-Tronco/metabolismo , Sindecana-4/deficiência , Túnica Íntima/metabolismo , Lesões do Sistema Vascular/metabolismo , Animais , Apoptose , Becaplermina , Transplante de Medula Óssea , Ciclina D1/metabolismo , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Artéria Femoral/lesões , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Hiperplasia , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/lesões , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2 , Proteínas Proto-Oncogênicas c-sis , Transdução de Sinais , Células-Tronco/patologia , Sindecana-4/genética , Fatores de Tempo , Túnica Íntima/lesões , Túnica Íntima/patologia , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA