Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nat Immunol ; 23(9): 1330-1341, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35999392

RESUMEN

Fibroblasts, the most abundant structural cells, exert homeostatic functions but also drive disease pathogenesis. Single-cell technologies have illuminated the shared characteristics of pathogenic fibroblasts in multiple diseases including autoimmune arthritis, cancer and inflammatory colitis. However, the molecular mechanisms underlying the disease-associated fibroblast phenotypes remain largely unclear. Here, we identify ETS1 as the key transcription factor governing the pathological tissue-remodeling programs in fibroblasts. In arthritis, ETS1 drives polarization toward tissue-destructive fibroblasts by orchestrating hitherto undescribed regulatory elements of the osteoclast differentiation factor receptor activator of nuclear factor-κB ligand (RANKL) as well as matrix metalloproteinases. Fibroblast-specific ETS1 deletion resulted in ameliorated bone and cartilage damage under arthritic conditions without affecting the inflammation level. Cross-tissue fibroblast single-cell data analyses and genetic loss-of-function experiments lent support to the notion that ETS1 defines the perturbation-specific fibroblasts shared among various disease settings. These findings provide a mechanistic basis for pathogenic fibroblast polarization and have important therapeutic implications.


Asunto(s)
Artritis Reumatoide , Fibroblastos , Proteína Proto-Oncogénica c-ets-1 , Artritis Reumatoide/metabolismo , Artritis Reumatoide/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Metaloproteinasas de la Matriz/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Proteína Proto-Oncogénica c-ets-1/metabolismo , Ligando RANK/genética , Factores de Transcripción/metabolismo
2.
Nat Immunol ; 21(10): 1172-1180, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32839611

RESUMEN

Fibroblasts are one of the most common but also neglected types of stromal cells, the heterogeneity of which underlies the specific function of tissue microenvironments in development and regeneration. In the thymus, autoreactive T cells are thought to be negatively selected by reference to the self-antigens expressed in medullary epithelial cells, but the contribution of other stromal cells to tolerance induction has been poorly examined. In the present study, we report a PDGFR+ gp38+ DPP4- thymic fibroblast subset that is required for T cell tolerance induction. The deletion of the lymphotoxin ß-receptor in thymic fibroblasts caused an autoimmune phenotype with decreased expression of tissue-restricted and fibroblast-specific antigens, offering insight into the long-sought target of lymphotoxin signaling in the context of the regulation of autoimmunity. Thus, thymic medullary fibroblasts play an essential role in the establishment of central tolerance by producing a diverse array of self-antigens.


Asunto(s)
Fibroblastos/inmunología , Linfocitos T/inmunología , Timo/metabolismo , Animales , Autoantígenos/inmunología , Autoinmunidad , Células Cultivadas , Microambiente Celular , Selección Clonal Mediada por Antígenos , Dipeptidil Peptidasa 4/metabolismo , Tolerancia Inmunológica , Receptor beta de Linfotoxina/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Timo/citología
3.
Nat Immunol ; 19(11): 1265-1276, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30323341

RESUMEN

The methylation of arginine residues in proteins is a post-translational modification that contributes to a wide range of biological processes. Many cytokines involved in T cell development and activation utilize the common cytokine receptor γ-chain (γc) and the kinase JAK3 for signal transduction, but the regulatory mechanism that underlies the expression of these factors remains unclear. Here we found that the arginine methyltransferase PRMT5 was essential for the maintenance of invariant natural killer T cells (iNKT cells), CD4+ T cells and CD8+ T cells. T cell-specific deletion of Prmt5 led to a marked reduction in signaling via γc-family cytokines and a substantial loss of thymic iNKT cells, as well as a decreased number of peripheral CD4+ T cells and CD8+ T cells. PRMT5 induced the symmetric dimethylation of Sm proteins that promoted the splicing of pre-mRNA encoding γc and JAK3, and this critically contributed to the expression of γc and JAK3. Thus, arginine methylation regulates strength of signaling via γc-family cytokines by facilitating the expression of signal-transducing components.


Asunto(s)
Arginina/metabolismo , Subunidad gamma Común de Receptores de Interleucina/inmunología , Proteína-Arginina N-Metiltransferasas/metabolismo , Transducción de Señal/inmunología , Linfocitos T/inmunología , Animales , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Metilación , Ratones , Proteína-Arginina N-Metiltransferasas/inmunología , Linfocitos T/metabolismo
4.
Int Immunol ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38916145

RESUMEN

The thymus is an organ required for T cell development and is also an eosinophil-rich organ; however, the nature and function of thymic eosinophils remain unclear. Here, we characterized the gene expression and differentiation mechanism of thymic eosinophils in mice. Thymic eosinophils showed a distinct gene expression profile compared with other organ-resident eosinophils. The number of thymic eosinophils was controlled by medullary thymic epithelial cells. In Rag-deficient mice, the unique gene expression signature of thymic eosinophils was lost but restored by pre-T cell receptor signaling, which induces CD4+ CD8+ thymocyte differentiation, indicating that T cell differentiation beyond the CD4- CD8- stage is necessary and sufficient for the induction of thymic eosinophils. These results demonstrate that thymic eosinophils are quantitatively and qualitatively regulated by medullary thymic epithelial cells and developing thymocytes, respectively, suggesting that thymic eosinophils are a distinct, thymus-specific cell subset, induced by interactions with thymic cells.

5.
Immunol Rev ; 302(1): 68-85, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34096078

RESUMEN

Fibroblasts have recently attracted attention as a key stromal component that controls the immune responses in lymphoid tissues. The thymus has a unique microenvironment comprised of a variety of stromal cells, including fibroblasts and thymic epithelial cells (TECs), the latter of which is known to be important for T cell development because of their ability to express self-antigens. Thymic fibroblasts contribute to thymus organogenesis during embryogenesis and form the capsule and medullary reticular network in the adult thymus. However, the immunological significance of thymic fibroblasts has thus far only been poorly elucidated. In this review, we will summarize the current views on the development and functions of thymic fibroblasts as revealed by new technologies such as multicolor flow cytometry and single cell-based transcriptome profiling. Furthermore, the recently discovered role of medullary fibroblasts in the establishment of T cell tolerance by producing a unique set of self-antigens will be highlighted.


Asunto(s)
Fibroblastos , Linfocitos T , Diferenciación Celular , Células Epiteliales , Activación de Linfocitos , Células del Estroma , Timo
6.
Int Immunol ; 34(1): 45-52, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34687536

RESUMEN

Medullary thymic epithelial cells (mTECs) help shape the thymic microenvironment for T-cell development by expressing a variety of peripheral tissue-restricted antigens (TRAs). The self-tolerance of T cells is established by negative selection of autoreactive T cells that bind to TRAs. To increase the diversity of TRAs, a fraction of mTECs terminally differentiates into distinct subsets resembling atypical types of epithelial cells in specific peripheral tissues. As such, thymic tuft cells that express peripheral tuft cell genes have recently emerged. Here, we show that the transcription factor SRY-box transcription factor 4 (Sox4) is highly expressed in mTECs and is essential for the development of thymic tuft cells. Mice lacking Sox4 specifically in TECs had a significantly reduced number of thymic tuft cells with no effect on the differentiation of other mTEC subsets, including autoimmune regulator (Aire)+ and Ccl21a+ mTECs. Furthermore, Sox4 expression was diminished in mice deficient in TEC-specific lymphotoxin ß receptor (LTßR), indicating a role for the LTßR-Sox4 axis in the differentiation of thymic tuft cells. Given that Sox4 promotes differentiation of peripheral tuft cells, our findings suggest that mTECs employ the same transcriptional program as peripheral epithelial cells. This mechanism may explain how mTECs diversify peripheral antigen expression to project an immunological self within the thymic medulla.


Asunto(s)
Receptor beta de Linfotoxina/genética , Factores de Transcripción SOXC/genética , Timo/inmunología , Animales , Diferenciación Celular/inmunología , Receptor beta de Linfotoxina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Transcripción SOXC/inmunología , Transducción de Señal/genética , Timo/citología
7.
Int J Mol Sci ; 22(9)2021 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-33926096

RESUMEN

The SOS response is induced upon DNA damage and the inhibition of Z ring formation by the product of the sulA gene, which is one of the LexA-regulated genes, allows time for repair of damaged DNA. On the other hand, severely DNA-damaged cells are eliminated from cell populations. Overexpression of sulA leads to cell lysis, suggesting SulA eliminates cells with unrepaired damaged DNA. Transcriptome analysis revealed that overexpression of sulA leads to up-regulation of numerous genes, including soxS. Deletion of soxS markedly reduced the extent of cell lysis by sulA overexpression and soxS overexpression alone led to cell lysis. Further experiments on the SoxS regulon suggested that LpxC is a main player downstream from SoxS. These findings suggested the SulA-dependent cell lysis (SDCL) cascade as follows: SulA→SoxS→LpxC. Other tests showed that the SDCL cascade pathway does not overlap with the apoptosis-like and mazEF cell death pathways.


Asunto(s)
Daño del ADN/fisiología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Amidohidrolasas/metabolismo , Apoptosis/genética , Proteínas Bacterianas/metabolismo , División Celular/genética , Daño del ADN/genética , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/genética , Genes Bacterianos/genética , Serina Endopeptidasas/metabolismo , Transactivadores/metabolismo
8.
Biochem Biophys Res Commun ; 496(1): 25-30, 2018 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-29291408

RESUMEN

Fine regulation of the Ras/mitogen-activating protein kinase (MAPK) pathway is crucial in controlling the survival, proliferation, and development of various types of cells. Ras-activating protein-like 3 (Rasal3) is a T cell-specific Ras GTPase-activating protein that negatively regulates T cell receptor (TCR)-induced activation of Ras/MAPK pathway. Rasal3-deficient mice showed a decreased number of naive T cells because Rasal3 is required for the survival of naive T cells. In the current study, we observed ameliorated Type1 T helper (Th1) cell- and Type2 T helper (Th2) cell-dependent contact hypersensitivity reactions in Rasal3-deficient mice, along with a marked shortage of T cells at regional lymph node. Activated Rasal3-deficient T cells showed an increased cell death with reduced Bcl2 expression, suggesting that Rasal3 is required for the survival of not only naïve T cells but also activated T cells. Collectively, Rasal3 controls the magnitude of inflammatory responses through the survival of both naive T cells and activated T cells in vivo.


Asunto(s)
Supervivencia Celular/inmunología , Dermatitis por Contacto/inmunología , Dermatitis por Contacto/patología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Proteínas Activadoras de ras GTPasa/inmunología , Animales , Ratones , Ratones Endogámicos C57BL
9.
J Immunol ; 197(6): 2269-79, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27511731

RESUMEN

ESET/SETDB1, one of the major histone methyltransferases, catalyzes histone 3 lysine 9 (H3K9) trimethylation. ESET is critical for suppressing expression of retroviral elements in embryonic stem cells; however, its role in the immune system is not known. We found that thymocyte-specific deletion of ESET caused impaired T cell development, with CD8 lineage cells being most severely affected. Increased apoptosis of CD8 single-positive cells was observed, and TCR-induced ERK activation was severely inhibited in ESET(-/-) thymocytes. Genome-wide comprehensive analysis of mRNA expression and H3K9 trimethylation revealed that ESET regulates expression of numerous genes in thymocytes. Among them, FcγRIIB, whose signaling can inhibit ERK activation, was strongly and ectopically expressed in ESET(-/-) thymocytes. Indeed, genetic depletion of FcγRIIB in ESET(-/-) thymocytes rescued impaired ERK activation and partially restored defective positive selection in ESET(-/-) mice. Therefore, impaired T cell development in ESET(-/-) mice is partly due to the aberrant expression of FcγRIIB. Collectively, to our knowledge, we identify ESET as the first trimethylated H3K9 histone methyltransferase playing a crucial role in T cell development.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/metabolismo , Animales , Apoptosis , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Genoma , N-Metiltransferasa de Histona-Lisina/deficiencia , Histonas/metabolismo , Lisina/metabolismo , Metilación , Ratones , Regiones Promotoras Genéticas , Receptores de IgG/genética , Receptores de IgG/metabolismo , Timocitos/inmunología , Timocitos/fisiología
10.
EMBO Rep ; 16(5): 638-53, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25770130

RESUMEN

The thymus provides a specialized microenvironment in which distinct subsets of thymic epithelial cells (TECs) support T-cell development. Here, we describe the significance of cortical TECs (cTECs) in T-cell development, using a newly established mouse model of cTEC deficiency. The deficiency of mature cTECs caused a massive loss of thymic cellularity and impaired the development of αßT cells and invariant natural killer T cells. Unexpectedly, the differentiation of certain γδT-cell subpopulations-interleukin-17-producing Vγ4 and Vγ6 cells-was strongly dysregulated, resulting in the perturbation of γδT-mediated inflammatory responses in peripheral tissues. These findings show that cTECs contribute to the shaping of the TCR repertoire, not only of "conventional" αßT cells but also of inflammatory "innate" γδT cells.


Asunto(s)
Epitelio/metabolismo , Interleucina-17/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/metabolismo , Timo/metabolismo , Animales , Diferenciación Celular , Supervivencia Celular/genética , Análisis Mutacional de ADN , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/inmunología , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Timocitos/citología , Timocitos/inmunología , Timocitos/metabolismo , Timo/inmunología , Timo/patología
11.
Int J Oral Sci ; 16(1): 18, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38413562

RESUMEN

The immune-stromal cell interactions play a key role in health and diseases. In periodontitis, the most prevalent infectious disease in humans, immune cells accumulate in the oral mucosa and promote bone destruction by inducing receptor activator of nuclear factor-κB ligand (RANKL) expression in osteogenic cells such as osteoblasts and periodontal ligament cells. However, the detailed mechanism underlying immune-bone cell interactions in periodontitis is not fully understood. Here, we performed single-cell RNA-sequencing analysis on mouse periodontal lesions and showed that neutrophil-osteogenic cell crosstalk is involved in periodontitis-induced bone loss. The periodontal lesions displayed marked infiltration of neutrophils, and in silico analyses suggested that the neutrophils interacted with osteogenic cells through cytokine production. Among the cytokines expressed in the periodontal neutrophils, oncostatin M (OSM) potently induced RANKL expression in the primary osteoblasts, and deletion of the OSM receptor in osteogenic cells significantly ameliorated periodontitis-induced bone loss. Epigenomic data analyses identified the OSM-regulated RANKL enhancer region in osteogenic cells, and mice lacking this enhancer showed decreased periodontal bone loss while maintaining physiological bone metabolism. These findings shed light on the role of neutrophils in bone regulation during bacterial infection, highlighting the novel mechanism underlying osteoimmune crosstalk.


Asunto(s)
Pérdida de Hueso Alveolar , Periodontitis , Humanos , Ratones , Animales , Neutrófilos/metabolismo , Neutrófilos/patología , Citocinas , Pérdida de Hueso Alveolar/microbiología , Osteogénesis , Ligando RANK
12.
Bone Res ; 11(1): 43, 2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37563119

RESUMEN

The bony skeleton is continuously renewed throughout adult life by the bone remodeling process, in which old or damaged bone is removed by osteoclasts via largely unknown mechanisms. Osteocytes regulate bone remodeling by producing the osteoclast differentiation factor RANKL (encoded by the TNFSF11 gene). However, the precise mechanisms underlying RANKL expression in osteocytes are still elusive. Here, we explored the epigenomic landscape of osteocytic cells and identified a hitherto-undescribed osteocytic cell-specific intronic enhancer in the TNFSF11 gene locus. Bioinformatics analyses showed that transcription factors involved in cell death and senescence act on this intronic enhancer region. Single-cell transcriptomic data analysis demonstrated that cell death signaling increased RANKL expression in osteocytic cells. Genetic deletion of the intronic enhancer led to a high-bone-mass phenotype with decreased levels of RANKL in osteocytic cells and osteoclastogenesis in the adult stage, while RANKL expression was not affected in osteoblasts or lymphocytes. These data suggest that osteocytes may utilize a specialized regulatory element to facilitate osteoclast formation at the bone surface to be resorbed by linking signals from cellular senescence/death and RANKL expression.

13.
Front Immunol ; 13: 1045881, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36713401

RESUMEN

The γδT cells that produce IL-17 (γδT17 cells) play a key role in various pathophysiologic processes in host defense and homeostasis. The development of γδT cells in the thymus requires γδT cell receptor (γδTCR) signaling mediated by the spleen tyrosine kinase (Syk) family proteins, Syk and Zap70. Here, we show a critical role of Syk in the early phase of γδT cell development using mice deficient for Syk specifically in lymphoid lineage cells (Syk-conditional knockout (cKO) mice). The development of γδT cells in the Syk-cKO mice was arrested at the precursor stage where the expression of Rag genes and αßT-lineage-associated genes were retained, indicating that Syk is required for γδT-cell lineage commitment. Loss of Syk in γδT cells weakened TCR signal-induced phosphorylation of Erk and Akt, which is mandatory for the thymic development of γδT17 cells. Syk-cKO mice exhibited a loss of γδT17 cells in the thymus as well as throughout the body, and thereby are protected from γδT17-dependent psoriasis-like skin inflammation. Collectively, our results indicate that Syk is a key player in the lineage commitment of γδT cells and the priming of γδT17 cell differentiation.


Asunto(s)
Transducción de Señal , Timo , Animales , Ratones , Quinasa Syk/genética , Diferenciación Celular/genética , Linaje de la Célula
14.
Nat Commun ; 13(1): 4166, 2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35851381

RESUMEN

The ontogeny and fate of stem cells have been extensively investigated by lineage-tracing approaches. At distinct anatomical sites, bone tissue harbors multiple types of skeletal stem cells, which may independently supply osteogenic cells in a site-specific manner. Periosteal stem cells (PSCs) and growth plate resting zone stem cells (RZSCs) critically contribute to intramembranous and endochondral bone formation, respectively. However, it remains unclear whether there is functional crosstalk between these two types of skeletal stem cells. Here we show PSCs are not only required for intramembranous bone formation, but also for the growth plate maintenance and prolonged longitudinal bone growth. Mice deficient in PSCs display progressive defects in intramembranous and endochondral bone formation, the latter of which is caused by a deficiency in PSC-derived Indian hedgehog (Ihh). PSC-specific deletion of Ihh impairs the maintenance of the RZSCs, leading to a severe defect in endochondral bone formation in postnatal life. Thus, crosstalk between periosteal and growth plate stem cells is essential for post-developmental skeletal growth.


Asunto(s)
Condrocitos , Placa de Crecimiento , Animales , Proteínas Hedgehog/genética , Ratones , Osteogénesis/genética , Células Madre
15.
Methods Mol Biol ; 2111: 193-203, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31933209

RESUMEN

The thymus is an organ where T cells develop throughout life. Using mice as a model animal, molecular mechanisms of intrathymic T cell development have been studied. Fetal thymus organ culture technique enables ex vivo reconstitution of fetal-specific T cell development, while bone marrow chimera technique allows in vivo reconstitution of T cell development in adult thymus. These techniques can be combined with retroviral gene transduction into the T cell progenitors to evaluate the function of genes of interest in developing T cells. Here, we describe the basic protocols for retrovirus gene transduction into fetal or adult T cell progenitors and reconstitution of thymic T cell development including experimental tips such as using cryopreserved fetal liver or bone marrow cells as sources of T cell progenitors.


Asunto(s)
Células de la Médula Ósea/citología , Técnicas de Cultivo de Órganos/métodos , Linfocitos T/citología , Timo/embriología , Animales , Células de la Médula Ósea/inmunología , Diferenciación Celular , Criopreservación , Ratones , Retroviridae/genética , Timo/citología , Timo/inmunología , Transducción Genética
16.
Cell Rep ; 32(10): 108124, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32905763

RESUMEN

Osteoprotegerin (OPG) is a circulating decoy receptor for RANKL, a multifunctional cytokine essential for the differentiation of tissue-specific cells in bone and immune systems such as osteoclasts, medullary thymic epithelial cells (mTECs), and intestinal microfold cells (M cells). However, it is unknown whether OPG functions only at the production site or circulates to other tissues acting in an endocrine fashion. Here we explore the cellular source of OPG by generating OPG-floxed mice and show that locally produced OPG, rather than circulating OPG, is crucial for bone and immune homeostasis. Deletion of OPG in osteoblastic cells leads to severe osteopenia without affecting serum OPG. Deletion of locally produced OPG increases mTEC and M cell numbers while retaining the normal serum OPG level. This study shows that OPG limits its functions within the tissue where it was produced, illuminating the importance of local regulation of the RANKL system.


Asunto(s)
Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoprotegerina/metabolismo , Animales , Ratones
17.
Nat Metab ; 2(12): 1382-1390, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33288951

RESUMEN

Osteoclasts are the exclusive bone-resorbing cells, playing a central role in bone metabolism, as well as the bone damage that occurs under pathological conditions1,2. In postnatal life, haematopoietic stem-cell-derived precursors give rise to osteoclasts in response to stimulation with macrophage colony-stimulating factor and receptor activator of nuclear factor-κB ligand, both of which are produced by osteoclastogenesis-supporting cells such as osteoblasts and osteocytes1-3. However, the precise mechanisms underlying cell fate specification during osteoclast differentiation remain unclear. Here, we report the transcriptional profiling of 7,228 murine cells undergoing in vitro osteoclastogenesis, describing the stepwise events that take place during the osteoclast fate decision process. Based on our single-cell transcriptomic dataset, we find that osteoclast precursor cells transiently express CD11c, and deletion of receptor activator of nuclear factor-κB specifically in CD11c-expressing cells inhibited osteoclast formation in vivo and in vitro. Furthermore, we identify Cbp/p300-interacting transactivator with Glu/Asp-rich carboxy-terminal domain 2 (Cited2) as the molecular switch triggering terminal differentiation of osteoclasts, and deletion of Cited2 in osteoclast precursors in vivo resulted in a failure to commit to osteoclast fate. Together, the results of this study provide a detailed molecular road map of the osteoclast differentiation process, refining and expanding our understanding of the molecular mechanisms underlying osteoclastogenesis.


Asunto(s)
Osteoclastos/fisiología , Osteogénesis/fisiología , Transducción de Señal/fisiología , Animales , Células de la Médula Ósea , Antígeno CD11c/metabolismo , Proliferación Celular , Bases de Datos Factuales , Femenino , Ratones , Ratones Endogámicos C57BL , Osteogénesis/genética , Embarazo , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Transactivadores/metabolismo , Factores de Transcripción p300-CBP
18.
Inflamm Regen ; 39: 6, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30976362

RESUMEN

T cells are central to the vertebrate immune system. Two distinct types of T cells, αßT and γδT cells, express different types of T cell antigen receptors (TCRs), αßTCR and γδTCR, respectively, that are composed of different sets of somatically rearranged TCR chains and CD3 subunits. γδT cells have recently attracted considerable attention due to their ability to produce abundant cytokines and versatile roles in host defense, tissue regeneration, inflammation, and autoimmune diseases. Both αßT and γδT cells develop in the thymus. Unlike the development of αßT cells, which depends on αßTCR-mediated positive and negative selection, the development of γδT cells, including the requirement of γδTCR, has been less well understood. αßT cells differentiate into effector cells in the peripheral tissues, whereas γδT cells acquire effector functions during their development in the thymus. In this review, we will discuss the current state of knowledge of the molecular mechanism of TCR signal transduction and its role in the thymic development of γδT cells, particularly highlighting a newly discovered mechanism that controls proinflammatory γδT cell development.

19.
Nat Metab ; 1(9): 868-875, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-32694743

RESUMEN

Receptor activator of NF-κB ligand (RANKL) is a multifunctional cytokine known to affect immune and skeletal systems, as well as oncogenesis and metastasis1-4. RANKL is synthesized as a membrane-bound molecule, and cleaved into its soluble form by proteases5-7. As the soluble form of RANKL does not contribute greatly to bone remodelling or ovariectomy-induced bone loss8, whether soluble RANKL has a role in pathological settings remains unclear. Here we show that soluble RANKL promotes the formation of tumour metastases in bone. Mice that selectively lack soluble RANKL (Tnfsf11ΔS/ΔS)5-7,9 have normal bone homoeostasis and develop a normal immune system but display markedly reduced numbers of bone metastases after intracardiac injection of RANK-expressing melanoma and breast cancer cells. Deletion of soluble RANKL does not affect osteoclast numbers in metastatic lesions or tumour metastasis to non-skeletal tissues. Therefore, soluble RANKL is dispensable for physiological regulation of bone and immune systems, but has a distinct and pivotal role in the promotion of bone metastases.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias/patología , Receptor Activador del Factor Nuclear kappa-B/fisiología , Animales , Remodelación Ósea/fisiología , Diferenciación Celular/fisiología , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Osteoclastos/citología , Receptor Activador del Factor Nuclear kappa-B/genética
20.
J Clin Invest ; 128(1): 415-426, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29202478

RESUMEN

γδT cells produce inflammatory cytokines and have been implicated in the pathogenesis of cancer, infectious diseases, and autoimmunity. The T cell receptor (TCR) signal transduction that specifically regulates the development of IL-17-producing γδT (γδT17) cells largely remains unclear. Here, we showed that the receptor proximal tyrosine kinase Syk is essential for γδTCR signal transduction and development of γδT17 in the mouse thymus. Zap70, another tyrosine kinase essential for the development of αßT cells, failed to functionally substitute for Syk in the development of γδT17. Syk induced the activation of the PI3K/Akt pathway upon γδTCR stimulation. Mice deficient in PI3K signaling exhibited a complete loss of γδT17, without impaired development of IFN-γ-producing γδT cells. Moreover, γδT17-dependent skin inflammation was ameliorated in mice deficient in RhoH, an adaptor known to recruit Syk. Thus, we deciphered lineage-specific TCR signaling and identified the Syk/PI3K pathway as a critical determinant of proinflammatory γδT cell differentiation.


Asunto(s)
Diferenciación Celular/inmunología , Fosfatidilinositol 3-Quinasas/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Transducción de Señal/inmunología , Quinasa Syk/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular/genética , Inflamación/genética , Inflamación/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Ratones , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Transducción de Señal/genética , Quinasa Syk/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA