Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Immunol ; 197(5): 1968-78, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27481847

ABSTRACT

Activation of TGF-ß by dendritic cells (DCs) expressing αvß8 integrin is essential for the generation of intestinal regulatory T cells (Tregs) that in turn promote tolerance to intestinal Ags. We have recently shown that αvß8 integrin is preferentially expressed by CD103(+) DCs and confers their ability to activate TGF-ß and generate Tregs. However, how these DCs become specialized for this vital function is unknown. In this study, we show that ß8 expression is controlled by a combination of factors that include DC lineage and signals derived from the tissue microenvironment and microbiota. Specifically, our data demonstrate that TGF-ß itself, along with retinoic acid and TLR signaling, drives expression of αvß8 in DCs. However, these signals only result in high levels of ß8 expression in cells of the cDC1 lineage, CD8α(+), or CD103(+)CD11b(-) DCs, and this is associated with epigenetic changes in the Itgb8 locus. Together, these data provide a key illustrative example of how microenvironmental factors and cell lineage drive the generation of regulatory αvß8-expressing DCs specialized for activation of TGF-ß to facilitate Treg generation.


Subject(s)
Cell Lineage , Cellular Microenvironment , Dendritic Cells/immunology , Integrin beta Chains/metabolism , Intestines/cytology , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Cell Differentiation , Dendritic Cells/physiology , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Integrin beta Chains/genetics , Integrin beta Chains/immunology , Intestines/immunology , Mice , T-Lymphocytes, Regulatory/physiology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Tretinoin/metabolism
2.
Biomed Res Int ; 2015: 137893, 2015.
Article in English | MEDLINE | ID: mdl-26583087

ABSTRACT

Maintaining the identity of Foxp3(+) regulatory T cells (Tregs) is critical for controlling immune responses in the gut, where an imbalance between Tregs and T effector cells has been linked to inflammatory bowel disease. Accumulating evidence suggests that Tregs can convert into Th17 cells and acquire an inflammatory phenotype. In this study, we used an adoptive transfer model of Ag-specific T cells to study the contribution of different factors to the reprogramming of in vitro-generated Treg cells (iTreg) into IL-17-producing cells in a mouse model of gut inflammation in vivo. Our results show that intestinal inflammation induces the reprogramming of iTreg cells into IL-17-producing cells and that vitamin A restrains reprogramming in the gut. We also demonstrate that the presence of IL-2 during the in vitro generation of iTreg cells confers resistance to Th17 conversion but that IL-2 and retinoic acid (RA) cooperate to maintain Foxp3 expression following stimulation under Th17-polarizing conditions. Additionally, although IL-2 and RA differentially regulate the expression of different Treg cell suppressive markers, Treg cells generated under different polarizing conditions present similar suppressive capacity.


Subject(s)
Inflammation/genetics , Interleukin-17/biosynthesis , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Vitamin A/administration & dosage , Animals , Cell Lineage/drug effects , Cell Lineage/immunology , Cellular Reprogramming/genetics , Cellular Reprogramming/immunology , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Humans , Immunity, Cellular/genetics , Inflammation/immunology , Interleukin-17/immunology , Interleukin-2/immunology , Intestinal Mucosa/metabolism , Intestines/pathology , Mice , T-Lymphocytes, Regulatory/drug effects , Th17 Cells/pathology , Tretinoin/administration & dosage
3.
Gastroenterology ; 148(5): 991-1001.e4, 2015 May.
Article in English | MEDLINE | ID: mdl-25678452

ABSTRACT

BACKGROUND & AIMS: Intraepithelial T lymphocyte cells (IEL) are the first immune cells to respond to pathogens; they help maintain the integrity of the epithelial barrier. We studied the function of the mouse glycoprotein Signaling Lymphocyte Activation Molecule Family receptor (SLAMF) 4 (encoded by Slamf4) on the surface of CD8αß αß T-cell receptor (TCR)(+) IELs, and the roles of these cells in homeostasis of the small intestine in mice. METHODS: SLAMF4(-) CD8(+) αßTCR(+) cells isolated from spleens of OT-I Rag1(-/-) mice were induced to express gut-homing receptors and transferred to C57BL/6J mice; levels of SLAMF4(+) cells were measured in small intestine tissues. After administration of anti-CD3 or antigen, with or without anti-SLAM4, to C57BL/6J and Slamf4(-/-) mice, CD8αß αßTCR(+) IELs were collected; cytokine production and cytotoxicity were measured. Depletion of CX3CR1(+) phagocytes was assessed in mice by live-cell confocal imaging or by cytofluorometry; small intestine tissues were analyzed by histology and inflammation was quantified. RESULTS: Splenic CD8(+) αßTCR(+) cells began to express SLAMF4 only after migrating to the small intestine. Injection of C57BL/6J mice with anti-SLAMF4 and anti-CD3 increased levels of interleukin 10 and interferon gamma secretion by IEL, compared with injection of anti-CD3 only. Similarly, the number of granzyme B(+) cytotoxic CD8(+) αßTCR(+) IELs increased in Slamf4(-/-) mice after injection of anti-CD3 and anti-SLAMF4, administration of antigen, or injection of anti-CD3. Surprisingly, in vivo activation of CD8αß(+) IELs with anti-CD3 or antigen caused transient depletion of CX3CR1(+) phagocytes, which was prolonged by co-injection with anti-SLAMF4 or in Slamf4(-/-) mice. Anti-CD3 aggravated inflammation in the small intestines of Slamf4(-/-) mice and Eat2a(-/-)Eat2b(-/-) mice, indicated by flattened villi and crypt hyperplasia. CONCLUSIONS: In mice, the intestinal environment induces SLAMF4 expression and localization to the surface of CD8(+) αßTCR(+) IELs. Signaling via SLAMF4 controls expansion of cytotoxic CD8αß(+) IELs, which regulate the reversible depletion of lamina propria phagocytes and inflammation in the small intestine.


Subject(s)
Antigens, CD/metabolism , Cell Proliferation , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Lymphocyte Activation , Receptors, Immunologic/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, CD/genetics , CX3C Chemokine Receptor 1 , Cell Movement , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Homeostasis , Hyperplasia , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/pathology , Mice, Inbred C57BL , Mice, Knockout , Phagocytes/immunology , Phagocytes/metabolism , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Signal Transduction , Signaling Lymphocytic Activation Molecule Family , Spleen/immunology , Spleen/metabolism , T-Lymphocytes, Cytotoxic/immunology
4.
Front Immunol ; 5: 186, 2014.
Article in English | MEDLINE | ID: mdl-24795728

ABSTRACT

Signaling lymphocytic activation molecule (SLAM)-associated protein (SAP) plays an essential role in the immune system mediating the function of several members of the SLAM family (SLAMF) of receptors, whose expression is essential for T, NK, and B-cell responses. Additionally, the expression of SAP in double-positive thymocytes is mandatory for natural killer T (NKT) cells and, in mouse, for innate CD8(+) T cell development. To date, only two members of the SLAMF of receptors, Slamf1 and Slamf6, have been shown to positively cooperate during NKT cell differentiation in mouse. However, it is less clear whether other members of this family may also participate in the development of these innate T cells. Here, we show that Slamf[1 + 6](-/-) and Slamf[1 + 5 + 6](-/-) B6 mice have ~70% reduction of NKT cells compared to wild-type B6 mice. Unexpectedly, the proportion of innate CD8(+) T cells slightly increased in the Slamf[1 + 5 + 6](-/-) , but not in the Slamf[1 + 6](-/-) strain, suggesting that Slamf5 may function as a negative regulator of innate CD8(+) T cell development. Accordingly, Slamf5(-/-) B6 mice showed an exclusive expansion of innate CD8(+) T cells, but not NKT cells. Interestingly, the SAP-independent Slamf7(-/-) strain showed an expansion of both splenic innate CD8(+) T cells and thymic NKT cells. On the other hand, and similar to what was recently shown in Slamf3(-/-) BALB/c mice, the proportions of thymic promyelocytic leukemia zinc finger (PLZF(hi)) NKT cells and innate CD8(+) T cells significantly increased in the SAP-independent Slamf8(-/-) BALB/c strain. In summary, these results show that NKT and innate CD8(+) T cell development can be regulated in a SAP-dependent and -independent fashion by SLAMF receptors, in which Slamf1, Slamf6, and Slamf8 affect development of NKT cells, and that Slamf5, Slamf7, and Slamf8 affect the development of innate CD8(+) T cells.

5.
Gut ; 63(9): 1431-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24030488

ABSTRACT

BACKGROUND AND OBJECTIVE: While pro-inflammatory monocyte trafficking to the intestine has been partially characterised, the molecules required for migration of tolerogenic mononuclear phagocytes (dendritic cells (DC) and macrophages) are unknown. We hypothesised that the gut-homing receptor integrin α4ß7 is required for this process. METHODS: We used a T cell-mediated colitis model to study the role of α4ß7 in the innate immune compartment. We then performed competitive bone marrow (BM) reconstitution experiments to assess the requirement of α4ß7 in the generation of intestinal retinoic acid (RA)-producing CD11c(hi) DC (ALDE(+)DC) and CD64 macrophages. Using mixed BM chimeras we also asked whether α4ß7 is required to give rise to tolerogenic mononuclear phagocytes. RESULTS: Lack of ß7 integrins in the innate immune compartment (ß7(-/-)RAG2(-/-) mice) markedly accelerated T cell-mediated colitis, which was correlated with lower numbers and frequencies of ALDE(+)DC in mesenteric lymph nodes. Consistent with a role of α4ß7 in the generation of intestinal mononuclear phagocytes, BM cells from ß7(-/-) mice poorly reconstituted small intestine ALDE(+)DC and Mφ when compared to their wild type counterparts. In addition, mice lacking ß7 integrins in the CD11c(hi) compartment showed decreased ability to induce Foxp3(+) T(REG) and IL-10-producing T cells. CONCLUSIONS: Mice lacking ß7 integrins in the innate immune compartment are more susceptible to intestinal inflammation, which is correlated with a requirement of ß7 integrins to reconstitute gut mononuclear phagocytes with tolerogenic potential.


Subject(s)
Colitis/immunology , Dendritic Cells/metabolism , Integrin beta Chains/metabolism , Integrins/metabolism , Intestinal Mucosa/immunology , Macrophages/metabolism , Animals , Biomarkers/metabolism , Cell Movement , Colitis/metabolism , Dendritic Cells/physiology , Immunity, Innate , Integrins/deficiency , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Macrophages/physiology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/metabolism , Tretinoin/metabolism
6.
Eur J Immunol ; 42(12): 3136-40, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23255010

ABSTRACT

Macrophages are becoming increasingly recognized as key cellular players in intestinal immune homeostasis. However, differentiating between macrophages and dendritic cells (DCs) is often difficult, and finding a specific phenotypic signature for intestinal macrophage identification has remained elusive. In this issue of the European Journal of Immunology, Tamoutounour et al. [Eur. J. Immunol. 2012. 42: 3150-3166] identify CD64 as a specific macrophage marker that can be used to discriminate DCs from macrophages in the murine small and large intestine, under both steady-state and inflammatory conditions. The authors also propose a sequential 'monocyte-waterfall' model for intestinal macrophage differentiation, with implications for immune tolerance and inflammation at the gut mucosal interface. This Commentary will discuss the advantages and potential limitations of CD64 as a marker for intestinal macrophages.


Subject(s)
Colitis/immunology , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Lymph Nodes/immunology , Macrophages/immunology , Mesentery/immunology , Receptors, IgG/immunology , Th1 Cells/immunology , Animals , Humans
7.
Methods Mol Biol ; 757: 411-34, 2012.
Article in English | MEDLINE | ID: mdl-21909925

ABSTRACT

Effector/memory T cells can migrate to most extra-lymphoid tissues in the body. However, migration to the intestinal mucosa requires the expression of very specific homing receptors on T cells, integrin α4ß7 and chemokine receptor CCR9. These receptors are induced by all-trans retinoic acid (RA), a vitamin A metabolite that is specifically synthesized by gut-associated dendritic cells (DC), but not by extra-intestinal DC. Here we summarize some general concepts on T cell homing with an emphasis on the gut mucosa. We also discuss experimental strategies to generate gut-homing T cells in vivo and in vitro and the techniques to track gut-homing T cells.


Subject(s)
Cell Movement/immunology , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Animals , Dendritic Cells/immunology , Intestinal Mucosa/metabolism , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Receptors, Lymphocyte Homing/immunology , Receptors, Lymphocyte Homing/metabolism , Staining and Labeling , T-Lymphocytes/metabolism
8.
Mol Aspects Med ; 33(1): 63-76, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22120429

ABSTRACT

The vitamin A (VA) metabolite all-trans retinoic acid (RA) plays a key role in mucosal immune responses. RA is produced by gut-associated dendritic cells (DC) and is required for generating gut-tropic lymphocytes and IgA-antibody-secreting cells (IgA-ASC). Moreover, RA modulates Foxp3(+) regulatory T cell (T(REG)) and Th17 effector T cell differentiation. Thus, although RA could be used as an effective "mucosal adjuvant" in vaccines, it also appears to be required for establishing intestinal immune tolerance. Here we discuss the roles proposed for RA in shaping intestinal immune responses and tolerance at the gut mucosal interface. We also focus on recent data exploring the mechanisms by which gut-associated DC acquire RA-producing capacity.


Subject(s)
Dendritic Cells/immunology , Immune Tolerance , Intestines/immunology , Tretinoin/physiology , Vitamin A/metabolism , Adjuvants, Immunologic/metabolism , Adjuvants, Immunologic/pharmacology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Differentiation/immunology , Dendritic Cells/drug effects , Humans , Immune Tolerance/drug effects , Immunity, Mucosal/drug effects , Intestines/drug effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tretinoin/metabolism , Tretinoin/pharmacology , Vitamin A/pharmacology
9.
J Immunol ; 187(4): 1877-83, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21765014

ABSTRACT

Vitamin A deficiency is highly prevalent in much of the developing world, where vaccination programs are of paramount importance to public health. However, the impact of vitamin A deficiency on the immunogenicity and protective efficacy of vaccines has not been defined previously. In this article, we show that the vitamin A metabolite retinoic acid is critical for trafficking of vaccine-elicited T lymphocytes to the gastrointestinal mucosa and for vaccine protective efficacy in mice. Moderate vitamin A deficiency abrogated Ag-specific T lymphocyte trafficking to the gastrointestinal tract, gastrointestinal cellular immune responses, and protection against a mucosal challenge following immunization with a recombinant adenovirus vaccine vector. Oral vitamin A supplementation as well as retinoic acid administration fully restored the mucosal immune responses and vaccine protective efficacy. These data suggest that oral vitamin A supplementation may be important for optimizing the success of vaccines against HIV-1 and other mucosal pathogens in the developing world, highlighting a critical relationship between host nutritional status and vaccine efficacy.


Subject(s)
AIDS Vaccines/immunology , Adenoviridae , Gastric Mucosa/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Vitamin A Deficiency/immunology , AIDS Vaccines/pharmacology , Animals , Cell Movement/drug effects , Cell Movement/immunology , Immunity, Mucosal/drug effects , Immunity, Mucosal/genetics , Mice , Mice, Knockout , T-Lymphocytes/immunology , Vitamin A Deficiency/drug therapy , Vitamin A Deficiency/genetics
10.
J Immunol ; 187(1): 141-50, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21646294

ABSTRACT

Gut-associated dendritic cells (DC) synthesize all-trans retinoic acid, which is required for inducing gut-tropic lymphocytes. Gut-associated DC from MyD88(-/-) mice, which lack most TLR signals, expressed low levels of retinal dehydrogenases (critical enzymes for all-trans retinoic acid biosynthesis) and were significantly impaired in their ability to induce gut-homing T cells. Pretreatment of extraintestinal DC with a TLR1/2 agonist was sufficient to induce retinal dehydrogenases and to confer these DC with the capacity to induce gut-homing lymphocytes via a mechanism dependent on MyD88 and JNK/MAPK. Moreover, gut-associated DC from TLR2(-/-) mice, or from mice in which JNK was pharmacologically blocked, were impaired in their education to imprint gut-homing T cells, which correlated with a decreased induction of gut-tropic T cells in TLR2(-/-) mice upon immunization. Thus, MyD88-dependent TLR2 signals are necessary and sufficient to educate DC with gut-specific imprinting properties and contribute in vivo to the generation of gut-tropic T cells.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Genomic Imprinting/immunology , Intestinal Mucosa/immunology , Myeloid Differentiation Factor 88/physiology , Signal Transduction/immunology , Toll-Like Receptor 1/physiology , Toll-Like Receptor 2/physiology , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line, Tumor , Coculture Techniques , Dendritic Cells/cytology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Radiation Chimera , Receptors, Lymphocyte Homing/deficiency , Receptors, Lymphocyte Homing/genetics , Receptors, Lymphocyte Homing/physiology , Signal Transduction/genetics
11.
Gastroenterology ; 141(1): 176-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21596042

ABSTRACT

BACKGROUND & AIMS: Gut-associated dendritic cells (DC) metabolize vitamin A into all-trans retinoic acid (RA), which is required to induce lymphocytes to localize to the gastrointestinal tract and promotes the differentiation of Foxp3+ regulatory T cells and IgA antibody-secreting cells. We investigated whether RA functions in a positive-feedback loop in DC to induce its own synthesis. METHODS: We measured levels of retinoids in intestinal tissues from mice and assessed the role of RA in the functional specialization of gut-associated DC in cell cultures and mice. We used pharmacologic antagonists to determine the signaling pathways involved in regulation of DC and used MyD88-/- mice to determine the contribution of Toll-like receptor signaling in RA-mediated effects on DC. RESULTS: The concentration of retinoids decreased in a proximal-to-distal gradient along the intestine, which correlated with the activity of gut-specific DC. Importantly, RA regulated the ability of gut-associated DC to produce RA, induce T cells to localize to the gastrointestinal tract, and generate regulatory T cells and IgA-secreting cells. RA was sufficient to induce its own production by extraintestinal DC in vitro and in vivo. RA-mediated regulation of DC required signaling through the mitogen-activated protein kinase signaling pathway and unexpectedly required MyD88, which is conventionally associated with Toll-like receptor, interleukin-1, and interleukin-18 signaling. CONCLUSIONS: RA is necessary and sufficient to induce DC to regulate T-cell localization to the gastrointestinal tract and IgA secretion. Our findings also indicate crosstalk between the RA receptor and MyD88-dependent Toll-like receptor signaling pathways.


Subject(s)
Dendritic Cells/metabolism , Intestinal Mucosa/metabolism , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Tretinoin/metabolism , Analysis of Variance , Animals , Cells, Cultured , Chemotaxis, Leukocyte , Coculture Techniques , Dendritic Cells/drug effects , Dendritic Cells/immunology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , Humans , Immunoglobulin A, Secretory/metabolism , Intestines/drug effects , Intestines/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Time Factors , Toll-Like Receptors/metabolism , Tretinoin/administration & dosage
12.
J Immunol ; 183(1): 277-84, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542439

ABSTRACT

The acquired immune response begins with Ag presentation by dendritic cells (DCs) to naive T cells in a heterocellular cell-cell contact-dependent process. Although both DCs and T cells are known to express connexin43, a gap junction protein subunit, the role of connexin43 on the initiation of T cell responses remains to be elucidated. In the present work, we report the formation of gap junctions between DCs and T cells and their role on T cell activation during Ag presentation by DCs. In cocultures of DCs and T cells, Lucifer yellow microinjected into DCs is transferred to adjacent transgenic CD4(+) T cells, only if the specific antigenic peptide was present at least during the first 24 h of cocultures. This dye transfer was sensitive to gap junction blockers, such as oleamide, and small peptides containing the extracellular loop sequences of conexin. Furthermore, in this system, gap junction blockers drastically reduced T cell activation as reflected by lower proliferation, CD69 expression, and IL-2 secretion. This lower T cell activation produced by gap junction blockers was not due to a lower expression of CD80, CD86, CD40, and MHC-II on DCs. Furthermore, gap junction blocker did not affect polyclonal activation of T cell induced with anti-CD3 plus anti-CD28 Abs in the absence of DCs. These results strongly suggest that functional gap junctions assemble at the interface between DCs and T cells during Ag presentation and that they play an essential role in T cell activation.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epitopes, T-Lymphocyte/physiology , Gap Junctions/immunology , Lymphocyte Activation/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Amino Acid Sequence , Animals , Biomarkers/metabolism , CD28 Antigens/physiology , CD3 Complex/physiology , Cell Communication/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Coculture Techniques , Gap Junctions/genetics , Gap Junctions/metabolism , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Spleen/cytology , Spleen/immunology , Spleen/metabolism
13.
Dev Biol ; 323(1): 114-29, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18775422

ABSTRACT

The neural crest is induced at the border of the neural plate in a multistep process by signals emanated from the epidermis, neural plate and mesoderm. In this work we show for the first time the existence of a neural crest maintenance step which is dependent on signals released from the mesoderm. We identified Endothelin-1 (Edn1) and its receptor (Ednra) as key players of this signal and we show that Edn1/Ednra signaling is required for maintenance of the neural crest by a dual mechanism of cell specification and cell survival. We show that: (i) Ednra is expressed in prospective neural crest; (ii) loss-of-function experiments with antisense morpholino or with specific chemical inhibitor suppress the expression of early neural crest markers; (iii) gain-of-function experiments expand the neural crest territory; (iv) epistatic experiments show that Ednra/Edn1 is downstream of the early neural crest gene Msx1 and upstream of the late genes Sox9 and Sox10; and (v) Edn1/Ednra signaling inhibits apoptosis and controls cell specification of the neural crest. Together, our results provide insight on a new role of Edn1/Ednra cell signaling pathway during early neural crest development.


Subject(s)
Embryonic Induction/genetics , Endothelin-1/metabolism , Neural Crest/physiology , Receptor, Endothelin A/metabolism , Signal Transduction/physiology , Animals , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/physiology , Endothelin-1/genetics , Immunohistochemistry , In Situ Hybridization , Models, Biological , Neural Crest/metabolism , Receptor, Endothelin A/genetics , Signal Transduction/genetics , Xenopus/embryology , Xenopus/genetics , Xenopus/metabolism
14.
Development ; 132(11): 2587-97, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15857909

ABSTRACT

Migration of neural crest cells is an elaborate process that requires the delamination of cells from an epithelium and cell movement into an extracellular matrix. In this work, it is shown for the first time that the non-canonical Wnt signalling [planar cell polarity (PCP) or Wnt-Ca2+] pathway controls migration of neural crest cells. By using specific Dsh mutants, we show that the canonical Wnt signalling pathway is needed for neural crest induction, while the non-canonical Wnt pathway is required for neural crest migration. Grafts of neural crest tissue expressing non-canonical Dsh mutants, as well as neural crest cultured in vitro, indicate that the PCP pathway works in a cell-autonomous manner to control neural crest migration. Expression analysis of non-canonical Wnt ligands and their putative receptors show that Wnt11 is expressed in tissue adjacent to neural crest cells expressing the Wnt receptor Frizzled7 (Fz7). Furthermore, loss- and gain-of-function experiments reveal that Wnt11 plays an essential role in neural crest migration. Inhibition of neural crest migration by blocking Wnt11 activity can be rescued by intracellular activation of the non-canonical Wnt pathway. When Wnt11 is expressed opposite its normal site of expression, neural crest migration is blocked. Finally, time-lapse analysis of cell movement and cell protrusion in neural crest cultured in vitro shows that the PCP or Wnt-Ca2+ pathway directs the formation of lamellipodia and filopodia in the neural crest cells that are required for their delamination and/or migration.


Subject(s)
Body Patterning/physiology , Cell Movement/physiology , Glycoproteins/metabolism , Neural Crest/physiology , Signal Transduction/physiology , Xenopus/embryology , Adaptor Proteins, Signal Transducing , Animals , Dishevelled Proteins , Immunohistochemistry , In Situ Hybridization , Micromanipulation , Microscopy, Electron, Scanning , Mutation/genetics , Neural Crest/ultrastructure , Phosphoproteins/genetics , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins , Xenopus Proteins
15.
Dev Dyn ; 229(1): 109-17, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14699582

ABSTRACT

In recent years, research on neural crest induction has allowed the identification of several molecules as candidates for neural crest inducers. Although many of these molecules have the ability to induce neural crest in different assays, a general mechanism of neural crest induction that includes a description of the tissues that produce the inductive signals and the time and steps in which this process takes place remains elusive. To better understand the mechanism of neural crest induction, we developed an assay that has been used previously by Nieuwkoop to study anterior-posterior pattern of the neural plate. Folds of competent ectoderm were implanted in different positions of a young neurula embryo, and the induction of neural crest was analyzed using the expression of the neural crest marker Xslug. We identified a very localized region of the early neurula where it is possible to get neural crest induction, whereas all of the regions tested showed a clear induction of the neural plate marker Xsox2. These results indicate that there is a region in the embryo with the appropriate combination of signals needed to induce neural crest cells; we called this region the neural crest competence territory. In addition, our results show that neural crest induction is always accompanied by neural plate induction, but there are many cases where neural plate was induced without neural crest. These results support the model in which the neural crest is induced by an interaction between neural plate and epidermis, but they also suggest that additional signals are required. By making grafts of different sizes and implanting them in the epidermis or the neural plate, we concluded that one of the inductive signals is produced in the dorsal region of the embryo and travels into the ectoderm. Finally, by performing gain- and loss-of-function of Wnt signaling experiments, we show that this pathway plays an important role not only in neural crest induction but also in the specification of the neural crest competence territory. Developmental Dynamics 229:109-117, 2004.


Subject(s)
Neural Crest/embryology , Neural Crest/metabolism , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins , Animals , Body Patterning , DNA-Binding Proteins/genetics , Ectoderm/transplantation , Embryonic Induction , Gene Expression Regulation, Developmental , HMGB Proteins , Models, Biological , Nuclear Proteins/genetics , SOXB1 Transcription Factors , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Wnt Proteins , Xenopus Proteins , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...