Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mucosal Immunol ; 16(4): 486-498, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37192682

RESUMEN

Curative therapies against autoimmune diseases are lacking. Indeed, most of the currently available treatments are only targeting symptoms. We have developed a novel strategy for a therapeutic vaccine against autoimmune diseases based on intranasal administration of a fusion protein tolerogen, which consists of a mutant, enzymatically inactive, cholera toxin A1 (CTA1)-subunit genetically fused to disease-relevant high-affinity peptides and a dimer of D-fragments from protein A (DD). The CTA1 R7K mutant - myelin oligodendrocyte glycoprotein (MOG), or proteolipid protein (PLP) - DD (CTA1R7K-MOG/PLP-DD) fusion proteins effectively reduced clinical symptoms in the experimental autoimmune encephalitis model of multiple sclerosis. The treatment induced Tr1 cells, in the draining lymph node, which produced interleukin (IL)-10 and suppressed effector clusters of differentiation 4+ T-cell responses. This effect was dependent on IL-27 signaling because treatment was ineffective in bone marrow chimeras lacking IL-27Ra within their hematopoietic compartment. Single-cell RNA sequencing of dendritic cells in draining lymph nodes demonstrated distinct gene transcriptional changes of classic dendritic cells 1, including enhanced lipid metabolic pathways, induced by the tolerogenic fusion protein. Thus, our results with the tolerogenic fusion protein demonstrate the possibility to vaccinate and protect against disease progression by reinstating tolerance in multiple sclerosis and other autoimmune diseases.


Asunto(s)
Esclerosis Múltiple , Linfocitos T Reguladores , Humanos , Administración Intranasal , Toxina del Cólera , Linfocitos T CD4-Positivos , Esclerosis Múltiple/tratamiento farmacológico
3.
Sci Immunol ; 7(73): eabc5500, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35776804

RESUMEN

T helper 17 (TH17) cells located at the Peyer's patch (PP) inductive site and at the lamina propria effector site of the intestinal immune system are responsive to both pathogenic and commensal bacteria. Their plasticity to convert into follicular helper T (TFH) cells has been proposed to be central to gut immunoglobulin A (IgA) responses. Here, we used an IL-17A fate reporter mouse and an MHC-II tetramer to analyze antigen-specific CD4+ T cell subsets and isolate them for single-cell RNA sequencing after oral immunization with cholera toxin and ovalbumin. We found a TFH-dominated response with only rare antigen-specific TH17 cells (<8%) in the PP. A clonotypic analysis provided little support that clonotypes were shared between TFH and TH17 cells, arguing against TH17 plasticity as a major contributor to TFH differentiation. Two mouse models of TH17 deficiency confirmed that gut IgA responses to oral immunization do not require TH17 cells, with CD4CreRorcfl/fl mice exhibiting normal germinal centers in PP and unperturbed total IgA production in the intestine.


Asunto(s)
Inmunoglobulina A , Ganglios Linfáticos Agregados , Células Th17 , Animales , Antígenos/inmunología , Toxina del Cólera , Inmunización , Inmunoglobulina A/inmunología , Ratones , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/inmunología , Células Th17/inmunología , Vacunación
4.
Mucosal Immunol ; 15(4): 745-761, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35418673

RESUMEN

Migratory dendritic cells expressing CD103 are the targets for mucosal vaccines. These belong to either of two lineage-restricted subsets, cDC1 or cDC2 cells, which have been linked to priming of functionally distinct CD4 T cells. However, recent studies have identified plasticity in cDC2 cells with overlapping functions with cDC1 cells, while the converse has not been reported. We genetically engineered a vaccine adjuvant platform that targeted the cholera toxin A1 (CTA1) ADP-ribosylating enzyme to CD103+ cDC1 and cDC2 cells using a single-chain antibody (scFv) to CD103. Unexpectedly, intranasal immunization with the CTA1-svFcCD103 adjuvant modified cDC1 cells to effectively prime Th17 cells, a function previously limited to cDC2 cells. In fact, cDC2 cells were dispensible, while cDC1 cells, lacking in Batf3-/- mice, were critical. Following intranasal immunizations isolated cDC1 cells from mLN exclusively promoted Rorgt+ T cells and IL-17, IL-21, and IL-22 production. Strong CD8 T cell responses through antigen cross presentation by cDC1 cells were also observed. Single-cell RNAseq analysis revealed upregulation of Th17-promoting gene signatures in sorted cDC1 cells. Gene expression in isolated cDC2 cells was largely unaffected. Our finding represents a major shift of paradigm as we have documented functional plasticity in cDC1 cells.


Asunto(s)
Gripe Humana , Infecciones por Orthomyxoviridae , Adenosina Difosfato/metabolismo , Adyuvantes Inmunológicos , Animales , Toxina del Cólera/metabolismo , Células Dendríticas , Humanos , Gripe Humana/metabolismo , Ratones , Infecciones por Orthomyxoviridae/metabolismo , Células Th17
5.
Mucosal Immunol ; 15(4): 717-729, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35260804

RESUMEN

The fate of tissue-resident memory CD4 T cells (Trm) has been incompletely investigated. Here we show that intranasal, but not parenteral, immunization with CTA1-3M2e-DD stimulated M2e-specific Th17 Trm cells, which conferred strong protection against influenza virus infection in the lung. These cells rapidly expanded upon infection and effectively restricted virus replication as determined by CD4 T cell depletion studies. Single-cell RNAseq transcriptomic and TCR VDJ-analysis of M2e-tetramer-sorted CD4 T cells on day 3 and 8 post infection revealed complete Th17-lineage dominance (no Th1 or Tregs) with extensive functional diversity and expression of gene markers signifying mature resident Trm cells (Cd69, Nfkbid, Brd2, FosB). Unexpectedly, the same TCR clonotype hosted cells with different Th17 subcluster functions (IL-17, IL-22), regulatory and cytotoxic cells, suggesting a tissue and context-dependent differentiation of reactivated Th17 Trm cells. A gene set enrichment analysis demonstrated up-regulation of regulatory genes (Lag3, Tigit, Ctla4, Pdcd1) in M2e-specific Trm cells on day 8, indicating a tissue damage preventing function. Thus, contrary to current thinking, lung M2e-specific Th17 Trm cells are sufficient for controlling infection and for protecting against tissue injury. These findings will have strong implications for vaccine development against respiratory virus infections and influenza virus infections, in particular.


Asunto(s)
Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Humanos , Memoria Inmunológica , Pulmón , Receptores de Antígenos de Linfocitos T , Células Th17
6.
Development ; 148(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34905617

RESUMEN

Development of the Drosophila visceral muscle depends on Anaplastic Lymphoma Kinase (Alk) receptor tyrosine kinase (RTK) signaling, which specifies founder cells (FCs) in the circular visceral mesoderm (VM). Although Alk activation by its ligand Jelly Belly (Jeb) is well characterized, few target molecules have been identified. Here, we used targeted DamID (TaDa) to identify Alk targets in embryos overexpressing Jeb versus embryos with abrogated Alk activity, revealing differentially expressed genes, including the Snail/Scratch family transcription factor Kahuli (Kah). We confirmed Kah mRNA and protein expression in the VM, and identified midgut constriction defects in Kah mutants similar to those of pointed (pnt). ChIP and RNA-Seq data analysis defined a Kah target-binding site similar to that of Snail, and identified a set of common target genes putatively regulated by Kah and Pnt during midgut constriction. Taken together, we report a rich dataset of Alk-responsive loci in the embryonic VM and functionally characterize the role of Kah in the regulation of embryonic midgut morphogenesis.


Asunto(s)
Quinasa de Linfoma Anaplásico , Proteínas de Unión al ADN , Proteínas de Drosophila , Desarrollo Embrionario , Proteínas del Tejido Nervioso , Proteínas Proto-Oncogénicas , Factores de Transcripción , Animales , Quinasa de Linfoma Anaplásico/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/genética , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Proteínas de Drosophila/genética , Desarrollo Embrionario/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Desarrollo de Músculos/genética , Músculos/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Proto-Oncogénicas/genética , ARN Mensajero/genética , RNA-Seq , Transducción de Señal/genética , Análisis de la Célula Individual , Factores de Transcripción/genética
7.
Cell Rep ; 36(2): 109372, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260908

RESUMEN

B lymphocytes are exquisitely sensitive to fluctuations in nutrient signaling by the Rag GTPases, and 15% of follicular lymphomas (FLs) harbor activating mutations in RRAGC. Hence, a potential therapeutic approach against malignant B cells is to inhibit Rag GTPase signaling, but because such inhibitors are still to be developed, efficacy and safety remain unknown. We generated knockin mice expressing a hypomorphic variant of RagC (Q119L); RagCQ119L/+ mice are viable and show attenuated nutrient signaling. B lymphocyte activation is cell-intrinsically impaired in RagCQ119L/+ mice, which also show significant suppression of genetically induced lymphomagenesis and autoimmunity. Surprisingly, no overt systemic trade-offs or phenotypic alterations caused by partial suppression of nutrient signaling are seen in other organs, and RagCQ119L/+ mice show normal longevity and normal age-dependent health decline. These results support the efficacy and safety of moderate inhibition of nutrient signaling against pathological B cells.


Asunto(s)
Linfocitos B/inmunología , Carcinogénesis/inmunología , Carcinogénesis/patología , Linfoma/inmunología , Linfoma/patología , Proteínas de Unión al GTP Monoméricas/metabolismo , Transducción de Señal , Animales , Femenino , Técnicas de Sustitución del Gen , Heterocigoto , Inmunidad Humoral , Longevidad , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Mutantes , Mutación/genética
8.
Genes Dev ; 35(15-16): 1190-1207, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34301765

RESUMEN

The meninges are important for brain development and pathology. Using single-cell RNA sequencing, we have generated the first comprehensive transcriptional atlas of neonatal mouse meningeal leukocytes under normal conditions and after perinatal brain injury. We identified almost all known leukocyte subtypes and found differences between neonatal and adult border-associated macrophages, thus highlighting that neonatal border-associated macrophages are functionally immature with regards to immune responses compared with their adult counterparts. We also identified novel meningeal microglia-like cell populations that may participate in white matter development. Early after the hypoxic-ischemic insult, neutrophil numbers increased and they exhibited increased granulopoiesis, suggesting that the meninges are an important site of immune cell expansion with implications for the initiation of inflammatory cascades after neonatal brain injury. Our study provides a single-cell resolution view of the importance of meningeal leukocytes at the early stage of development in health and disease.


Asunto(s)
Meninges , Microglía , Animales , Encéfalo/patología , Femenino , Leucocitos , Macrófagos , Ratones , Embarazo
9.
Cell Rep ; 35(12): 109286, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34161770

RESUMEN

B cell responses are critical for antiviral immunity. However, a comprehensive picture of antigen-specific B cell differentiation, clonal proliferation, and dynamics in different organs after infection is lacking. Here, by combining single-cell RNA and B cell receptor (BCR) sequencing of antigen-specific cells in lymph nodes, spleen, and lungs after influenza infection in mice, we identify several germinal center (GC) B cell subpopulations and organ-specific differences that persist over the course of the response. We discover transcriptional differences between memory cells in lungs and lymphoid organs and organ-restricted clonal expansion. Remarkably, we find significant clonal overlap between GC-derived memory and plasma cells. By combining BCR-mutational analyses with monoclonal antibody (mAb) expression and affinity measurements, we find that memory B cells are highly diverse and can be selected from both low- and high-affinity precursors. By linking antigen recognition with transcriptional programming, clonal proliferation, and differentiation, these finding provide important advances in our understanding of antiviral immunity.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos B/inmunología , Perfilación de la Expresión Génica , Gripe Humana/genética , Gripe Humana/inmunología , Receptores de Antígenos de Linfocitos B/metabolismo , Análisis de la Célula Individual , Animales , Anticuerpos Monoclonales/metabolismo , Diferenciación Celular/genética , Proliferación Celular , Células Clonales , Centro Germinal/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Células B de Memoria/metabolismo , Ratones Endogámicos C57BL , Mutación/genética , Tasa de Mutación , Especificidad de Órganos , Células Plasmáticas/metabolismo , ARN/metabolismo , Transcripción Genética
10.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33538776

RESUMEN

B cells emerge from the bone marrow as transitional (TS) B cells that differentiate through T1, T2, and T3 stages to become naive B cells. We have identified a bifurcation of human B cell maturation from the T1 stage forming IgMhi and IgMlo developmental trajectories. IgMhi T2 cells have higher expression of α4ß7 integrin and lower expression of IL-4 receptor (IL4R) compared with the IgMlo branch and are selectively recruited into gut-associated lymphoid tissue. IgMhi T2 cells also share transcriptomic features with marginal zone B cells (MZBs). Lineage progression from T1 cells to MZBs via an IgMhi trajectory is identified by pseudotime analysis of scRNA-sequencing data. Reduced frequency of IgMhi gut-homing T2 cells is observed in severe SLE and is associated with reduction of MZBs and their putative IgMhi precursors. The collapse of the gut-associated MZB maturational axis in severe SLE affirms its existence in health.


Asunto(s)
Diferenciación Celular/inmunología , Tracto Gastrointestinal/inmunología , Inmunoglobulina M/metabolismo , Nefritis Lúpica/inmunología , Tejido Linfoide/inmunología , Células Precursoras de Linfocitos B/inmunología , Adulto , Anciano , Donantes de Sangre , Estudios de Casos y Controles , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Células Cultivadas , Femenino , Humanos , Cadenas beta de Integrinas/metabolismo , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Nefritis Lúpica/sangre , Nefritis Lúpica/patología , Masculino , Persona de Mediana Edad , Fenotipo , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma , Adulto Joven
11.
Nat Metab ; 1(8): 775-789, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31579886

RESUMEN

The humoral immune response demands that B cells undergo a sudden anabolic shift and high cellular nutrient levels which are required to sustain the subsequent proliferative burst. Follicular lymphoma (FL) originates from B cells that have participated in the humoral response, and 15% of FL samples harbor point, activating mutations in RRAGC, an essential activator of mTORC1 downstream of the sensing of cellular nutrients. The impact of recurrent RRAGC mutations in B cell function and lymphoma is unexplored. RRAGC mutations, targeted to the endogenous locus in mice, confer a partial insensitivity to nutrient deprivation, but strongly exacerbate B cell responses and accelerate lymphomagenesis, while creating a selective vulnerability to pharmacological inhibition of mTORC1. This moderate increase in nutrient signaling synergizes with paracrine cues from the supportive T cell microenvironment that activates B cells via the PI3K-Akt-mTORC1 axis. Hence, Rragc mutations sustain induced germinal centers and murine and human FL in the presence of decreased T cell help. Our results support a model in which activating mutations in the nutrient signaling pathway foster lymphomagenesis by corrupting a nutrient-dependent control over paracrine signals from the T cell microenvironment.


Asunto(s)
GTP Fosfohidrolasas/metabolismo , Activación de Linfocitos , Linfoma Folicular/tratamiento farmacológico , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Humanos , Linfoma Folicular/patología , Ratones , Ratones Transgénicos
12.
Nat Commun ; 10(1): 2423, 2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31160559

RESUMEN

The germinal center (GC) reaction in Peyer's patches (PP) requires continuous access to antigens, but how this is achieved is not known. Here we show that activated antigen-specific CCR6+CCR1+GL7- B cells make close contact with M cells in the subepithelial dome (SED). Using in situ photoactivation analysis of antigen-specific SED B cells, we find migration of cells towards the GC. Following antigen injection into ligated intestinal loops containing PPs, 40% of antigen-specific SED B cells bind antigen within 2 h, whereas unspecifc cells do not, indicating B cell-receptor involvment. Antigen-loading is not observed in M cell-deficient mice, but is unperturbed in mice depleted of classical dendritic cells (DC). Thus, we report a M cell-B cell antigen-specific transporting pathway in PP that is independent of DC. We propose that this antigen transporting pathway has a critical role in gut IgA responses, and should be taken into account when developing mucosal vaccines.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos/inmunología , Linfocitos B/inmunología , Células Epiteliales/inmunología , Ganglios Linfáticos Agregados/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Animales , Células Dendríticas/inmunología , Centro Germinal/inmunología , Inmunoglobulina A/inmunología , Activación de Linfocitos , Ratones
13.
Curr Opin Pharmacol ; 41: 42-51, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29702466

RESUMEN

Most infections are caused by pathogens that access the body at mucosal sites. Hence, development of mucosal vaccines to prevent local infection or invasion of pathogens appears highly warranted, especially since only mucosal immunization will stimulate strong local IgA responses and tissue resident memory CD4 and CD8 T cells. The most significant obstacle to developing such vaccines is the lack of approved adjuvants that can effectively and safely enhance relevant mucosal and systemic immune responses. The most potent mucosal adjuvants known today are the adenosine diphosphate (ADP)-ribosylating bacterial enterotoxins cholera toxin (CT) and Escherichia coli heat-labile toxins (LTs). Unfortunately, these molecules are also very toxic, which precludes their clinical use. However, much effort has been devoted to developing derivatives of these enterotoxins with low or no toxicity and retained adjuvant activity. Although it is fair to say that we know more about how these toxins affect the immune system than ever before, we still lack a detailed understanding of how and why these toxins are effective adjuvants. In the present review, we provide a state-of-the-art overview of the mechanism of action of the holotoxins and the strategies used for improving the toxin-based adjuvants.


Asunto(s)
ADP-Ribosilación , Adyuvantes Inmunológicos/química , Adyuvantes Inmunológicos/toxicidad , Toxinas Bacterianas/química , Toxinas Bacterianas/toxicidad , Toxina del Cólera/química , Toxina del Cólera/toxicidad , Enterotoxinas/química , Enterotoxinas/toxicidad , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidad , Adyuvantes Inmunológicos/farmacología , Toxinas Bacterianas/farmacología , Toxina del Cólera/farmacología , Enterotoxinas/farmacología , Proteínas de Escherichia coli/farmacología , Humanos , Inmunidad Mucosa
14.
Front Immunol ; 8: 1133, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28959261

RESUMEN

Reinstating tissue-specific tolerance has attracted much attention as a means to treat autoimmune diseases. However, despite promising results in rodent models of autoimmune diseases, no established tolerogenic therapy is clinically available yet. In the experimental autoimmune myasthenia gravis (EAMG) model several protocols have been reported that induce tolerance against the prime disease-associated antigen, the acetylcholine receptor (AChR) at the neuromuscular junction. Using the whole AChR, the extracellular part or peptides derived from the receptor, investigators have reported variable success with their treatments, though, usually relatively large amounts of antigen has been required. Hence, there is a need for better formulations and strategies to improve on the efficacy of the tolerance-inducing therapies. Here, we report on a novel targeted fusion protein carrying the immunodominant peptide from AChR, mCTA1-T146, which given intranasally in repeated microgram doses strongly suppressed induction as well as ongoing EAMG disease in mice. The results corroborate our previous findings, using the same fusion protein approach, in the collagen-induced arthritis model showing dramatic suppressive effects on Th1 and Th17 autoaggressive CD4 T cells and upregulated regulatory T cell activities with enhanced IL10 production. A suppressive gene signature with upregulated expression of mRNA for TGFß, IL10, IL27, and Foxp3 was clearly detectable in lymph node and spleen following intranasal treatment with mCTA1-T146. Amelioration of EAMG disease was accompanied by reduced loss of muscle AChR and lower levels of anti-AChR serum antibodies. We believe this targeted highly effective fusion protein mCTA1-T146 is a promising candidate for clinical evaluation in myasthenia gravis patients.

15.
Immun Inflamm Dis ; 4(2): 191-200, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27957327

RESUMEN

Several Butyrophilin (BTN) and Btn-like (BTNL) molecules control T lymphocyte responses, and are genetically associated with inflammatory disorders and cancer. In this study, we present a comprehensive expression analysis of human and murine BTN and BTNL genes in conditions associated with intestinal inflammation and cancer. Using real-time PCR, expression of human BTN and BTNL genes was analyzed in samples from patients with ulcerative colitis, irritable bowel syndrome, and colon tumors. Expression of murine Btn and Btnl genes was examined in mouse models of spontaneous colitis (Muc2-/-) and intestinal tumorigenesis (ApcMin/+). Our analysis indicates a strong association of several of the human genes with ulcerative colitis and colon cancer; while especially BTN1A1, BTN2A2, BTN3A3, and BTNL8 were significantly altered in inflammation, colonic tumors exhibited significantly decreased levels of BTNL2, BTNL3, BTNL8, and BTNL9 as compared to unaffected tissue. Colonic inflammation in Muc2-/- mice significantly down-regulated the expression of particularly Btnl1, Btnl4, and Btnl6 mRNA, and intestinal polyps derived from ApcMin/+ mice displayed altered levels of Btn1a1, Btn2a2, and Btnl1 transcripts. Thus, our data present an association of BTN and BTNL genes with intestinal inflammation and cancer and represent a valuable resource for further studies of this gene family.


Asunto(s)
Butirofilinas/metabolismo , Neoplasias del Colon/metabolismo , Inflamación/metabolismo , Animales , Colitis Ulcerosa/metabolismo , Colon , Humanos , Mucosa Intestinal , Ratones , Ratones Endogámicos C57BL , Linfocitos T
16.
Sci Rep ; 6: 31524, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27528202

RESUMEN

Murine Butyrophilin-like (Btnl) 1 and Btnl6 are primarily restricted to intestinal epithelium where they regulate the function of intraepithelial T lymphocytes. We recently demonstrated that Btnl1 and Btnl6 can form an intra-family heterocomplex and that the Btnl1-Btnl6 complex selectively expands Vγ7Vδ4 TCR IELs. To define the regulation of Btnl expression in the small intestine during ontogeny we examined the presence of Btnl1 and Btnl6 in the small bowel of newborn to 4-week-old mice. Although RNA expression of Btnl1 and Btnl6 was detected in the small intestine at day 0, Btnl1 and Btnl6 protein expression was substantially delayed and was not detectable in the intestinal epithelium until the mice reached 2-3 weeks of age. The markedly elevated Btnl protein level at week 3 coincided with a significant increase of γδ TCR IELs, particularly those bearing the Vγ7Vδ4 receptor. This was not dependent on gut microbial colonization as mice housed in germ-free conditions had normal Btnl protein levels. Taken together, our data show that the expression of Btnl1 and Btnl6 is delayed in the murine neonatal gut and that the appearance of the Btnl1 and Btnl6 proteins in the intestinal mucosa associates with the expansion of Vγ7Vδ4 TCR IELs.


Asunto(s)
Butirofilinas/metabolismo , Intestino Delgado/metabolismo , Animales , Animales Recién Nacidos , Butirofilinas/genética , Femenino , Vida Libre de Gérmenes , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo , ARN/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología
17.
Front Immunol ; 7: 1, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26834743

RESUMEN

To date, few molecular conduits mediating the cross-talk between intestinal epithelial cells and intraepithelial lymphocytes (IELs) have been described. We recently showed that butyrophilin-like (Btnl) 1 can attenuate the epithelial response to activated IELs, resulting in reduced production of proinflammatory mediators, such as IL-6 and CXCL1. We here report that like Btnl1, murine Btnl6 expression is primarily confined to the intestinal epithelium. Although Btnl1 can exist in a cell surface-expressed homomeric form, we found that it additionally forms heteromeric complexes with Btnl6, and that the engagement of Btnl1 is a prerequisite for surface expression of Btnl6 on intestinal epithelial cells. In an IEL-epithelial cell coculture system, enforced epithelial cell expression of Btnl1 significantly enhanced the proliferation of IELs in the absence of exogenous activation. The effect on proliferation was dependent on the presence of IL-2 or IL-15 and restricted to IELs upregulating CD25. In the γδ T-cell subset, the Btnl1-Btnl6 complex, but not Btnl1, specifically elevated the proliferation of IELs bearing the Vγ7Vδ4 receptor. Thus, our results show that murine epithelial cell-specific Btnl proteins can form intrafamily heterocomplexes and suggest that the interaction between Btnl proteins and IELs regulates the expansion of IELs in the intestinal mucosa.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...