Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Immunity ; 46(2): 233-244, 2017 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-28214225

RESUMEN

Arginase 1 (Arg1) and indoleamine 2,3-dioxygenase 1 (IDO1) are immunoregulatory enzymes catalyzing the degradation of l-arginine and l-tryptophan, respectively, resulting in local amino acid deprivation. In addition, unlike Arg1, IDO1 is also endowed with non-enzymatic signaling activity in dendritic cells (DCs). Despite considerable knowledge of their individual biology, no integrated functions of Arg1 and IDO1 have been reported yet. We found that IDO1 phosphorylation and consequent activation of IDO1 signaling in DCs was strictly dependent on prior expression of Arg1 and Arg1-dependent production of polyamines. Polyamines, either produced by DCs or released by bystander Arg1+ myeloid-derived suppressor cells, conditioned DCs toward an IDO1-dependent, immunosuppressive phenotype via activation of the Src kinase, which has IDO1-phosphorylating activity. Thus our data indicate that Arg1 and IDO1 are linked by an entwined pathway in immunometabolism and that their joint modulation could represent an important target for effective immunotherapy in several disease settings.


Asunto(s)
Arginasa/inmunología , Células Dendríticas/inmunología , Tolerancia Inmunológica/fisiología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Transducción de Señal/inmunología , Animales , Arginasa/metabolismo , Arginina/inmunología , Arginina/metabolismo , Western Blotting , Células Dendríticas/metabolismo , Femenino , Perfilación de la Expresión Génica , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma , Triptófano/inmunología , Triptófano/metabolismo
2.
Nat Immunol ; 13(9): 843-50, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22863752

RESUMEN

A large gap in our understanding of infant immunity is why natural killer (NK) cell responses are deficient, which makes infants more prone to viral infection. Here we demonstrate that transforming growth factor-ß (TGF-ß) was responsible for NK cell immaturity during infancy. We found more fully mature NK cells in CD11c(dnR) mice, whose NK cells lack TGF-ß receptor (TGF-ßR) signaling. Ontogenic maturation of NK cells progressed faster in the absence of TGF-ß signaling, which results in the formation of a mature NK cell pool early in life. As a consequence, infant CD11c(dnR) mice efficiently controlled viral infections. These data thus demonstrate an unprecedented role for TGF-ß in ontogeny that can explain why NK cell responses are deficient early in life.


Asunto(s)
Sistema Inmunológico/citología , Sistema Inmunológico/crecimiento & desarrollo , Células Asesinas Naturales/citología , Factor de Crecimiento Transformador beta/inmunología , Animales , Animales Recién Nacidos , Diferenciación Celular/inmunología , Citometría de Flujo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Madre , Factor de Crecimiento Transformador beta/metabolismo
3.
J Immunol ; 200(6): 1982-1987, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29440507

RESUMEN

B6.SJL-Ptprca Pepcb /Boy (CD45.1) mice have been used in hundreds of congenic competitive transplants, with the presumption that they differ from C57BL/6 mice only at the CD45 locus. In this study, we describe a point mutation in the natural cytotoxicity receptor 1 (Ncr1) locus fortuitously identified in the CD45.1 strain. This point mutation was mapped at the 40th nucleotide of the Ncr1 locus causing a single amino acid mutation from cysteine to arginine at position 14 from the start codon, resulting in loss of NCR1 expression. We found that these mice were more resistant to CMV due to a hyper innate IFN-γ response in the absence of NCR1. In contrast, loss of NCR1 increased susceptibility to influenza virus, a result that is consistent with the role of NCR1 in the recognition of influenza Ag, hemagglutinin. This work sheds light on potential confounding experimental interpretation when this congenic strain is used as a tool for tracking lymphocyte development.


Asunto(s)
Antígenos Ly/genética , Antígenos Comunes de Leucocito/genética , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Mutación Puntual/genética , Animales , Hemaglutininas/inmunología , Inmunidad Innata , Interferón gamma/inmunología , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/genética
4.
Gut ; 67(5): 847-859, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28341747

RESUMEN

BACKGROUND: Interactions between host immune cells and gut microbiota are crucial for the integrity and function of the intestine. How these interactions regulate immune cell responses in the intestine remains a major gap in the field. AIM: We have identified the signalling lymphocyte activation molecule family member 4 (SLAMF4) as an immunomodulator of the intestinal immunity. The aim is to determine how SLAMF4 is acquired in the gut and what its contribution to intestinal immunity is. METHODS: Expression of SLAMF4 was assessed in mice and humans. The mechanism of induction was studied using GFPtg bone marrow chimaera mice, lymphotoxin α and TNLG8A-deficient mice, as well as gnotobiotic mice. Role in immune protection was revealed using oral infection with Listeria monocytogenes and Cytobacter rodentium. RESULTS: SLAMF4 is a selective marker of intestinal immune cells of mice and humans. SLAMF4 induction occurs directly in the intestinal mucosa without the involvement of the gut-associated lymphoid tissue. Gut bacterial products, particularly those of gut anaerobes, and gut-resident antigen-presenting cell (APC) TNLG8A are key contributors of SLAMF4 induction in the intestine. Importantly, lack of SLAMF4 expression leads the increased susceptibility of mice to infection by oral pathogens culminating in their premature death. CONCLUSIONS: SLAMF4 is a marker of intestinal immune cells which contributes to the protection against enteric pathogens and whose expression is dependent on the presence of the gut microbiota. This discovery provides a possible mechanism for answering the long-standing question of how the intertwining of the host and gut microbial biology regulates immune cell responses in the gut.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa/fisiología , Mucosa Intestinal/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Animales , Citometría de Flujo , Vida Libre de Gérmenes , Humanos , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Simbiosis
5.
J Immunol ; 195(3): 1112-20, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26116500

RESUMEN

A small pool of NK1.1(+) CD8(+) T cells is harbored among the conventional CD8(+) T cell compartment. Conclusions drawn from the analysis of immune responses mediated by cytotoxic CD8(+) T cells are often based on the total population, which includes these contaminating NK1.1(+) CD8(+) T cells. An unresolved question is whether NK1.1(+) CD8(+) cells are conventional T cells that acquire NK1.1 expression upon activation or delineation into memory phenotype or whether they are a distinct cell population that induces immune responses in a different manner than conventional T cells. To address this question, we used the Listeria monocytogenes model of infection and followed CD8(+) NK1.1(+) T cells and NK1.1(-) CD8(+) T cells during each phase of the immune response: innate, effector, and memory. Our central finding is that CD8(+) NK1.1(+) cells and conventional NK1.1(-) CD8(+) T cells both contribute to the adaptive immune response to Listeria, but only CD8(+) NK1.1(+) cells were equipped with the ability to provide a rapid innate immune response, as demonstrated by early and Ag-independent IFN-γ production, granzyme B expression, and degranulation. More importantly, purified conventional CD8(+) T cells alone, in the absence of any contaminating CD8(+) NK1.1(+) cells, were not sufficient to provide early protection to lethally infected mice. These results highlight the role of CD8(+) NK1.1(+) T cells in mounting early innate responses that are important for host defense and support the therapeutic potential of this subset to improve the effectiveness of protective immunity.


Asunto(s)
Inmunidad Innata/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Células T Asesinas Naturales/inmunología , Linfocitos T Citotóxicos/inmunología , Traslado Adoptivo , Animales , Antígenos Ly/genética , Granzimas/biosíntesis , Interferón gamma/biosíntesis , Listeriosis/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Subfamilia B de Receptores Similares a Lectina de Células NK/genética , Células T Asesinas Naturales/citología , Células T Asesinas Naturales/trasplante , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/trasplante
6.
Proc Natl Acad Sci U S A ; 111(35): 12841-6, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25143585

RESUMEN

Invariant natural killer T (iNKT) cells to date represent the best example of cells known to have a hybrid function, representing both innate and adaptive immunity. Shared phenotypic similarities with NK cells together with a rapid response to a cytokine stimulus and a productive TCR engagement are the features that underline the hybrid nature of iNKT cells. Using these criteria, we provide molecular and functional evidence demonstrating that CD1d-independent (CD1d(ind)) NKT cells, a population of CD1d-unrestricted NKT cells, are endowed with a hybrid function far superior to that of iNKT cells: (i) an extensive shared program with NK cells, (ii) a closer Euclidian distance with NK cells, and (iii) the ability to respond to innate stimuli (Poly:IC) with cytotoxic potential in the same manner as NK cells identify a hybrid feature in CD1d(ind)NKT cells that truly fulfills the dual function of an NK and a T cell. Our finding that CD1d(ind)NKT cells are programmed to act like NK cells in response to innate signals while being capable of adaptive responses is unprecedented, and thus might reemphasize CD1d-unrestricted NKT cells as a subset of lymphocytes that could affect biological processes of antimicrobial and tumor immunity in a unique way.


Asunto(s)
Inmunidad Adaptativa/inmunología , Antígenos CD1d/inmunología , Células T Asesinas Naturales/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos CD1d/genética , Linaje de la Célula/inmunología , Femenino , Genómica , Granzimas/inmunología , Inmunofenotipificación , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Am J Physiol Lung Cell Mol Physiol ; 308(1): L86-95, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25361568

RESUMEN

Hematopoietic stem cell transplantation (HSCT) is complicated by pulmonary infections that manifest posttransplantation. Despite engraftment, susceptibility to infections persists long after reconstitution. Previous work using a murine bone marrow transplant (BMT) model implicated increased cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) in promoting impaired alveolar macrophage (AM) responses. However, mechanisms driving COX-2 overexpression remained elusive. Previously, transforming growth factor-ß (TGF-ß) signaling after BMT was shown to promote hypomethylation of the COX-2 gene. Here, we provide mechanistic insight into how this occurs and show that TGF-ß induces microRNA (miR)-29b while decreasing DNA methyltransferases (DNMT)1, DNMT3a, and DNMT3b in AMs after BMT. De novo DNMT3a and DNMT3b were decreased upon transient transfection of miR-29b, resulting in decreased methylation of the COX-2 promoter and induction of COX-2. As a consequence, miR-29b-driven upregulation of COX-2 promoted AM dysfunction, and transfection of BMT AMs with a miR-29b inhibitor rescued the bacterial-killing defect. MiR-29b-mediated defects in BMT AMs were dependent on increased levels of PGE2, as miR-29b-transfected AMs treated with a novel E prostanoid receptor 2 antagonist abrogated the impaired bacterial killing. We also demonstrate that patients that have undergone HSCT exhibit increased miR-29b; thus these studies highlight miR-29b in driving defective AM responses and identify this miRNA as a potential therapeutic target.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Células Madre Hematopoyéticas , Macrófagos Alveolares/metabolismo , MicroARNs/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto , Aloinjertos , Animales , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Dinoprostona/genética , Dinoprostona/metabolismo , Femenino , Humanos , Macrófagos Alveolares/patología , Masculino , Ratones , MicroARNs/genética , Persona de Mediana Edad , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética
8.
Am J Physiol Lung Cell Mol Physiol ; 307(3): L219-30, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24879051

RESUMEN

Patients with idiopathic pulmonary fibrosis (IPF) often do worse following infection, but the cause of the decline is not fully understood. We previously demonstrated that infection with a murine gamma herpes virus (γHV-68) could exacerbate established lung fibrosis following administration of fluorescein isothiocyanate (McMillan et al. Am J Respir Crit Care Med 177: 771-780, 2008). In the present study, we anesthetized mice and injected saline or bleomycin intratracheally on day 0. On day 14, mice were anesthetized again and infected with either a Gram-negative bacteria (Pseudomonas aeruginosa), or with H1N1 or γHV-68 viruses. Measurements were then made on days 15, 21, or 35. We demonstrate that infection with P. aeruginosa does not exacerbate extracellular matrix deposition post-bleomycin. Furthermore, fibrotic mice are effectively able to clear P. aeruginosa infection. In contrast, bleomycin-treated mice develop worse lung fibrosis when infected with γHV-68, but not when infected with H1N1. The differential ability of γHV-68 to cause increased collagen deposition could not be explained by differences in inflammatory cell recruitment or whole lung chemokine and cytokine responses. Alveolar epithelial cells from γHV-68-infected mice displayed increased expression of TGFß receptor 1, increased SMAD3 phosphorylation, and evidence of apoptosis measured by cleaved poly-ADP ribose polymerase (PARP). The ability of γHV-68 to augment fibrosis required the ability of the virus to reactivate from latency. This property appears unique to γHV-68, as the ß-herpes virus, cytomegalovirus, did not have the same effect.


Asunto(s)
Herpesviridae/patogenicidad , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Pseudomonas aeruginosa/patogenicidad , Fibrosis Pulmonar/microbiología , Fibrosis Pulmonar/virología , Animales , Apoptosis/efectos de los fármacos , Bleomicina/efectos adversos , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Células Epiteliales/patología , Células Epiteliales/virología , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/virología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
9.
J Immunol ; 189(9): 4528-36, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23008450

RESUMEN

Hematopoietic stem cell transplant therapy is limited by pulmonary infections. Mice with fully reconstituted hematopoietic compartments, including alveolar macrophages (AMs), after bone marrow transplantation (BMT) have impaired host defense against Gram-negative Pseudomonas aeruginosa. Impaired innate immunity is related to increased production of PGE(2) by AMs. Cyclooxygenase (COX)-2 is the rate-limiting enzyme for synthesis of PGE(2) from arachidonic acid, and COX-2 expression is elevated in AMs post-BMT. We hypothesized that epigenetic mechanisms may be responsible for upregulation of COX-2 in AMs. Using bisulfite sequencing, we observed the 5'-untranslated region and exon 1 of the COX-2 gene is hypomethylated in the AMs of BMT mice compared with control. COX-2 expression was increased in primary AMs and in the AM cell line (MHS) after treatment with 5-aza-2'-deoxycytidine (a methyltransferase inhibitor). Methylation by SssI methyltransferase of a 698-bp region of the COX-2 promoter including the beginning of exon 1 driving a luciferase reporter silenced luciferase expression. Because TGF-ß1 is elevated in lungs post-BMT, we tested whether TGF-ß1 could promote expression of COX-2 in a hypermethylated COX-2 vector, and observed TGF-ß1-induced modest expression of COX-2, suggesting an ability to demethylate the promoter. Finally, BMTs performed with marrow from mice expressing a dominant-negative form of the TGF-ßRII on CD11c-expressing cells (which includes AMs) demonstrated improved host defense and AM function. Our findings suggest impaired innate immunity and PGE(2) elevation post-BMT are due to hypomethylation of the COX-2 gene, which is at least partly regulated by TGF-ß1.


Asunto(s)
Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Metilación de ADN/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología , Regiones no Traducidas 5'/inmunología , Animales , Línea Celular , Ciclooxigenasa 2/deficiencia , Metilación de ADN/genética , Exones/inmunología , Técnicas de Silenciamiento del Gen , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología
10.
Eur Heart J ; 34(48): 3717-27, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22613345

RESUMEN

AIMS: The importance of transforming growth factor beta (TGFß) as an immune regulatory cytokine in atherosclerosis has been established. However, the role of TGFß signalling in dendritic cells (DCs) and in DC-mediated T cell proliferation and differentiation in atherosclerosis is unknown. METHODS AND RESULTS: Here, we investigated the effect of disrupted TGFß signalling in DCs on atherosclerosis by using mice carrying a transgene resulting in functional inactivation of TGFß receptor II (TGFßRII) signalling in CD11c(+) cells (Apoe(-/-)CD11cDNR). Apoe(-/-)CD11cDNR mice exhibited an over two-fold increase in the plaque area compared with Apoe(-/-) mice. Plaques of Apoe(-/-)CD11cDNR mice showed an increase in CD45(+) leucocyte content, and specifically in CD3(+), CD4(+) and CD8(+) cells, whereas macrophage content was not affected. In lymphoid organs, Apoe(-/-)CD11cDNR mice had equal amounts of CD11c(+) cells, and CD11c(+)CD8(+) and CD11c(+)CD8(-) subsets, but showed a subtle shift in the CD11c(+)CD8(-) population towards the more inflammatory CD11c(+)CD8(-)CD4(-) DC subset. In addition, the number of plasmacytoid-DCs decreased. Maturation markers such as MHCII, CD86 and CD40 on CD11c(hi) cells did not change, but the CD11cDNR DCs produced more TNFα and IL-12. CD11c(+) cells from CD11cDNR mice strongly induced T-cell proliferation and activation, resulting in increased amounts of effector T cells producing high amounts of Th1 (IFN-γ), Th2 (IL-4, IL-10), Th17 (IL-17), and Treg (IL-10) cytokines. CONCLUSION: Here, we show that loss of TGFßRII signalling in CD11c(+) cells induces subtle changes in DC subsets, which provoke uncontrolled T cell activation and maturation. This results in increased atherosclerosis and an inflammatory plaque phenotype during hypercholesterolaemia.


Asunto(s)
Aterosclerosis/inmunología , Células Dendríticas/inmunología , Inmunidad Celular/inmunología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Linfocitos T/inmunología , Animales , Antígenos CD11/inmunología , Células Cultivadas , Hipercolesterolemia/inmunología , Ratones , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/deficiencia , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Transducción de Señal/inmunología
11.
bioRxiv ; 2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38659946

RESUMEN

In an era where the established lines between cell identities are blurred by intra-lineage plasticity, distinguishing between stable and transitional states becomes imperative. This challenge is particularly pronounced within the Group 1 ILC lineage, where the similarity and plasticity between NK cells and ILC1s obscure their classification and the assignment of their unique contributions to immune regulation. This study exploits the unique property of Asialo-GM1 (AsGM1)-a membrane lipid associated with cytotoxic attributes absent in ILC1s-as a definitive criterion to distinguish between these cells. By prioritizing cytotoxic potential as the cardinal differentiator, our strategic use of the AsGM1 signature achieved precise delineation of NK cells and ILC1s across tissues, validated by RNA-seq analysis. This capability extends beyond steady-state classifications, adeptly capturing the binary classification of NK cells and ILC1s during acute liver injury. By leveraging two established models of NK-to-ILC1 plasticity driven by TGFß and Toxoplasma gondii , we demonstrate the stability of the AsGM1 signature, which sharply contrasts with the loss of Eomes. This signature identified a spectrum of known and novel NK cell derivatives-ILC1-like entities that bridge traditional binary classifications in aging and infection. The early detection of the AsGM1 signature at the immature NK (iNK) stage, preceding Eomes, and its stability, unaffected by transcriptional reprogramming that typically alters Eomes, position AsGM1 as a unique, site-agnostic marker for fate mapping NK-to-ILC1 plasticity. This provides a powerful tool to explore the expanding heterogeneity within the Group 1 ILC landscape, effectively transcending the ambiguity inherent to the NK-to-ILC1 continuum.

12.
J Exp Med ; 203(2): 413-24, 2006 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-16461338

RESUMEN

The live attenuated yellow fever vaccine 17D (YF-17D) is one of the most effective vaccines available, with a 65-yr history of use in >400 million people globally. Despite this efficacy, there is presently no information about the immunological mechanisms by which YF-17D acts. Here, we present data that suggest that YF-17D activates multiple Toll-like receptors (TLRs) on dendritic cells (DCs) to elicit a broad spectrum of innate and adaptive immune responses. Specifically, YF-17D activates multiple DC subsets via TLRs 2, 7, 8, and 9 to elicit the proinflammatory cytokines interleukin (IL)-12p40, IL-6, and interferon-alpha. Interestingly, the resulting adaptive immune responses are characterized by a mixed T helper cell (Th)1/Th2 cytokine profile and antigen-specific CD8+ T cells. Furthermore, distinct TLRs appear to differentially control the Th1/Th2 balance; thus, whilst MyD88-deficient mice show a profound impairment of Th1 cytokines, TLR2-deficient mice show greatly enhanced Th1 and Tc1 responses to YF-17D. Together, these data enhance our understanding of the molecular mechanism of action of YF-17D, and highlight the potential of vaccination strategies that use combinations of different TLR ligands to stimulate polyvalent immune responses.


Asunto(s)
Células Dendríticas/inmunología , Receptores Toll-Like/fisiología , Vacuna contra la Fiebre Amarilla/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Células Dendríticas/metabolismo , Humanos , Inmunidad Activa , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Factor 88 de Diferenciación Mieloide , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/fisiología , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 7/fisiología , Receptor Toll-Like 8/fisiología , Receptor Toll-Like 9/fisiología , Vacunas Atenuadas/inmunología
13.
J Clin Invest ; 119(1): 182-92, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19104149

RESUMEN

Parkinson disease (PD) is a neurodegenerative disorder characterized by a loss of dopamine-containing neurons. Mounting evidence suggests that dopaminergic cell death is influenced by the innate immune system. However, the pathogenic role of the adaptive immune system in PD remains enigmatic. Here we showed that CD8+ and CD4+ T cells but not B cells had invaded the brain in both postmortem human PD specimens and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD during the course of neuronal degeneration. We further demonstrated that MPTP-induced dopaminergic cell death was markedly attenuated in the absence of mature T lymphocytes in 2 different immunodeficient mouse strains (Rag1-/- and Tcrb-/- mice). Importantly, similar attenuation of MPTP-induced dopaminergic cell death was seen in mice lacking CD4 as well as in Rag1-/- mice reconstituted with FasL-deficient splenocytes. However, mice lacking CD8 and Rag1-/- mice reconstituted with IFN-gamma-deficient splenocytes were not protected. These data indicate that T cell-mediated dopaminergic toxicity is almost exclusively arbitrated by CD4+ T cells and requires the expression of FasL but not IFNgamma. Further, our data may provide a rationale for targeting the adaptive arm of the immune system as a therapeutic strategy in PD.


Asunto(s)
Encéfalo , Linfocitos T CD4-Positivos/inmunología , Degeneración Nerviosa , Enfermedad de Parkinson , Trastornos Parkinsonianos , Anciano , Anciano de 80 o más Años , Animales , Encéfalo/citología , Encéfalo/inmunología , Encéfalo/patología , Linfocitos T CD4-Positivos/citología , Muerte Celular/fisiología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteína Ligando Fas/genética , Proteína Ligando Fas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Sistema Inmunológico/fisiología , Interferón gamma/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Neuronas/citología , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/inmunología , Enfermedad de Parkinson/patología , Trastornos Parkinsonianos/inmunología , Trastornos Parkinsonianos/patología
14.
Am J Pathol ; 179(5): 2382-96, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21924228

RESUMEN

Pulmonary infections and pneumonitis occur frequently after hematopoietic stem cell transplantation. Using a syngeneic mouse model of bone marrow transplantation (BMT), we have previously demonstrated that BMT mice are more susceptible to acute gammaherpesvirus 68 (MHV-68) replication at day 7 after infection. By day 21, the virus is latent in lungs of BMT and control mice, and there is no difference in viral load. Despite similar latent viral load, BMT mice develop severe pneumonitis associated with reduced oxygen saturation, fibrosis, peripheral inflammation, hyaline membranes, and foamy alveolar macrophages, a phenotype that persists for 7 weeks after infection. BMT mice demonstrate increased bronchoalveolar lavage (BAL) cells, and this population is enriched in neutrophils and T cells. Alternatively, activated macrophages appear earlier than do classically activated macrophages. BAL fluid from BMT mice at day 21 after infection contains increased levels of hydrogen peroxide, nitrite, and transforming growth factor-ß (TGF-ß). Mice expressing the dominant-negative transgene dn-TGFßRII in multiple cell types were used as BMT donors. BMT mice with T-cell dnTGFßRII are largely protected from the pneumonitis phenotype, whereas mice with CD11c-dnTGFßRII BMT mice are only modestly protected from pneumonitis. Protection in BMT mice with T-cell dnTGFßRII is associated with decreased TGF-ß derived from parenchymal cells in the BAL fluid, lower nitrite levels, and reduced apoptosis, whereas alternatively activated macrophage markers are unchanged.


Asunto(s)
Trasplante de Médula Ósea/efectos adversos , Gammaherpesvirinae , Infecciones por Herpesviridae , Neumonía/virología , Fibrosis Pulmonar/virología , Factor de Crecimiento Transformador beta/fisiología , Animales , Apoptosis/fisiología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/virología , Activación de Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Infiltración Neutrófila , Transducción de Señal/fisiología , Linfocitos T Reguladores/fisiología , Transgenes , Carga Viral
15.
J Immunol ; 183(11): 7297-306, 2009 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19917689

RESUMEN

Chronic allograft rejection (CR) is the main barrier to long-term transplant survival. CR is a progressive disease defined by interstitial fibrosis, vascular neointimal development, and graft dysfunction. The underlying mechanisms responsible for CR remain poorly defined. TGFbeta has been implicated in promoting fibrotic diseases including CR, but is beneficial in the transplant setting due to its immunosuppressive activity. To assess the requirement for T cell TGFbeta signaling in allograft acceptance and the progression of CR, we used mice with abrogated T cell TGFbeta signaling as allograft recipients. We compared responses from recipients that were transiently depleted of CD4(+) cells (that develop CR and express intragraft TGFbeta) with responses from mice that received anti-CD40L mAb therapy (that do not develop CR and do not express intragraft TGFbeta). Allograft acceptance and suppression of graft-reactive T and B cells were independent of T cell TGFbeta signaling in mice treated with anti-CD40L mAb. In recipients transiently depleted of CD4(+) T cells, T cell TGFbeta signaling was required for the development of fibrosis associated with CR, long-term graft acceptance, and suppression of graft-reactive T and B cell responses. Furthermore, IL-17 was identified as a critical element in TGFbeta-driven allograft fibrosis. Thus, IL-17 may provide a therapeutic target for preventing graft fibrosis, a measure of CR, while sparing the immunosuppressive activity of TGFbeta.


Asunto(s)
Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Interleucina-17/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Enfermedad Crónica , Fibrosis , Citometría de Flujo , Rechazo de Injerto/patología , Trasplante de Corazón/inmunología , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Trasplante Homólogo
16.
Proc Natl Acad Sci U S A ; 105(31): 10865-70, 2008 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-18669656

RESUMEN

One unresolved issue in immune tolerance is what prevents self-reactive T cells from activation. In this study, we used a transgenic mouse model of targeted functional inactivation of TGF-betaR signaling in CD11c(+) cells (CD11c(dnR) mice) and showed a direct impact on the development of experimental autoimmune encephalomyelitis (EAE). We found that MOG(35-55) immunization of CD11c(dnR) mice results in strong inflammation of CNS, high frequency of T cells in CNS, increased levels of T helper 1 (T(H)1) and T(H)17 cytokines in the periphery, and lack of remission from EAE. Once crossed with mice prone to autoimmunity, double-transgenic CD11c(dnR)Mog(TCR) mice revealed a spontaneous EAE-like disease characterized by early infiltration of activated myelin-specific T cells into CNS, activation of microglial cells, inflammation of CNS, dysfunction of locomotion, and premature death. We constructed chimeric mice and demonstrated that inactivation of TGF-betaR signaling in dendritic cells (DCs) results in augmented EAE-associated T cell responses. Our data provide direct evidence that TGF-beta can control autoimmunity via actions on DCs.


Asunto(s)
Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Glicoproteínas/inmunología , Tolerancia Inmunológica/inmunología , Fragmentos de Péptidos/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Traslado Adoptivo , Análisis de Varianza , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito
17.
PLoS One ; 11(6): e0156762, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27285085

RESUMEN

Despite our knowledge of the protective role of antibodies passed to infants through breast milk, our understanding of immunity transfer via maternal leukocytes is still limited. To emulate the immunological interface between the mother and her infant while breast-feeding, we used murine pups fostered after birth onto MHC-matched and MHC-mismatched dams. Overall, data revealed that: 1) Survival of breast milk leukocytes in suckling infants is possible, but not significant after the foster-nursing ceases; 2) Most breast milk lymphocytes establish themselves in specific areas of the intestine termed Peyer's patches (PPs); 3) While most leukocytes in the milk bolus were myeloid cells, the majority of breast milk leukocytes localized to PPs were T lymphocytes, and cytotoxic T cells (CTLs) in particular; 4) These CTLs exhibit high levels of the gut-homing molecules α4ß7 and CCR9, but a reduced expression of the systemic homing marker CD62L; 5) Under the same activation conditions, transferred CD8 T cells through breast milk have a superior capacity to produce potent cytolytic and inflammatory mediators when compared to those generated by the breastfed infant. It is therefore possible that maternal CTLs found in breast milk are directed to the PPs to compensate for the immature adaptive immune system of the infant in order to protect it against constant oral infectious risks during the postnatal phase.


Asunto(s)
Quimiotaxis de Leucocito , Inmunidad Materno-Adquirida/inmunología , Inmunización Pasiva , Lactancia/inmunología , Leche/inmunología , Ganglios Linfáticos Agregados/inmunología , Linfocitos T Citotóxicos/fisiología , Animales , Animales Recién Nacidos , Animales Lactantes , Células Cultivadas , Quimiotaxis de Leucocito/fisiología , Femenino , Inmunización Pasiva/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Leche/citología , Madres , Ganglios Linfáticos Agregados/citología , Linfocitos T Citotóxicos/citología
18.
PLoS One ; 9(7): e102390, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25072375

RESUMEN

While the role of Transforming Growth Factor ß (TGF-ß) as an intrinsic pathway has been well established in driving de novo differentiation of Th17 cells, no study has directly assessed the capacity of TGF-ß signaling initiated within dendritic cells (DCs) to regulate Th17 differentiation. The central finding of this study is the demonstration that Th17 cell fate during autoimmune inflammation is shaped by TGF-ß extrinsic pathway via DCs. First, we provide evidence that TGF-ß limits at the site of inflammation the differentiation of highly mature DCs as a means of restricting Th17 cell differentiation and controlling autoimmunity. Second, we demonstrate that TGF-ß controls DC differentiation in the inflammatory site but not in the priming site. Third, we show that TGF-ß controls DC numbers at a precursor level but not at a mature stage. While it is undisputable that TGF-ß intrinsic pathway drives Th17 differentiation, our data provide the first evidence that TGF-ß can restrict Th17 differentiation via DC suppression but such a control occurs in the site of inflammation, not at the site of priming. Such a demarcation of the role of TGF-ß in DC lineage is unprecedented and holds serious implications vis-à-vis future DC-based therapeutic targets.


Asunto(s)
Diferenciación Celular , Células Dendríticas/metabolismo , Transducción de Señal , Células Th17/citología , Células Th17/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antígeno CD11c , Recuento de Células , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Ratones , Imagen Molecular , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Índice de Severidad de la Enfermedad , Células Th17/inmunología
19.
Am J Physiol Cell Physiol ; 296(1): C193-204, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19005162

RESUMEN

Increased extracellular pressure stimulates beta1-integrin-dependent cancer cell adhesion. We asked whether pressure-induced adhesion is mediated by changes in beta1-integrin binding affinity or avidity and whether these changes are phosphorylation dependent. We evaluated integrin affinity and clustering in human SW620 colon cancer cells by measuring differences in binding between soluble Arg-Gly-Asp (RGD)-Fc ligands and RGD-Fc-F(ab')2 multimeric complexes under ambient and 15-mmHg increased pressures. Phosphorylation of beta1-integrin S785 and T788/9 residues in SW620 and primary malignant colonocytes was assessed in parallel. We further used GD25-beta1-integrin-null murine fibroblasts stably transfected with either wild-type beta1A-integrin, S785A, TT788/9AA, or T788D mutants to investigate the role of beta1-integrin site-specific phosphorylation. SW620 binding of RGD-Fc-F(ab')2 multimeric complexes, but not soluble RGD-Fc ligands, was sensitive to integrin clustering. RGD-Fc ligand binding was significantly increased under elevated pressure, suggesting that pressure modulates beta1-integrin affinity. Pressure stimulated both beta1-integrin S785 and T788/9 phosphorylation. GD25-beta1A-integrin wild-type and S785A cells displayed an increase in adhesion to fibronectin under elevated pressure, an effect absent in beta1-integrin-null and TT788/9AA cells. T788D substitution significantly elevated basal cell adhesion but displayed no further increase under pressure. These results suggest pressure-induced cell adhesion is mediated by beta1-integrin T788/9 phosphorylation-dependent changes in integrin binding affinity.


Asunto(s)
Adhesión Celular , Neoplasias del Colon/metabolismo , Integrina beta1/metabolismo , Mecanotransducción Celular , Actinina/metabolismo , Animales , Sitios de Unión , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Humanos , Integrina beta1/química , Integrina beta1/genética , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Oligopéptidos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Presión , Conformación Proteica , Proteínas Recombinantes de Fusión/metabolismo , Treonina , Factores de Tiempo , Transfección , Células Tumorales Cultivadas , Familia-src Quinasas/metabolismo
20.
Nat Med ; 14(6): 681-7, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18516051

RESUMEN

Alzheimer's disease is the most common dementia and is pathologically characterized by deposition of amyloid-beta peptide (Abeta) into beta-amyloid plaques, neuronal injury and low-level, chronic activation of brain immunity. Transforming growth factor-betas (TGF-betas) are pleiotropic cytokines that have key roles in immune cell activation, inflammation and repair after injury. We genetically interrupted TGF-beta and downstream Smad2/3 signaling (TGF-beta-Smad2/3) in innate immune cells by inducing expression of CD11c promoter-driven dominant-negative TGF-beta receptor type II in C57BL/6 mice (CD11c-DNR), crossed these mice with mice overexpressing mutant human amyloid precursor protein, the Tg2576 Alzheimer's disease mouse model, and evaluated Alzheimer's disease-like pathology. Aged double-transgenic mice showed complete mitigation of Tg2576-associated hyperactivity and partial mitigation of defective spatial working memory. Brain parenchymal and cerebrovascular beta-amyloid deposits and Abeta abundance were markedly (up to 90%) attenuated in Tg2576-CD11c-DNR mice. This was associated with increased infiltration of Abeta-containing peripheral macrophages around cerebral vessels and beta-amyloid plaques. In vitro, cultures of peripheral macrophages, but not microglia, from CD11c-DNR mice showed blockade of classical TGF-beta-activated Smad2/3 but also showed hyperactivation of alternative bone morphogenic protein-activated Smad1/5/8 signaling and increased Abeta phagocytosis. Similar effects were noted after pharmacological inhibition of activin-like kinase-5, a type I TGF-beta receptor. Taken together, our results suggest that blockade of TGF-beta-Smad2/3 signaling in peripheral macrophages represents a new therapeutic target for Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/patología , Inmunidad Innata , Transducción de Señal , Proteína Smad2/inmunología , Proteína smad3/inmunología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Femenino , Heterocigoto , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Smad2/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda