Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(6): 114333, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38865244

RESUMEN

Histone methyltransferases (HMTs) are crucial in gene regulation and function, yet their role in natural killer (NK) cell biology within the tumor microenvironment (TME) remains largely unknown. We demonstrate that the HMT DOT1L limits NK cell conversion to CD49a+ CD49b+ intILC1, a subset that can be observed in the TME in response to stimulation with transforming growth factor (TGF)-ß and is correlated with impaired tumor control. Deleting Dot1l in NKp46-expressing cells reveals its pivotal role in maintaining NK cell phenotype and function. Loss of DOT1L skews NK cells toward intILC1s even in the absence of TGF-ß. Transcriptionally, DOT1L-null NK cells closely resemble intILC1s and ILC1s, correlating with altered NK cell responses and impaired solid tumor control. These findings deepen our understanding of NK cell biology and could inform approaches to prevent NK cell conversion to intILC1s in adoptive NK cell therapies for cancer.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Células Asesinas Naturales , Neoplasias , Animales , Humanos , Ratones , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones Endogámicos C57BL , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Fenotipo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/inmunología
2.
Kidney Int ; 105(4): 731-743, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38158181

RESUMEN

Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a leading cause of kidney failure and is associated with substantial morbidity and mortality. Interstitial inflammation is attributed to the action of infiltrating macrophages and is a feature thought to aggravate disease progression. Here, we investigated the therapeutic potential of the anti-inflammatory IL37b cytokine as a treatment for ADPKD using genetic mouse models, demonstrating that transgenic expression of human IL37b reduced collecting duct cyst burden in both early and adult-onset ADPKD rodent models. Moreover, injection of recombinant human IL37b could also reduce cyst burden in early onset ADPKD mice, an observation not associated with increased macrophage number at early stages of cyst formation. Interestingly, transgenic IL37b expression also did not alter macrophage numbers in advanced disease. Whole kidney RNA-seq highlighted an IL37b-mediated upregulation of the interferon signaling pathway and single-cell RNA-seq established that these changes originate at least partly from kidney resident macrophages. We further found that blocking type I interferon signaling in mice expressing IL37b resulted in increased cyst number, confirming this as an important pathway by which IL37b exerts its beneficial effects. Thus, our studies show that IL37b promotes interferon signaling in kidney resident macrophages which suppresses cyst initiation, identifying this protein as a potential therapy for ADPKD.


Asunto(s)
Quistes , Riñón Poliquístico Autosómico Dominante , Ratones , Humanos , Animales , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Inflamación/genética , Inflamación/complicaciones , Riñón/metabolismo , Quistes/complicaciones , Interleucinas , Interferones
3.
Mucosal Immunol ; 16(5): 642-657, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37392971

RESUMEN

T follicular helper (Tfh) cells are an important component of germinal center (GC)-mediated humoral immunity. Yet, how a chronic type 1 versus protective type 2 helminth infection modulates Tfh-GC responses remains poorly understood. Here, we employ the helminth Trichuris muris model and demonstrate that Tfh cell phenotypes and GC are differentially regulated in acute versus chronic infection. The latter failed to induce Tfh-GC B cell responses, with Tfh cells expressing Τ-bet and interferon-γ. In contrast, interleukin-4-producing Tfh cells dominate responses to an acute, resolving infection. Heightened expression and increased chromatin accessibility of T helper (Th)1- and Th2 cell-associated genes are observed in chronic and acute induced Tfh cells, respectively. Blockade of the Th1 cell response by T-cell-intrinsic T-bet deletion promoted Tfh cell expansion during chronic infection, pointing to a correlation between a robust Tfh cell response and protective immunity to parasites. Finally, blockade of Tfh-GC interactions impaired type 2 immunity, revealing the critical protective role of GC-dependent Th2-like Tfh cell responses during acute infection. Collectively, these results provide new insights into the protective roles of Tfh-GC responses and identify distinct transcriptional and epigenetic features of Tfh cells that emerge during resolving or chronic T. muris infection.

4.
J Immunol ; 210(3): 297-309, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36524995

RESUMEN

CD8 virtual memory T (TVM) cells are Ag-naive CD8 T cells that have undergone partial differentiation in response to common γ-chain cytokines, particularly IL-15 and IL-4. TVM cells from young individuals are highly proliferative in response to TCR and cytokine stimulation but, with age, they lose TCR-mediated proliferative capacity and exhibit hallmarks of senescence. Helminth infection can drive an increase in TVM cells, which is associated with improved pathogen clearance during subsequent infectious challenge in young mice. Given the cytokine-dependent profile of TVM cells and their age-associated dysfunction, we traced proliferative and functional changes in TVM cells, compared with true naive CD8 T cells, after helminth infection of young and aged C57BL/6 mice. We show that IL-15 is essential for the helminth-induced increase in TVM cells, which is driven only by proliferation of existing TVM cells, with negligible contribution from true naive cell differentiation. Additionally, TVM cells showed the greatest proliferation in response to helminth infection and IL-15 compared with other CD8 T cells. Furthermore, TVM cells from aged mice did not undergo expansion after helminth infection due to both TVM cell-intrinsic and -extrinsic changes associated with aging.


Asunto(s)
Helmintiasis , Interleucina-15 , Animales , Ratones , Envejecimiento/inmunología , Linfocitos T CD8-positivos/parasitología , Citocinas , Helmintiasis/inmunología , Helmintiasis/metabolismo , Helmintos/patogenicidad , Memoria Inmunológica , Interleucina-15/metabolismo , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T
6.
Front Immunol ; 12: 667922, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34194431

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are emerging as important cellular regulators of homeostatic and disease-associated immune processes. The cytokine interleukin-33 (IL-33) promotes ILC2-dependent inflammation and immunity, with IL-33 having been shown to activate NF-κB in a wide variety of cell types. However, it is currently unclear which NF-κB members play an important role in IL-33-dependent ILC2 biology. Here, we identify the NF-κB family member c-Rel as a critical component of the IL-33-dependent activation of ILC2s. Although c-Rel is dispensable for ILC2 development, it is critical for ILC2 function in the lung, with c-Rel-deficient (c-Rel-/- ) mice present a significantly reduced response to papain- and IL-33-induced lung inflammation. We also show that the absence of c-Rel reduces the IL-33-dependent expansion of ILC2 precursors and lower levels of IL-5 and IL-13 cytokine production by mature ILC2s in the lung. Together, these results identify the IL-33-c-Rel axis as a central control point of ILC2 activation and function.


Asunto(s)
Inmunidad Innata/efectos de los fármacos , Interleucina-33/farmacología , Pulmón/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Neumonía/metabolismo , Proteínas Proto-Oncogénicas c-rel/metabolismo , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Médula Ósea/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Interleucina-13/metabolismo , Interleucina-5/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Linfocitos/inmunología , Linfocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Subunidad p50 de NF-kappa B/genética , Subunidad p50 de NF-kappa B/metabolismo , Papaína , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/inmunología , Proteínas Proto-Oncogénicas c-rel/genética
7.
Eur J Immunol ; 51(8): 2006-2026, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33960413

RESUMEN

The NF-κB transcription factor c-Rel is a critical regulator of Treg ontogeny, controlling multiple points of the stepwise developmental pathway. Here, we found that the thymic Treg defect in c-Rel-deficient (cRel-/- ) mice is quantitative, not qualitative, based on analyses of TCR repertoire and TCR signaling strength. However, these parameters are altered in the thymic Treg-precursor population, which is also markedly diminished in cRel-/- mice. Moreover, c-Rel governs the transcriptional programme of both thymic and peripheral Tregs, controlling a core of genes involved with immune signaling, and separately in the periphery, cell cycle progression. Last, the immune suppressive function of peripheral cRel-/- tTregs is diminished in a lymphopenic model of T cell proliferation and is associated with decreased stability of Foxp3 expression. Collectively, we show that c-Rel is a transcriptional regulator that controls multiple aspects of Treg development, differentiation, and function via distinct mechanisms.


Asunto(s)
Proteínas Proto-Oncogénicas c-rel/inmunología , Proteínas Proto-Oncogénicas c-rel/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Diferenciación Celular/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Timo/inmunología , Timo/metabolismo
8.
PLoS Negl Trop Dis ; 15(5): e0009340, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33983946

RESUMEN

The current approaches to reduce the burden of chronic helminth infections in endemic areas are adequate sanitation and periodic administration of deworming drugs. Yet, resistance against some deworming drugs and reinfection can still rapidly occur even after treatment. A vaccine against helminths would be an effective solution at preventing reinfection. However, vaccines against helminth parasites have yet to be successfully developed. While T helper cells and innate lymphoid cells have been established as important components of the protective type 2 response, the roles of B cells and antibodies remain the most controversial. Here, we review the roles of B cells during intestinal helminth infection. We discuss the potential factors that contribute to the context-specific roles for B cells in protection against diverse intestinal helminth parasite species, using evidence from well-defined murine model systems. Understanding the precise roles of B cells during resistance and susceptibility to helminth infection may offer a new perspective of type 2 protective immunity.


Asunto(s)
Anticuerpos Antihelmínticos/inmunología , Linfocitos B/inmunología , Helmintiasis/inmunología , Helmintos/inmunología , Parasitosis Intestinales/inmunología , Animales , Antihelmínticos/uso terapéutico , Centro Germinal/inmunología , Helmintiasis/tratamiento farmacológico , Helmintiasis/parasitología , Helmintos/efectos de los fármacos , Humanos , Parasitosis Intestinales/tratamiento farmacológico , Parasitosis Intestinales/parasitología , Ratones , Vacunas Antiprotozoos/inmunología , Reinfección/parasitología , Reinfección/prevención & control , Linfocitos T Colaboradores-Inductores/inmunología
9.
PLoS Pathog ; 17(3): e1009476, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33788902

RESUMEN

Infectious and inflammatory diseases in the intestine remain a serious threat for patients world-wide. Reprogramming of the intestinal epithelium towards a protective effector state is important to manage inflammation and immunity and can be therapeutically targeted. The role of epigenetic regulatory enzymes within these processes is not yet defined. Here, we use a mouse model that has an intestinal-epithelial specific deletion of the histone demethylase Lsd1 (cKO mice), which maintains the epithelium in a fixed reparative state. Challenge of cKO mice with bacteria-induced colitis or a helminth infection model both resulted in increased pathogenesis. Mechanistically, we discovered that LSD1 is important for goblet cell maturation and goblet-cell effector molecules such as RELMß. We propose that this may be in part mediated by directly controlling genes that facilitate cytoskeletal organization, which is important in goblet cell biology. This study therefore identifies intestinal-epithelial epigenetic regulation by LSD1 as a critical element in host protection from infection.


Asunto(s)
Infecciones por Enterobacteriaceae/inmunología , Células Caliciformes/inmunología , Histona Demetilasas/inmunología , Mucosa Intestinal/metabolismo , Tricuriasis/inmunología , Animales , Citrobacter rodentium , Células Caliciformes/metabolismo , Histona Demetilasas/metabolismo , Mucosa Intestinal/inmunología , Ratones , Ratones Noqueados , Trichuris
10.
Cell Rep ; 33(11): 108505, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326781

RESUMEN

CD4+ T helper (Th) cell differentiation is controlled by lineage-specific expression of transcription factors and effector proteins, as well as silencing of lineage-promiscuous genes. Lysine methyltransferases (KMTs) comprise a major class of epigenetic enzymes that are emerging as important regulators of Th cell biology. Here, we show that the KMT DOT1L regulates Th cell function and lineage integrity. DOT1L-dependent dimethylation of lysine 79 of histone H3 (H3K79me2) is associated with lineage-specific gene expression. However, DOT1L-deficient Th cells overproduce IFN-γ under lineage-specific and lineage-promiscuous conditions. Consistent with the increased IFN-γ response, mice with a T-cell-specific deletion of DOT1L are susceptible to infection with the helminth parasite Trichuris muris and are resistant to the development of allergic lung inflammation. These results identify a central role for DOT1L in Th2 cell lineage commitment and stability and suggest that inhibition of DOT1L may provide a therapeutic strategy to limit type 2 immune responses.


Asunto(s)
Antígenos CD4/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Infecciones/inmunología , Inflamación/inmunología , Metiltransferasas/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones
11.
Cell Rep ; 33(11): 108504, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326791

RESUMEN

Histone modifiers are essential for the ability of immune cells to reprogram their gene expression during differentiation. The recruitment of the histone methyltransferase DOT1L (disruptor of telomeric silencing 1-like) induces oncogenic gene expression in a subset of B cell leukemias. Despite its importance, its role in the humoral immune system is unclear. Here, we demonstrate that DOT1L is a critical regulator of B cell biology. B cell development is defective in Dot1lf/fMb1Cre/+ mice, culminating in a reduction of peripheral mature B cells. Upon immunization or influenza infection of Dot1lf/fCd23Cre/+ mice, class-switched antibody-secreting cells are significantly attenuated and germinal centers fail to form. Consequently, DOT1L is essential for B cell memory formation. Transcriptome, pathway, and histological analyses identified a role for DOT1L in reprogramming gene expression for appropriate localization of B cells during the initial stage of the response. Together, these results demonstrate an essential role for DOT1L in generating an effective humoral immune response.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/inmunología , Inmunidad Humoral/inmunología , Metiltransferasas/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones
12.
Nat Commun ; 10(1): 19, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30604761

RESUMEN

Protein methyltransferases (PMTs) comprise a major class of epigenetic regulatory enzymes with therapeutic relevance. Here we present a collection of chemical probes and associated reagents and data to elucidate the function of human and murine PMTs in cellular studies. Our collection provides inhibitors and antagonists that together modulate most of the key regulatory methylation marks on histones H3 and H4, providing an important resource for modulating cellular epigenomes. We describe a comprehensive and comparative characterization of the probe collection with respect to their potency, selectivity, and mode of inhibition. We demonstrate the utility of this collection in CD4+ T cell differentiation assays revealing the potential of individual probes to alter multiple T cell subpopulations which may have implications for T cell-mediated processes such as inflammation and immuno-oncology. In particular, we demonstrate a role for DOT1L in limiting Th1 cell differentiation and maintaining lineage integrity. This chemical probe collection and associated data form a resource for the study of methylation-mediated signaling in epigenetics, inflammation and beyond.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Histonas/metabolismo , Proteína Metiltransferasas/antagonistas & inhibidores , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Pruebas de Enzimas/métodos , Epigenómica/métodos , Células HEK293 , N-Metiltransferasa de Histona-Lisina , Humanos , Células Jurkat , Metilación/efectos de los fármacos , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/metabolismo , Ratones Endogámicos C57BL , Proteína Metiltransferasas/metabolismo , Procesamiento Proteico-Postraduccional/genética , Células TH1/efectos de los fármacos , Células TH1/fisiología
13.
PLoS One ; 13(12): e0208343, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30521606

RESUMEN

Successful T-dependent humoral responses require the production of antibody-secreting plasmablasts, as well as the formation of germinal centers which eventually form high-affinity B cell memory. The ability of B cells to differentiate into germinal center and plasma cells, as well as the ability to tailor responses to different pathogens, is driven by transcription factors. In T cells, the T-box transcription factors T-bet and Eomesodermin (Eomes) regulate effector and memory T cell differentiation, respectively. While T-bet has a critical role in regulating anti-viral B cell responses, a role for Eomes in B cells has yet to be described. We therefore investigated whether Eomes was required for B cell differentiation during either Th1 or Th2 cell-biased immune responses. Here, we demonstrate that deletion of Eomes specifically in B cells did not affect B cell differentiation in response to vaccination, as well as following viral or helminth infection. In contrast to its established role in CD8+ T cells, Eomes did not influence memory B cell differentiation. Finally, the use of an Eomes reporter mouse confirmed the lack of Eomes expression during immune responses. Thus, germinal center and plasma cell differentiation and the formation of isotype-switched memory B cells in response to infection are independent of Eomes expression.


Asunto(s)
Linfocitos B/citología , Linfocitos B/metabolismo , Proteínas de Dominio T Box/metabolismo , Células TH1/metabolismo , Células Th2/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Activación de Linfocitos , Ratones , Proteínas de Dominio T Box/genética
14.
PLoS Pathog ; 14(2): e1006869, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29470558

RESUMEN

The intestinal immune system must be able to respond to a wide variety of infectious organisms while maintaining tolerance to non-pathogenic microbes and food antigens. The Vitamin A metabolite all-trans-retinoic acid (atRA) has been implicated in the regulation of this balance, partially by regulating innate lymphoid cell (ILC) responses in the intestine. However, the molecular mechanisms of atRA-dependent intestinal immunity and homeostasis remain elusive. Here we define a role for the transcriptional repressor Hypermethylated in cancer 1 (HIC1, ZBTB29) in the regulation of ILC responses in the intestine. Intestinal ILCs express HIC1 in a vitamin A-dependent manner. In the absence of HIC1, group 3 ILCs (ILC3s) that produce IL-22 are lost, resulting in increased susceptibility to infection with the bacterial pathogen Citrobacter rodentium. Thus, atRA-dependent expression of HIC1 in ILC3s regulates intestinal homeostasis and protective immunity.


Asunto(s)
Inmunidad Innata , Intestinos/efectos de los fármacos , Intestinos/inmunología , Factores de Transcripción de Tipo Kruppel/fisiología , Linfocitos/efectos de los fármacos , Tretinoina/farmacología , Animales , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Homeostasis/genética , Homeostasis/inmunología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Intestinos/microbiología , Factores de Transcripción de Tipo Kruppel/genética , Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tretinoina/metabolismo
15.
Cell Stem Cell ; 22(2): 177-190.e7, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29395054

RESUMEN

The development of cell therapy for repairing damaged or diseased skeletal muscle has been hindered by the inability to significantly expand immature, transplantable myogenic stem cells (MuSCs) in culture. To overcome this limitation, a deeper understanding of the mechanisms regulating the transition between activated, proliferating MuSCs and differentiation-primed, poorly engrafting progenitors is needed. Here, we show that methyltransferase Setd7 facilitates such transition by regulating the nuclear accumulation of ß-catenin in proliferating MuSCs. Genetic or pharmacological inhibition of Setd7 promotes in vitro expansion of MuSCs and increases the yield of primary myogenic cell cultures. Upon transplantation, both mouse and human MuSCs expanded with a Setd7 small-molecule inhibitor are better able to repopulate the satellite cell niche, and treated mouse MuSCs show enhanced therapeutic potential in preclinical models of muscular dystrophy. Thus, Setd7 inhibition may help bypass a key obstacle in the translation of cell therapy for muscle disease.


Asunto(s)
Desarrollo de Músculos , Proteína Metiltransferasas/antagonistas & inhibidores , Trasplante de Células Madre , Células Madre/citología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Linaje de la Célula/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Células Cultivadas , Eliminación de Gen , N-Metiltransferasa de Histona-Lisina , Ratones , Músculo Esquelético/fisiología , Proteína MioD/metabolismo , Unión Proteica/efectos de los fármacos , Proteína Metiltransferasas/metabolismo , Pirrolidinas/farmacología , Regeneración/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Sulfonamidas/farmacología , Tetrahidroisoquinolinas/farmacología , beta Catenina/metabolismo
16.
Am J Respir Cell Mol Biol ; 57(6): 651-661, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28683207

RESUMEN

Survival during lung injury requires a coordinated program of damage limitation and rapid repair. CD34 is a cell surface sialomucin expressed by epithelial, vascular, and stromal cells that promotes cell adhesion, coordinates inflammatory cell recruitment, and drives angiogenesis. To test whether CD34 also orchestrates pulmonary damage and repair, we induced acute lung injury in wild-type (WT) and Cd34-/- mice by bleomycin administration. We found that Cd34-/- mice displayed severe weight loss and early mortality compared with WT controls. Despite equivalent early airway inflammation to WT mice, CD34-deficient animals developed interstitial edema and endothelial delamination, suggesting impaired endothelial function. Chimeric Cd34-/- mice reconstituted with WT hematopoietic cells exhibited early mortality compared with WT mice reconstituted with Cd34-/- cells, supporting an endothelial defect. CD34-deficient mice were also more sensitive to lung damage caused by influenza infection, showing greater weight loss and more extensive pulmonary remodeling. Together, our data suggest that CD34 plays an essential role in maintaining vascular integrity in the lung in response to chemical- and infection-induced tissue damage.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Antígenos CD34/genética , Endotelio Vascular/metabolismo , Lesión Pulmonar/metabolismo , Edema Pulmonar/metabolismo , Animales , Antígenos CD34/metabolismo , Bleomicina/efectos adversos , Bleomicina/farmacología , Endotelio Vascular/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Ratones , Ratones Noqueados , Edema Pulmonar/inducido químicamente , Edema Pulmonar/genética , Edema Pulmonar/patología
17.
Front Immunol ; 8: 429, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28443098

RESUMEN

G9a (KMT1C, EHMT2) is a lysine methyltransferase (KMT) whose primary function is to di-methylate lysine 9 of histone H3 (H3K9me2). G9a-dependent H3K9me2 is associated with gene silencing and acts primarily through the recruitment of H3K9me2-binding proteins that prevent transcriptional activation. Gene repression via G9a-dependent H3K9me2 is critically required in embryonic stem (ES) cells for the development of cellular lineages by repressing expression of pluripotency factors. In the immune system, lymphoid cells such as T cells and innate lymphoid cells (ILCs) can differentiate from a naïve state into one of several effector lineages that require both activating and repressive mechanisms to maintain the correct gene expression program. Furthermore, the long-term immunity to re-infection is mediated by memory T cells, which also require specific gene expression and repression to maintain a quiescent state. In this review, we examine the molecular machinery of G9a-dependent functions, address the role of G9a in lymphoid cell differentiation and function, and identify potential functions of T cells and ILCs that may be controlled by G9a. Together, this review will highlight the dynamic nature of G9a-dependent H3K9me2 in the immune system and shed light on the nature of repressive epigenetic modifications in cellular lineage choice.

18.
Eur J Immunol ; 47(2): 236-239, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28185248

RESUMEN

Mast cells are innate immune cells that respond rapidly to infection in barrier tissues such as the skin and intestinal mucosa. Expulsion of parasitic worms in the gut involves a robust type 2 host response, and an acute mastocytosis is often generated at the site of infection. However, the role of mast cells in resistance to worm infections appears to be parasite specific. Mast cells are also involved in tissue repair, but the long-term contribution of mast cell activation after worm expulsion has not been definitively studied. In this issue of European Journal of Immunology, Sorobetea et al. [Eur. J. Immunol. 2017. 47: 257-268] demonstrate that activated mast cells persist in the large intestinal lamina propria and intraepithelial compartment long after worm expulsion, resulting in continued local and systemic presence of the mast cell protease mast cell protease 1 (MCPt-1) and enhanced intestinal permeability. In this commentary, we discuss these findings in the wider context of mast cell function in health and disease.


Asunto(s)
Amigos , Mastocitosis/inmunología , Quimasas , Humanos , Mucosa Intestinal/inmunología , Intestinos/inmunología , Mastocitos/citología , Permeabilidad
19.
J Leukoc Biol ; 101(4): 893-900, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28034915

RESUMEN

The incidence of inflammatory bowel diseases (IBDs) has steadily increased in recent decades-a phenomenon that cannot be explained by genetic mutations alone. Other factors, including the composition of the intestinal microbiome, are potentially important contributors to the increased occurrence of this group of diseases. Previous reports have shown a correlation between early-life antibiotic (Abx) treatment and an increased incidence of IBD. In this report, we investigated the effects of early-life Abx treatments on the pathogenicity of CD4+ T cells using an experimental T cell transfer model of IBD. Our results show that CD4+ T cells isolated from adult mice that had been treated with Abx during gestation and in early life induced a faster onset of IBD in Rag1-deficient mice compared with CD4+ T cells of untreated mice. Ex vivo functional analyses of IBD-inducing CD4+ T cells did not show significant differences in their immunologic potential ex vivo, despite their in vivo phenotype. However, genome-wide gene-expression analysis revealed that these cells displayed dysregulated expression of genes associated with cell-cycle regulation, metabolism, and cellular stress. Analysis of Abx-treated CD4+ T cell donors showed systemically elevated levels of the stress hormone corticosterone throughout life compared with untreated donors. The cohousing of Abx-treated mice with untreated mice decreased serum corticosterone, and a consequent transfer of the cells from cohoused mice into Rag1-deficient mice restored the onset and severity of disease to that of untreated animals. Thus, our results suggest that early-life Abx treatment results in a stress response with high levels of corticosterone that influences CD4+ T cell function.


Asunto(s)
Antibacterianos/farmacología , Linfocitos T CD4-Positivos/inmunología , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/patología , Animales , Antibacterianos/uso terapéutico , Linfocitos T CD4-Positivos/efectos de los fármacos , Corticosterona/sangre , Citocinas/genética , Citocinas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Genoma , Vivienda para Animales , Inflamación/sangre , Enfermedades Inflamatorias del Intestino/sangre , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/efectos de los fármacos , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/metabolismo
20.
PLoS Pathog ; 12(9): e1005876, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27598373

RESUMEN

The intestine is a common site for a variety of pathogenic infections. Helminth infections continue to be major causes of disease worldwide, and are a significant burden on health care systems. Lysine methyltransferases are part of a family of novel attractive targets for drug discovery. SETD7 is a member of the Suppressor of variegation 3-9-Enhancer of zeste-Trithorax (SET) domain-containing family of lysine methyltransferases, and has been shown to methylate and alter the function of a wide variety of proteins in vitro. A few of these putative methylation targets have been shown to be important in resistance against pathogens. We therefore sought to study the role of SETD7 during parasitic infections. We find that Setd7-/- mice display increased resistance to infection with the helminth Trichuris muris but not Heligmosomoides polygyrus bakeri. Resistance to T. muris relies on an appropriate type 2 immune response that in turn prompts intestinal epithelial cells (IECs) to alter differentiation and proliferation kinetics. Here we show that SETD7 does not affect immune cell responses during infection. Instead, we found that IEC-specific deletion of Setd7 renders mice resistant to T. muris by controlling IEC turnover, an important aspect of anti-helminth immune responses. We further show that SETD7 controls IEC turnover by modulating developmental signaling pathways such as Hippo/YAP and Wnt/ß-Catenin. We show that the Hippo pathway specifically is relevant during T. muris infection as verteporfin (a YAP inhibitor) treated mice became susceptible to T. muris. We conclude that SETD7 plays an important role in IEC biology during infection.


Asunto(s)
Intestinos/inmunología , Proteína Metiltransferasas/metabolismo , Transducción de Señal , Tricuriasis/inmunología , Trichuris/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Proliferación Celular , Citocinas/metabolismo , Resistencia a la Enfermedad , Células Epiteliales/parasitología , Células Epiteliales/fisiología , Eliminación de Gen , N-Metiltransferasa de Histona-Lisina , Humanos , Intestinos/parasitología , Intestinos/fisiología , Ratones , Especificidad de Órganos , Fosfoproteínas/metabolismo , Porfirinas/efectos adversos , Proteína Metiltransferasas/genética , Tricuriasis/parasitología , Tricuriasis/patología , Verteporfina , Proteínas Señalizadoras YAP , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA