Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Nat Immunol ; 25(5): 902-915, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38589618

RESUMEN

Repetitive exposure to antigen in chronic infection and cancer drives T cell exhaustion, limiting adaptive immunity. In contrast, aberrant, sustained T cell responses can persist over decades in human allergic disease. To understand these divergent outcomes, we employed bioinformatic, immunophenotyping and functional approaches with human diseased tissues, identifying an abundant population of type 2 helper T (TH2) cells with co-expression of TCF7 and LEF1, and features of chronic activation. These cells, which we termed TH2-multipotent progenitors (TH2-MPP) could self-renew and differentiate into cytokine-producing effector cells, regulatory T (Treg) cells and follicular helper T (TFH) cells. Single-cell T-cell-receptor lineage tracing confirmed lineage relationships between TH2-MPP, TH2 effectors, Treg cells and TFH cells. TH2-MPP persisted despite in vivo IL-4 receptor blockade, while thymic stromal lymphopoietin (TSLP) drove selective expansion of progenitor cells and rendered them insensitive to glucocorticoid-induced apoptosis in vitro. Together, our data identify TH2-MPP as an aberrant T cell population with the potential to sustain type 2 inflammation and support the paradigm that chronic T cell responses can be coordinated over time by progenitor cells.


Asunto(s)
Factor Nuclear 1-alfa del Hepatocito , Hipersensibilidad , Factor de Unión 1 al Potenciador Linfoide , Células Madre Multipotentes , Factor 1 de Transcripción de Linfocitos T , Células Th2 , Humanos , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factor de Unión 1 al Potenciador Linfoide/genética , Células Th2/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , Hipersensibilidad/inmunología , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Diferenciación Celular , Citocinas/metabolismo , Linfopoyetina del Estroma Tímico , Animales , Células Cultivadas , Ratones
2.
Immunity ; 57(6): 1274-1288.e6, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38821053

RESUMEN

Severe asthma and sinus disease are consequences of type 2 inflammation (T2I), mediated by interleukin (IL)-33 signaling through its membrane-bound receptor, ST2. Soluble (s)ST2 reduces available IL-33 and limits T2I, but little is known about its regulation. We demonstrate that prostaglandin E2 (PGE2) drives production of sST2 to limit features of lung T2I. PGE2-deficient mice display diminished sST2. In humans with severe respiratory T2I, urinary PGE2 metabolites correlate with serum sST2. In mice, PGE2 enhanced sST2 secretion by mast cells (MCs). Mice lacking MCs, ST2 expression by MCs, or E prostanoid (EP)2 receptors by MCs showed reduced sST2 lung concentrations and strong T2I. Recombinant sST2 reduced T2I in mice lacking PGE2 or ST2 expression by MCs back to control levels. PGE2 deficiency also reversed the hyperinflammatory phenotype in mice lacking ST2 expression by MCs. PGE2 thus suppresses T2I through MC-derived sST2, explaining the severe T2I observed in low PGE2 states.


Asunto(s)
Dinoprostona , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Pulmón , Mastocitos , Ratones Noqueados , Animales , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Proteína 1 Similar al Receptor de Interleucina-1/genética , Mastocitos/inmunología , Mastocitos/metabolismo , Dinoprostona/metabolismo , Ratones , Interleucina-33/metabolismo , Humanos , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Asma/inmunología , Asma/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Ratones Endogámicos C57BL , Inflamación/inmunología , Femenino , Masculino , Transducción de Señal , Neumonía/inmunología , Neumonía/metabolismo
3.
J Immunol ; 209(4): 660-664, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35905999

RESUMEN

Immune checkpoint blockade is limited by resistance to treatment, with many patients not achieving durable antitumor responses. Self-renewing (T cell factor 1+ [TCF1+]) CD8+ T cells have recently been implicated in efficacy of anti-programmed cell death protein 1 (anti-PD-1). Mice challenged with syngeneic tumors were treated with anti-PD-1 and/or a reversible inhibitor of PI3K δ, designed to promote T cell self-renewal. Growth of tumors in untreated mice was characterized by waning proportions of TCF1+ T cells, suggesting self-renewing T cells become limiting for successful immunotherapy. Higher proportions of TCF1+ T cells in tumor and blood correlated with better control of tumor growth. Combining anti-PD-1 and inhibitor of PI3K δ conferred superior protection compared with either monotherapy and was associated with higher frequency of TCF1+ T cells in tumor and blood compared with anti-PD-1 alone. These findings reveal predictive importance of self-renewing T cells in anti-tumor immunity and suggest that resistance-directed strategies to enhance T cell self-renewal could potentiate the efficacy of PD-1 blockade.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Animales , Muerte Celular , Inmunoterapia , Ratones , Fosfatidilinositol 3-Quinasas , Factor 1 de Transcripción de Linfocitos T
4.
J Immunol ; 199(2): 581-588, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28626066

RESUMEN

Long noncoding RNAs (lncRNAs) have emerged as critical regulators of inflammation. To further understand the interaction between inflammatory signaling pathways and lncRNAs, we characterized the function of cardiac and apoptosis-related lncRNA (Carlr), an lncRNA expressed in both mouse and human cells of diverse tissues. Carlr expression is increased following NF-κB signaling in macrophages, with concomitant translocation to, and enrichment of, the transcript in the cytoplasm. Knockdown of Carlr results in impaired expression of NF-κB pathway genes and influences the interaction between macrophages and intestinal cells in an inflammatory environment. In human celiac disease patient samples, increased levels of the Carlr transcript were detected in the cytoplasm, alongside elevated expression of NF-κB pathway genes. These findings suggest that increased Carlr expression and/or cytoplasmic localization is required for efficient NF-κB signaling and is associated with the inflamed tissue state observed in human celiac disease.


Asunto(s)
Citoplasma/genética , Regulación de la Expresión Génica , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , Animales , Apoptosis , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Citoplasma/inmunología , Citoplasma/metabolismo , Expresión Génica , Humanos , Inflamación , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , FN-kappa B/inmunología , Fosforilación , Transducción de Señal
5.
Immunol Cell Biol ; 96(8): 863-871, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29570858

RESUMEN

Growth signals drive hematopoietic progenitor cells to proliferate and branch into divergent cell fates, but how unequal outcomes arise from a common progenitor is not fully understood. We used steady-state analysis of in vivo hematopoiesis and Fms-related tyrosine kinase 3 ligand (Flt3L)-induced in vitro differentiation of dendritic cells (DCs) to determine how growth signals regulate lineage bias. We found that Flt3L signaling induced anabolic activation and proliferation of DC progenitors, which was associated with DC differentiation. Perturbation of processes associated with quiescence and catabolism, including AMP-activated protein kinase signaling, fatty acid oxidation, or mitochondrial clearance increased development of cDC2 cells at the expense of cDC1 cells. Conversely, scavenging anabolism-associated reactive oxygen species skewed differentiation toward cDC1 cells. Sibling daughter cells of dividing DC progenitors exhibited unequal expression of the transcription factor interferon regulatory factor 8, which correlated with clonal divergence in FoxO3a signaling and population-level bifurcation of cell fate. We propose that unequal transmission of growth signals during cell division might support fate branches during proliferative expansion of progenitors.


Asunto(s)
Células Dendríticas/fisiología , Ácidos Grasos/metabolismo , Células Madre Hematopoyéticas/fisiología , Factores Reguladores del Interferón/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Hematopoyesis , Factores Reguladores del Interferón/genética , Metabolismo de los Lípidos , Metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
6.
J Virol ; 87(17): 9431-40, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23804637

RESUMEN

Alphaherpesviruses, including pseudorabies virus (PRV), spread directionally within the nervous systems of their mammalian hosts. Three viral membrane proteins are required for efficient anterograde-directed spread of infection in neurons, including Us9 and a heterodimer composed of the glycoproteins gE and gI. We previously demonstrated that the kinesin-3 motor KIF1A mediates anterograde-directed transport of viral particles in axons of cultured peripheral nervous system (PNS) neurons. The PRV Us9 protein copurifies with KIF1A, recruiting the motor to transport vesicles, but at least one unidentified additional viral protein is necessary for this interaction. Here we show that gE/gI are required for efficient anterograde transport of viral particles in axons by mediating the interaction between Us9 and KIF1A. In the absence of gE/gI, viral particles containing green fluorescent protein (GFP)-tagged Us9 are assembled in the cell body but are not sorted efficiently into axons. Importantly, we found that gE/gI are necessary for efficient copurification of KIF1A with Us9, especially at early times after infection. We also constructed a PRV recombinant that expresses a functional gE-GFP fusion protein and used affinity purification coupled with mass spectrometry to identify gE-interacting proteins. Several viral and host proteins were found to associate with gE-GFP. Importantly, both gI and Us9, but not KIF1A, copurified with gE-GFP. We propose that gE/gI are required for efficient KIF1A-mediated anterograde transport of viral particles because they indirectly facilitate or stabilize the interaction between Us9 and KIF1A.


Asunto(s)
Alphaherpesvirinae/fisiología , Herpesvirus Suido 1/fisiología , Cinesinas/fisiología , Lipoproteínas/fisiología , Neuronas/fisiología , Neuronas/virología , Fosfoproteínas/fisiología , Proteínas del Envoltorio Viral/fisiología , Proteínas Virales/fisiología , Alphaherpesvirinae/genética , Alphaherpesvirinae/patogenicidad , Animales , Transporte Axonal/fisiología , Línea Celular , Células Cultivadas , Herpesvirus Suido 1/genética , Herpesvirus Suido 1/patogenicidad , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intracelular , Lipoproteínas/genética , Células PC12 , Fosfoproteínas/genética , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/fisiología , Porcinos , Proteínas del Envoltorio Viral/genética , Proteínas Virales/genética , Virión/fisiología
7.
Cancer Immunol Res ; 11(2): 164-170, 2023 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-36512052

RESUMEN

Treatment with immune checkpoint blockade (ICB) often fails to elicit durable antitumor immunity. Recent studies suggest that ICB does not restore potency to terminally dysfunctional T cells, but instead drives proliferation and differentiation of self-renewing progenitor T cells into fresh, effector-like T cells. Antitumor immunity catalyzed by ICB is characterized by mobilization of antitumor T cells in systemic circulation and tumor. To address whether abundance of self-renewing T cells in blood is associated with immunotherapy response, we used flow cytometry of peripheral blood from a cohort of patients with metastatic non-small cell lung cancer (NSCLC) treated with ICB. At baseline, expression of T-cell factor 1 (TCF1), a marker of self-renewing T cells, was detected at higher frequency in effector-memory (CCR7-) CD8+ T cells from patients who experienced durable clinical benefit compared to those with primary resistance to ICB. On-treatment blood samples from patients benefiting from ICB also exhibited a greater frequency of TCF1+CCR7-CD8+ T cells and higher proportions of TCF1 expression in treatment-expanded PD-1+CCR7-CD8+ T cells. The observed correlation of TCF1 frequency in CCR7-CD8+ T cells and response to ICB suggests that broader examination of self-renewing T-cell abundance in blood will determine its potential as a noninvasive, predictive biomarker of response and resistance to immunotherapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Receptores CCR7 , Linfocitos T CD8-positivos , Inmunoterapia
8.
JCI Insight ; 8(24)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38131378

RESUMEN

Secondary lymphedema occurs in up to 20% of patients after lymphadenectomy performed for the surgical management of tumors involving the breast, prostate, uterus, and skin. Patients develop progressive edema of the affected extremity due to retention of protein-rich lymphatic fluid. Despite compression therapy, patients progress to chronic lymphedema in which noncompressible fibrosis and adipose tissue are deposited within the extremity. The presence of fibrosis led to our hypothesis that rosiglitazone, a PPARγ agonist that inhibits fibrosis, would reduce fibrosis in a mouse model of secondary lymphedema after hind limb lymphadenectomy. In vivo, rosiglitazone reduced fibrosis in the hind limb after lymphadenectomy. Our findings verified that rosiglitazone reestablished the adipogenic features of TGF-ß1-treated mesenchymal cells in vitro. Despite this, rosiglitazone led to a reduction in adipose tissue deposition. Single-cell RNA-Seq data obtained from human tissues and flow cytometric and histological evaluation of mouse tissues demonstrated increased presence of PDGFRα+ cells in lymphedema; human tissue analysis verified these cells have the capacity for adipogenic and fibrogenic differentiation. Upon treatment with rosiglitazone, we noted a reduction in the overall quantity of PDGFRα+ cells and LipidTOX+ cells. Our findings provide a framework for treating secondary lymphedema as a condition of fibrosis and adipose tissue deposition, both of which, paradoxically, can be prevented with a pro-adipogenic agent.


Asunto(s)
Linfedema , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Masculino , Femenino , Humanos , Ratones , Animales , PPAR gamma , Rosiglitazona/farmacología , Rosiglitazona/uso terapéutico , Linfedema/tratamiento farmacológico , Fibrosis
9.
Blood Adv ; 2(14): 1685-1690, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30021780

RESUMEN

Expression of the transcription factor T-cell factor 1 (TCF1) identifies antigen-experienced murine CD8+ T cells that retain potential for lymphoid recirculation and the ability to self-renew while producing more differentiated effector cells. We found that CD8+ T cells in the blood of both healthy and chronically infected humans expressed TCF1 at 3 distinct levels: high (TCF1-hi), intermediate (TCF1-int), and low (TCF1-lo). TCF1-hi cells could be found within both the naive and memory compartments and were characterized by relative quiescence and lack of immediate effector function. A substantial fraction of TCF1-int cells were found among memory cells, and TCF1-int cells exhibited robust immediate effector functions. TCF1-lo cells were most enriched in effector memory cells that expressed the senescence marker CD57. Following reactivation, TCF1-hi cells gave rise to TCF1-lo descendants while self-renewing the TCF1-hi progenitor. By contrast, reactivation of TCF1-lo cells produced more TCF1-lo cells without evidence of de-differentiating into TCF1-hi cells. Flow cytometric analyses of TCF1 expression from patient specimens may become a useful biomarker for adaptive immune function in response to vaccination, infection, autoimmunity, and cancer.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Memoria Inmunológica , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Femenino , Factor Nuclear 1-alfa del Hepatocito/inmunología , Humanos , Masculino
10.
JCI Insight ; 3(22)2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30429372

RESUMEN

Tissue-resident memory T cells (TRMs) accelerate pathogen clearance through rapid and enhanced functional responses in situ. TRMs are prevalent in diverse anatomic sites throughout the human lifespan, yet their phenotypic and functional diversity has not been fully described. Here, we identify subpopulations of human TRMs based on the ability to efflux fluorescent dyes [efflux(+) TRMs] located within mucosal and lymphoid sites with distinct transcriptional profiles, turnover, and functional capacities. Compared with efflux(-) TRMs, efflux(+) TRMs showed transcriptional and phenotypic features of quiescence including reduced turnover, decreased expression of exhaustion markers, and increased proliferative capacity and signaling in response to homeostatic cytokines. Moreover, upon activation, efflux(+) TRMs secreted lower levels of inflammatory cytokines such as IFN-γ and IL-2 and underwent reduced degranulation. Interestingly, analysis of TRM subsets following activation revealed that both efflux(+) and efflux(-) TRMs undergo extensive transcriptional changes following TCR ligation but retain core TRM transcriptional properties including retention markers, suggesting that TRMs carry out effector function in situ. Overall, our results suggest a model for tissue-resident immunity wherein heterogeneous subsets have differential capacities for longevity and effector function.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Colorantes Fluorescentes , Humanos , Tejido Linfoide/citología , Mitocondrias/metabolismo , Modelos Inmunológicos , Fenotipo , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Distribución Tisular , Transcriptoma
11.
Cell Rep ; 22(4): 860-868, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29420173

RESUMEN

Unequal transmission of nutritive signaling during cell division establishes fate disparity between sibling lymphocytes, but how asymmetric signaling becomes organized is not understood. We show that receptor-associated class I phosphatidylinositol 3-kinase (PI3K) signaling activity, indexed by phosphatidylinositol (3,4,5)-trisphosphate (PIP3) staining, is spatially restricted to the microtubule-organizing center and subsequently to one pole of the mitotic spindle in activated T and B lymphocytes. Asymmetric PI3K activity co-localizes with polarization of antigen receptor components implicated in class I PI3K signaling and with facultative glucose transporters whose trafficking is PI3K dependent and whose abundance marks cells destined for differentiation. Perturbation of class I PI3K activity disrupts asymmetry of upstream antigen receptors and downstream glucose transporter traffic. The roles of PI3K signaling in nutrient utilization, proliferation, and gene expression may have converged with the conserved role of PI3K signaling in cellular symmetry breaking to form a logic for regenerative lymphocyte divisions.


Asunto(s)
Linfocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Diferenciación Celular , Humanos , Transducción de Señal
12.
Immunohorizons ; 1(7): 156-161, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28944344

RESUMEN

Anabolic metabolism in lymphocytes promotes plasmablast and cytotoxic T cell differentiation at the expense of self-renewal. Heightened expression and function of the transcription factor IFN regulatory factor 4 (IRF4) accompany enhanced anabolic induction and full commitment to functional differentiation in B cells and CD8+ T cells. In this study, we used a genetic approach to determine whether IRF4 plays an analogous role in Th1 cell induction. Our findings indicate that IRF4 promotes determined Th1 cell differentiation in tandem with anabolic metabolism of CD4+ T cells. IRF4-deficient CD4+ T cells stimulated in vitro exhibit impaired induction of Th1 gene expression and defective silencing of T cell factor 1 expression. IRF4-deficient CD4+ T cells also undergo a shift toward catabolic metabolism, with reduced mammalian target of rapamycin activation, cell size, and nutrient uptake, as well as increased mitochondrial clearance. These findings suggest that the ability to remodel metabolic states can be an essential gateway for altering cell fate.

13.
J Exp Med ; 214(1): 39-47, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27923906

RESUMEN

Upon infection, an activated CD4+ T cell produces terminally differentiated effector cells and renews itself for continued defense. In this study, we show that differentiation and self-renewal arise as opposing outcomes of sibling CD4+ T cells. After influenza challenge, antigen-specific cells underwent several divisions in draining lymph nodes (LN; DLNs) while maintaining expression of TCF1. After four or five divisions, some cells silenced, whereas some cells maintained TCF1 expression. TCF1-silenced cells were T helper 1-like effectors and concentrated in the lungs. Cells from earliest divisions were memory-like and concentrated in nondraining LN. TCF1-expressing cells from later divisions in the DLN could self-renew, clonally yielding a TCF1-silenced daughter cell as well as a sibling cell maintaining TCF1 expression. Some TCF1-expressing cells in DLNs acquired an alternative, follicular helper-like fate. Modeled differentiation experiments in vitro suggested that unequal PI3K/mechanistic target of rapamycin signaling drives intraclonal cell fate heterogeneity. Asymmetric division enables self-renewal to be coupled to production of differentiated CD4+ effector T cells during clonal selection.


Asunto(s)
División Celular Asimétrica/fisiología , Linfocitos T CD4-Positivos/inmunología , Animales , División Celular , Células Cultivadas , Factor Nuclear 1-alfa del Hepatocito/análisis , Factor Nuclear 1-alfa del Hepatocito/genética , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/fisiología , Serina-Treonina Quinasas TOR/fisiología
14.
Science ; 352(6281): 91-5, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27034373

RESUMEN

Recent studies have implicated long noncoding RNAs (lncRNAs) as regulators of many important biological processes. Here we report on the identification and characterization of a lncRNA, lnc13, that harbors a celiac disease-associated haplotype block and represses expression of certain inflammatory genes under homeostatic conditions. Lnc13 regulates gene expression by binding to hnRNPD, a member of a family of ubiquitously expressed heterogeneous nuclear ribonucleoproteins (hnRNPs). Upon stimulation, lnc13 levels are reduced, thereby allowing increased expression of the repressed genes. Lnc13 levels are significantly decreased in small intestinal biopsy samples from patients with celiac disease, which suggests that down-regulation of lnc13 may contribute to the inflammation seen in this disease. Furthermore, the lnc13 disease-associated variant binds hnRNPD less efficiently than its wild-type counterpart, thus helping to explain how these single-nucleotide polymorphisms contribute to celiac disease.


Asunto(s)
Enfermedad Celíaca/genética , Predisposición Genética a la Enfermedad , Inflamación/genética , ARN Largo no Codificante/genética , Animales , Secuencia de Bases , Enfermedad Celíaca/patología , Regulación hacia Abajo , Regulación de la Expresión Génica , Haplotipos , Ribonucleoproteínas Nucleares Heterogéneas/genética , Humanos , Ratones , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple
15.
Cell Rep ; 17(12): 3142-3152, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28009285

RESUMEN

Regeneration requires related cells to diverge in fate. We show that activated lymphocytes yield sibling cells with unequal elimination of aged mitochondria. Disparate mitochondrial clearance impacts cell fate and reflects larger constellations of opposing metabolic states. Differentiation driven by an anabolic constellation of PI3K/mTOR activation, aerobic glycolysis, inhibited autophagy, mitochondrial stasis, and ROS production is balanced with self-renewal maintained by a catabolic constellation of AMPK activation, mitochondrial elimination, oxidative metabolism, and maintenance of FoxO1 activity. Perturbations up and down the metabolic pathways shift the balance of nutritive constellations and cell fate owing to self-reinforcement and reciprocal inhibition between anabolism and catabolism. Cell fate and metabolic state are linked by transcriptional regulators, such as IRF4 and FoxO1, with dual roles in lineage and metabolic choice. Instructing some cells to utilize nutrients for anabolism and differentiation while other cells catabolically self-digest and self-renew may enable growth and repair in metazoa.


Asunto(s)
Proteína Forkhead Box O1/genética , Factores Reguladores del Interferón/genética , Activación de Linfocitos/genética , Linfocitos/metabolismo , Mitocondrias/metabolismo , Animales , Autofagia/genética , Diferenciación Celular/genética , Proteína Forkhead Box O1/metabolismo , Glucólisis , Hematopoyesis/genética , Factores Reguladores del Interferón/metabolismo , Metabolismo/genética , Ratones , Mitocondrias/genética , Fosfatidilinositol 3-Quinasas/genética , Especies Reactivas de Oxígeno/metabolismo , Regeneración/genética , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética
16.
Cell Rep ; 17(7): 1773-1782, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27829149

RESUMEN

Selected CD8+ T cells must divide, produce differentiated effector cells, and self-renew, often repeatedly. We now show that silencing expression of the transcription factor TCF1 marks loss of self-renewal by determined effector cells and that this requires cell division. In acute infections, the first three CD8+ T cell divisions produce daughter cells with unequal proliferative signaling but uniform maintenance of TCF1 expression. The more quiescent initial daughter cells resemble canonical central memory cells. The more proliferative, effector-prone cells from initial divisions can subsequently undergo division-dependent production of a TCF1-negative effector daughter cell along with a self-renewing TCF1-positive daughter cell, the latter also contributing to the memory cell pool upon resolution of infection. Self-renewal in the face of effector cell determination may promote clonal amplification and memory cell formation in acute infections, sustain effector regeneration during persistent subclinical infections, and be rate limiting, but remediable, in chronic active infections and cancer.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Autorrenovación de las Células , Animales , Ciclo Celular , Diferenciación Celular , División Celular , Proliferación Celular , Células Clonales , Silenciador del Gen , Ratones Endogámicos C57BL , Factor 1 de Transcripción de Linfocitos T/metabolismo
17.
Cell Rep ; 13(10): 2203-18, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26628372

RESUMEN

Metazoan sibling cells often diverge in activity and identity, suggesting links between growth signals and cell fate. We show that unequal transduction of nutrient-sensitive PI3K/AKT/mTOR signaling during cell division bifurcates transcriptional networks and fates of kindred cells. A sibling B lymphocyte with stronger signaling, indexed by FoxO1 inactivation and IRF4 induction, undergoes PI3K-driven Pax5 repression and plasma cell determination, while its sibling with weaker PI3K activity renews a memory or germinal center B cell fate. PI3K-driven effector T cell determination silences TCF1 in one sibling cell, while its PI3K-attenuated sibling self-renews in tandem. Prior to bifurcations achieving irreversible plasma or effector cell fate determination, asymmetric signaling during initial divisions specifies a more proliferative, differentiation-prone lymphocyte in tandem with a more quiescent memory cell sibling. By triggering cell division but transmitting unequal intensity between sibling cells, nutrient-sensitive signaling may be a frequent arbiter of cell fate bifurcations during development and repair.


Asunto(s)
Linfocitos T CD8-positivos/citología , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Células Plasmáticas/citología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/metabolismo , Linaje de la Célula , Citometría de Flujo , Técnicas de Sustitución del Gen , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Células Plasmáticas/metabolismo , Transducción de Señal/fisiología
18.
PLoS One ; 8(3): e58776, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23527020

RESUMEN

Alphaherpes viruses, such as pseudorabies virus (PRV), undergo anterograde transport in neuronal axons to facilitate anterograde spread within hosts. Axonal sorting and anterograde transport of virions is dependent on the viral membrane protein Us9, which interacts with the host motor protein Kif1A to direct transport. Us9-Kif1A interactions are necessary but not sufficient for these processes, indicating that additional cofactors or post-translational modifications are needed. In this study, we characterized two conserved serine phosphorylation sites (S51 and S53) in the PRV Us9 protein that are necessary for anterograde spread in vivo. We assessed the subcellular localization of phospho-Us9 subspecies during infection of neurons and found that the phospho-form is detectable on the majority, but not all, of axonal vesicles containing Us9 protein. In biochemical assays, phospho-Us9 was enriched in lipid raft membrane microdomains, though Us9 phosphorylation did not require prior lipid raft association. During infections of chambered neuronal cultures, we observed only a modest reduction in anterograde spread capacity for diserine mutant Us9, and no defect for monoserine mutants. Conversely, mutation of the kinase recognition sequence residues adjacent to the phosphorylation sites completely abrogated anterograde spread. In live-cell imaging analyses, anterograde transport of virions was reduced during infection with a recombinant PRV strain expressing GFP-tagged diserine mutant Us9. Phosphorylation was not required for Us9-Kif1A interaction, suggesting that Us9-Kif1A binding is a distinct step from the activation and/or stabilization of the transport complex. Taken together, our findings indicate that, while not essential, Us9 phosphorylation enhances Us9-Kif1A-based transport of virions in axons to modulate the overall efficiency of long-distance anterograde spread of infection.


Asunto(s)
Herpesvirus Suido 1/metabolismo , Lipoproteínas/química , Lipoproteínas/metabolismo , Neuronas/metabolismo , Neuronas/virología , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Animales , Transporte Axonal , Sitios de Unión , Línea Celular , Células Cultivadas , Herpesvirus Suido 1/genética , Interacciones Huésped-Patógeno , Péptidos y Proteínas de Señalización Intracelular , Cinesinas/metabolismo , Lipoproteínas/genética , Mutagénesis Sitio-Dirigida , Células PC12 , Fosfoproteínas/genética , Fosforilación , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Porcinos , Proteínas Virales/genética
19.
J Vis Exp ; (78)2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23978901

RESUMEN

Advances in live cell fluorescence microscopy techniques, as well as the construction of recombinant viral strains that express fluorescent fusion proteins have enabled real-time visualization of transport and spread of alphaherpes virus infection of neurons. The utility of novel fluorescent fusion proteins to viral membrane, tegument, and capsids, in conjunction with live cell imaging, identified viral particle assemblies undergoing transport within axons. Similar tools have been successfully employed for analyses of cell-cell spread of viral particles to quantify the number and diversity of virions transmitted between cells. Importantly, the techniques of live cell imaging of anterograde transport and spread produce a wealth of information including particle transport velocities, distributions of particles, and temporal analyses of protein localization. Alongside classical viral genetic techniques, these methodologies have provided critical insights into important mechanistic questions. In this article we describe in detail the imaging methods that were developed to answer basic questions of alphaherpes virus transport and spread.


Asunto(s)
Infecciones por Herpesviridae/virología , Microscopía Fluorescente/métodos , Alphaherpesvirinae , Animales , Ratas , Ganglio Cervical Superior/citología , Ganglio Cervical Superior/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA