Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 158
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(14): 3774-3793.e25, 2021 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-34115982

RESUMEN

Cytomegaloviruses (CMVs) have co-evolved with their mammalian hosts for millions of years, leading to remarkable host specificity and high infection prevalence. Macrophages, which already populate barrier tissues in the embryo, are the predominant immune cells at potential CMV entry sites. Here we show that, upon CMV infection, macrophages undergo a morphological, immunophenotypic, and metabolic transformation process with features of stemness, altered migration, enhanced invasiveness, and provision of the cell cycle machinery for viral proliferation. This complex process depends on Wnt signaling and the transcription factor ZEB1. In pulmonary infection, mouse CMV primarily targets and reprograms alveolar macrophages, which alters lung physiology and facilitates primary CMV and secondary bacterial infection by attenuating the inflammatory response. Thus, CMV profoundly perturbs macrophage identity beyond established limits of plasticity and rewires specific differentiation processes, allowing viral spread and impairing innate tissue immunity.


Asunto(s)
Citomegalovirus/fisiología , Macrófagos Alveolares/virología , Animales , Presentación de Antígeno , Efecto Espectador , Ciclo Celular , Línea Celular Transformada , Reprogramación Celular , Citomegalovirus/patogenicidad , Citomegalovirus/ultraestructura , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Proteínas Fluorescentes Verdes/metabolismo , Pulmón/patología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/ultraestructura , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fenotipo , Células Madre/patología , Replicación Viral/fisiología , Vía de Señalización Wnt
2.
Nat Immunol ; 21(1): 30-41, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31819254

RESUMEN

NLRP3-inflammasome-driven inflammation is involved in the pathogenesis of a variety of diseases. Identification of endogenous inflammasome activators is essential for the development of new anti-inflammatory treatment strategies. Here, we identified that apolipoprotein C3 (ApoC3) activates the NLRP3 inflammasome in human monocytes by inducing an alternative NLRP3 inflammasome via caspase-8 and dimerization of Toll-like receptors 2 and 4. Alternative inflammasome activation in human monocytes is mediated by the Toll-like receptor adapter protein SCIMP. This triggers Lyn/Syk-dependent calcium entry and the production of reactive oxygen species, leading to activation of caspase-8. In humanized mouse models, ApoC3 activated human monocytes in vivo to impede endothelial regeneration and promote kidney injury in an NLRP3- and caspase-8-dependent manner. These data provide new insights into the regulation of the NLRP3 inflammasome and the pathophysiological role of triglyceride-rich lipoproteins containing ApoC3. Targeting ApoC3 might prevent organ damage and provide an anti-inflammatory treatment for vascular and kidney diseases.


Asunto(s)
Lesión Renal Aguda/inmunología , Apolipoproteína C-III/inmunología , Caspasa 8/metabolismo , Enfermedades Renales/inmunología , Monocitos/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Lesión Renal Aguda/patología , Proteínas Adaptadoras Transductoras de Señales , Animales , Apolipoproteína C-III/genética , Línea Celular , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Inflamasomas/inmunología , Inflamación/genética , Inflamación/inmunología , Enfermedades Renales/patología , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 4/metabolismo
3.
Nat Immunol ; 20(5): 652-662, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30858620

RESUMEN

αß T cell antigen receptors (TCRs) bind complexes of peptide and major histocompatibility complex (pMHC) with low affinity, which poses a considerable challenge for the direct identification of αß T cell cognate peptides. Here we describe a platform for the discovery of MHC class II epitopes based on the screening of engineered reporter cells expressing novel pMHC-TCR (MCR) hybrid molecules carrying cDNA-derived peptides. This technology identifies natural epitopes of CD4+ T cells in an unbiased and efficient manner and allows detailed analysis of TCR cross-reactivity that provides recognition patterns beyond discrete peptides. We determine the cognate peptides of virus- and tumor-specific T cells in mouse disease models and present a proof of concept for human T cells. Furthermore, we use MCR to identify immunogenic tumor neo-antigens and show that vaccination with a peptide naturally recognized by tumor-infiltrating lymphocytes efficiently protects mice from tumor challenge. Thus, the MCR technology holds promise for basic research and clinical applications, allowing the personalized identification of T cell-specific neo-antigens in patients.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Tumoral , Células Cultivadas , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Humanos , Complejo Mayor de Histocompatibilidad/genética , Ratones Endogámicos C57BL , Péptidos/genética , Péptidos/inmunología , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo
4.
Nat Immunol ; 20(5): 663, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30944415

RESUMEN

In the version of this article initially published, a reference (23) was cited incorrectly and two references were not included in the second sentence of the first paragraph of the second Results subsection ('Screening for gp61 mimotopes with different functional properties'). The correct citation is as follows: "... we replaced the very stable GFP with a slow fluorescent timer (FT)27,28." Full details on the added references can be found in the correction notice. The errors have been corrected in the print, PDF and HTML versions of the paper.

7.
Nat Immunol ; 18(3): 313-320, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28068307

RESUMEN

Notch2 and B cell antigen receptor (BCR) signaling determine whether transitional B cells become marginal zone B (MZB) or follicular B (FoB) cells in the spleen, but it is unknown how these pathways are related. We generated Taok3-/- mice, lacking the serine/threonine kinase Taok3, and found cell-intrinsic defects in the development of MZB but not FoB cells. Type 1 transitional (T1) B cells required Taok3 to rapidly respond to ligation by the Notch ligand Delta-like 1. BCR ligation by endogenous or exogenous ligands induced the surface expression of the metalloproteinase ADAM10 on T1 B cells in a Taok3-dependent manner. T1 B cells expressing surface ADAM10 were committed to becoming MZB cells in vivo, whereas T1 B cells lacking expression of ADAM10 were not. Thus, during positive selection in the spleen, BCR signaling causes immature T1 B cells to become receptive to Notch ligands via Taok3-mediated surface expression of ADAM10.


Asunto(s)
Proteína ADAM10/metabolismo , Inmunidad Adaptativa , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Linfocitos B/fisiología , Diferenciación Celular , Linaje de la Célula , Centro Germinal/inmunología , Proteínas de la Membrana/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Células Cultivadas , Selección Clonal Mediada por Antígenos , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Receptor Notch2/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal
8.
Immunity ; 53(3): 597-613.e6, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32735846

RESUMEN

CD4+ T helper (Th) cells are fundamental players in immunity. Based on the expression of signature cytokines and transcription factors, several Th subsets have been defined. Th cells are thought to be far more heterogeneous and multifunctional than originally believed, but characterization of the full diversity has been hindered by technical limitations. Here, we employ mass cytometry to analyze the diversity of Th cell responses generated in vitro and in animal disease models, revealing a vast heterogeneity of effector states with distinct cytokine footprints. The diversities of cytokine responses established during primary antigen encounters in Th1- and Th2-cell-polarizing conditions are largely maintained after secondary challenge, regardless of the new inflammatory environment, highlighting many of the identified states as stable Th cell sublineages. We also find that Th17 cells tend to upregulate Th2-cell-associated cytokines upon challenge, indicating a closer developmental connection between Th17 and Th2 cells than previously anticipated.


Asunto(s)
Citocinas/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología , Animales , Asma/inmunología , Diferenciación Celular/inmunología , Células Cultivadas , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pyroglyphidae/inmunología , Células TH1/citología , Células Th17/citología , Células Th2/citología
9.
Nature ; 624(7992): 645-652, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38093014

RESUMEN

People with diabetes feature a life-risking susceptibility to respiratory viral infection, including influenza and SARS-CoV-2 (ref. 1), whose mechanism remains unknown. In acquired and genetic mouse models of diabetes, induced with an acute pulmonary viral infection, we demonstrate that hyperglycaemia leads to impaired costimulatory molecule expression, antigen transport and T cell priming in distinct lung dendritic cell (DC) subsets, driving a defective antiviral adaptive immune response, delayed viral clearance and enhanced mortality. Mechanistically, hyperglycaemia induces an altered metabolic DC circuitry characterized by increased glucose-to-acetyl-CoA shunting and downstream histone acetylation, leading to global chromatin alterations. These, in turn, drive impaired expression of key DC effectors including central antigen presentation-related genes. Either glucose-lowering treatment or pharmacological modulation of histone acetylation rescues DC function and antiviral immunity. Collectively, we highlight a hyperglycaemia-driven metabolic-immune axis orchestrating DC dysfunction during pulmonary viral infection and identify metabolic checkpoints that may be therapeutically exploited in mitigating exacerbated disease in infected diabetics.


Asunto(s)
Células Dendríticas , Complicaciones de la Diabetes , Diabetes Mellitus , Susceptibilidad a Enfermedades , Hiperglucemia , Pulmón , Virosis , Animales , Ratones , Acetilcoenzima A/metabolismo , Acetilación , Cromatina/genética , Cromatina/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Complicaciones de la Diabetes/inmunología , Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Glucosa/metabolismo , Histonas/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/inmunología , Hiperglucemia/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/virología , Linfocitos T/inmunología , Virosis/complicaciones , Virosis/inmunología , Virosis/mortalidad , Virus/inmunología , Modelos Animales de Enfermedad , Humanos
10.
Immunity ; 50(3): 645-654.e6, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30770250

RESUMEN

The epidermal growth factor receptor ligand Amphiregulin has a well-documented role in the restoration of tissue homeostasis after injury; however, the mechanism by which Amphiregulin contributes to wound repair remains unknown. Here we show that Amphiregulin functioned by releasing bioactive transforming growth factor beta (TGF-ß) from latent complexes via integrin-αV activation. Using acute injury models in two different tissues, we found that by inducing TGF-ß activation on mesenchymal stromal cells (pericytes), Amphiregulin induced their differentiation into myofibroblasts, thereby selectively contributing to the restoration of vascular barrier function within injured tissue. Furthermore, we identified macrophages as a critical source of Amphiregulin, revealing a direct effector mechanism by which these cells contribute to tissue restoration after acute injury. Combined, these observations expose a so far under-appreciated mechanism of how cells of the immune system selectively control the differentiation of tissue progenitor cells during tissue repair and inflammation.


Asunto(s)
Anfirregulina/metabolismo , Macrófagos/metabolismo , Pericitos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Diferenciación Celular/fisiología , Femenino , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/metabolismo
11.
Nat Methods ; 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39313558

RESUMEN

Transposon (IS200/IS605)-encoded TnpB proteins are predecessors of class 2 type V CRISPR effectors and have emerged as one of the most compact genome editors identified thus far. Here, we optimized the design of Deinococcus radiodurans (ISDra2) TnpB for application in mammalian cells (TnpBmax), leading to an average 4.4-fold improvement in editing. In addition, we developed variants mutated at position K76 that recognize alternative target-adjacent motifs (TAMs), expanding the targeting range of ISDra2 TnpB. We further generated an extensive dataset on TnpBmax editing efficiencies at 10,211 target sites. This enabled us to delineate rules for on-target and off-target editing and to devise a deep learning model, termed TnpB editing efficiency predictor (TEEP; https://www.tnpb.app ), capable of predicting ISDra2 TnpB guiding RNA (ωRNA) activity with high performance (r > 0.8). Employing TEEP, we achieved editing efficiencies up to 75.3% in the murine liver and 65.9% in the murine brain after adeno-associated virus (AAV) vector delivery of TnpBmax. Overall, the set of tools presented in this study facilitates the application of TnpB as an ultracompact programmable endonuclease in research and therapeutics.

12.
Nat Immunol ; 16(1): 36-44, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25521683

RESUMEN

Gas exchange is the vital function of the lungs. It occurs in the alveoli, where oxygen and carbon dioxide diffuse across the alveolar epithelium and the capillary endothelium surrounding the alveoli, separated only by a fused basement membrane 0.2-0.5 µm in thickness. This tenuous barrier is exposed to dangerous or innocuous particles, toxins, allergens and infectious agents inhaled with the air or carried in the blood. The lung immune system has evolved to ward off pathogens and restrain inflammation-mediated damage to maintain gas exchange. Lung-resident macrophages and dendritic cells are located in close proximity to the epithelial surface of the respiratory system and the capillaries to sample and examine the air-borne and blood-borne material. In communication with alveolar epithelial cells, they set the threshold and the quality of the immune response.


Asunto(s)
Células Dendríticas/inmunología , Pulmón/inmunología , Macrófagos Alveolares/inmunología , Alveolos Pulmonares/inmunología , Mucosa Respiratoria/inmunología , Animales , Citocinas/inmunología , Células Dendríticas/citología , Humanos , Pulmón/citología , Macrófagos Alveolares/citología , Alveolos Pulmonares/citología , Mucosa Respiratoria/citología , Infecciones del Sistema Respiratorio/inmunología
13.
Immunity ; 48(6): 1220-1232.e5, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29802020

RESUMEN

Despite the importance of Th17 cells in autoimmune diseases, it remains unclear how they control other inflammatory cells in autoimmune tissue damage. Using a model of spontaneous autoimmune arthritis, we showed that arthritogenic Th17 cells stimulated fibroblast-like synoviocytes via interleukin-17 (IL-17) to secrete the cytokine GM-CSF and also expanded synovial-resident innate lymphoid cells (ILCs) in inflamed joints. Activated synovial ILCs, which expressed CD25, IL-33Ra, and TLR9, produced abundant GM-CSF upon stimulation by IL-2, IL-33, or CpG DNA. Loss of GM-CSF production by either ILCs or radio-resistant stromal cells prevented Th17 cell-mediated arthritis. GM-CSF production by Th17 cells augmented chronic inflammation but was dispensable for the initiation of arthritis. We showed that GM-CSF-producing ILCs were present in inflamed joints of rheumatoid arthritis patients. Thus, a cellular cascade of autoimmune Th17 cells, ILCs, and stromal cells, via IL-17 and GM-CSF, mediates chronic joint inflammation and can be a target for therapeutic intervention.


Asunto(s)
Artritis Reumatoide/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Linfocitos/inmunología , Células del Estroma/inmunología , Células Th17/inmunología , Animales , Artritis Reumatoide/metabolismo , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Linfocitos/metabolismo , Ratones , Células del Estroma/metabolismo , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Células Th17/metabolismo
14.
Nat Immunol ; 15(11): 1026-37, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25263125

RESUMEN

Tissue-resident macrophages constitute heterogeneous populations with unique functions and distinct gene-expression signatures. While it has been established that they originate mostly from embryonic progenitor cells, the signals that induce a characteristic tissue-specific differentiation program remain unknown. We found that the nuclear receptor PPAR-γ determined the perinatal differentiation and identity of alveolar macrophages (AMs). In contrast, PPAR-γ was dispensable for the development of macrophages located in the peritoneum, liver, brain, heart, kidneys, intestine and fat. Transcriptome analysis of the precursors of AMs from newborn mice showed that PPAR-γ conferred a unique signature, including several transcription factors and genes associated with the differentiation and function of AMs. Expression of PPAR-γ in fetal lung monocytes was dependent on the cytokine GM-CSF. Therefore, GM-CSF has a lung-specific role in the perinatal development of AMs through the induction of PPAR-γ in fetal monocytes.


Asunto(s)
Diferenciación Celular/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Macrófagos Alveolares/citología , Monocitos/citología , PPAR gamma/biosíntesis , Animales , Antígeno CD11c/genética , Antígeno CD11c/inmunología , Diferenciación Celular/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Pulmón/citología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , PPAR gamma/genética
15.
Nat Immunol ; 19(4): 320-322, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29563630
16.
Nat Immunol ; 14(10): 1045-53, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23995233

RESUMEN

Chronic inflammation is a fundamental aspect of metabolic disorders such as obesity, diabetes and cardiovascular disease. Cholesterol crystals are metabolic signals that trigger sterile inflammation in atherosclerosis, presumably by activating inflammasomes for IL-1ß production. We found here that atherogenesis was mediated by IL-1α and we identified fatty acids as potent inducers of IL-1α-driven vascular inflammation. Fatty acids selectively stimulated the release of IL-1α but not of IL-1ß by uncoupling mitochondrial respiration. Fatty acid-induced mitochondrial uncoupling abrogated IL-1ß secretion, which deviated the cholesterol crystal-elicited response toward selective production of IL-1α. Our findings delineate a previously unknown pathway for vascular immunopathology that links the cellular response to metabolic stress with innate inflammation, and suggest that IL-1α, not IL-1ß, should be targeted in patients with cardiovascular disease.


Asunto(s)
Aterosclerosis/metabolismo , Ácidos Grasos/metabolismo , Inflamasomas/metabolismo , Interleucina-1alfa/metabolismo , Mitocondrias/metabolismo , Vasculitis/metabolismo , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Señalización del Calcio , Grasas de la Dieta/metabolismo , Ácidos Grasos/farmacología , Femenino , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Canales Iónicos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Ácido Oléico/farmacología , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Proteína Desacopladora 2 , Vasculitis/patología
17.
EMBO J ; 39(3): e103205, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31894879

RESUMEN

Tissue-resident macrophages (MΦTR ) originate from at least two distinct waves of erythro-myeloid progenitors (EMP) arising in the yolk sac (YS) at E7.5 and E8.5 with the latter going through a liver monocyte intermediate. The relative potential of these precursors in determining development and functional capacity of MΦTR remains unclear. Here, we studied development of alveolar macrophages (AM) after single and competitive transplantation of different precursors from YS, fetal liver, and fetal lung into neonatal Csf2ra-/- mice, which lack endogenous AM. Fetal monocytes, promoted by Myb, outcompeted primitive MΦ (pMΦ) in empty AM niches and preferentially developed to mature AM, which is associated with enhanced mitochondrial respiratory and glycolytic capacity and repression of the transcription factors c-Maf and MafB. Interestingly, AM derived from pMΦ failed to efficiently clear alveolar proteinosis and protect from fatal lung failure following influenza virus infection. Thus, our data demonstrate superior developmental and functional capacity of fetal monocytes over pMΦ in AM development and underlying mechanisms explaining replacement of pMΦ in fetal tissues.


Asunto(s)
Hígado/embriología , Pulmón/embriología , Monocitos/citología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Saco Vitelino/embriología , Animales , Células Cultivadas , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Glucólisis , Hígado/citología , Hígado/metabolismo , Pulmón/citología , Pulmón/metabolismo , Macrófagos Alveolares , Factor de Transcripción MafB/metabolismo , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Proteínas Proto-Oncogénicas c-myb/farmacología , Saco Vitelino/citología , Saco Vitelino/metabolismo
18.
Eur J Immunol ; 53(1): e2249948, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36285367

RESUMEN

The thioredoxin (TRX) system is an important contributor to cellular redox balance and regulates cell growth, apoptosis, gene expression, and antioxidant defense in nearly all living cells. Oxidative stress, the imbalance between reactive oxygen species (ROS) and antioxidants, can lead to cell death and tissue damage, thereby contributing to aging and to the development of several diseases, including cardiovascular and allergic diseases, diabetes, and neurological disorders. Targeting its activity is also considered as a promising strategy in the treatment of cancer. Over the past years, immunologists have established an essential function of TRX for activation, proliferation, and responses in T cells, B cells, and macrophages. Upon activation, immune cells rearrange their redox system and activate the TRX pathway to promote proliferation through sustainment of nucleotide biosynthesis, and to support inflammatory responses in myeloid cells by allowing NF-κB and NLRP3 inflammasome responses. Consequently, targeting the TRX system may therapeutically be exploited to inhibit immune responses in inflammatory conditions. In this review, we summarize recent insights revealing key roles of the TRX pathway in immune cells in health and disease, and lessons learnt for cancer therapy.


Asunto(s)
Neoplasias , Estrés Oxidativo , Humanos , Antioxidantes/metabolismo , Oxidación-Reducción , Tiorredoxinas/metabolismo
19.
Trends Immunol ; 42(6): 495-507, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33972166

RESUMEN

Tissue-resident macrophages (MTR) have recently emerged as a key rheostat capable of regulating the balance between organ health and disease. In most organs, ontogenetically and functionally distinct macrophage subsets fulfill a plethora of functions specific to their tissue environment. In this review, we summarize recent findings regarding the ontogeny and functions of macrophage populations in different mammalian tissues, describing how these cells regulate tissue homeostasis and how they can contribute to inflammation. Furthermore, we highlight new developments concerning certain general principles of tissue macrophage biology, including the importance of metabolism for understanding macrophage activation states and the influence of intrinsic and extrinsic factors on macrophage metabolic control. We also shed light on certain open questions in the field and how answering these might pave the way for tissue-specific therapeutic approaches.


Asunto(s)
Activación de Macrófagos , Macrófagos , Animales , Homeostasis , Inflamación
20.
Immunity ; 42(4): 597-9, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25902476

RESUMEN

In the present issue of Immunity, Hoeffel et al. (2015) reconcile a controversy by demonstrating that a distinct wave of yolk-sac-derived erythro-myeloid progenitors (EMPs) differentiate to fetal monocytes in the liver and further to adult macrophages in the majority of tissues.


Asunto(s)
Envejecimiento/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Células Progenitoras Mieloides/inmunología , Proteínas Proto-Oncogénicas c-myb/inmunología , Animales , Femenino , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA