Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
1.
Blood ; 143(26): 2763-2777, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38603633

RESUMEN

ABSTRACT: Inflammatory responses must be tightly coordinated with the activation of emergency myelopoiesis to produce potent myeloid cells that fight infection without causing excessive host damage. Here, we show that granulocyte-macrophage colony-stimulating factor (GM-CSF) programs myeloid-committed progenitors to produce trained macrophages (increased cytokine response), but programs the upstream noncommitted LKS+ progenitors (defined as Lin- c-Kit+ Sca-1+ cells) to produce tolerized macrophages (decreased cytokine response). In myeloid progenitors, GM-CSF strongly activates signal transducer and activator of transcription 5 (STAT5), Ras-Raf-extracellular signal regulated kinase (ERK), and Akt-mTOR signaling pathways, which are essential to establish a training program, whereas in LKS+ progenitors, GM-CSF induces NF-κB translocation to the nucleus to establish a tolerization program. These differences arise from higher GM-CSF receptor expression in myeloid progenitors compared with LKS+ cells. We demonstrate that ß-catenin regulation of NF-κB nuclear translocation is central in this process. In myeloid progenitors, glycogen synthase kinase 3 (GSK3) inactivation by strong ERK and phosphatidylinositol 3 kinase (PI3K)-Akt signaling increases cytoplasmic ß-catenin levels to block NF-κB nuclear translocation. In contrast, when ERK and PI3K-Akt signaling are weak, active GSK3 causes a decrease in ß-catenin, allowing NF-κB nuclear translocation in LKS+ progenitors. Finally, GM-CSF-induced LKS+ tolerization takes place in several murine models of trained immunity and in human CD34+ CD38- progenitors. Our study reveals that in addition to activating myelopoiesis, GM-CSF also programs early and immediate myeloid progenitors to produce opposing immune memory phenotypes. We propose that the inflammatory response from immediate myeloid progenitors may be balanced by the tolerized phenotype of early progenitors, thus providing a mechanism for appropriate resolution of inflammation and protection against a prolonged cytokine storm.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos , Mielopoyesis , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Animales , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Ratones , Fenotipo , Transducción de Señal , FN-kappa B/metabolismo , Memoria Inmunológica , Ratones Endogámicos C57BL , Macrófagos/metabolismo , Macrófagos/inmunología , Inmunidad Innata , Células Progenitoras Mieloides/metabolismo , Células Progenitoras Mieloides/inmunología , beta Catenina/metabolismo , beta Catenina/genética
2.
Front Immunol ; 12: 765741, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925335

RESUMEN

A link between high sodium chloride (salt) intake and the development of autoimmune diseases was previously reported. These earlier studies demonstrated exacerbation of experimental autoimmune encephalomyelitis and colitis by excess salt intake associated with Th17- and macrophage-mediated mechanisms. Little is known about the impact of dietary salt intake on experimental arthritides. Here, we investigated if salt restriction can exert beneficial effects on collagen-induced arthritis (CIA) and K/BxN serum transfer-induced arthritis (STIA). CIA depends on both adaptive and innate immunity, while STIA predominantly mimics the innate immune cell-driven effector phase of arthritis. In both models, low salt (LS) diet significantly decreased arthritis severity compared to regular salt (RS) and high salt (HS) diet. We did not observe an aggravation of arthritis with HS diet compared to RS diet. Remarkably, in STIA, LS diet was as effective as IL-1 receptor blocking treatment. Complement-fixing anti-CII IgG2a antibodies are associated with inflammatory cell infiltration and cartilage destruction. LS diet reduced anti-CII IgG2a levels in CIA and decreased the anti-CII IgG2a/IgG1 ratios pointing toward a more Th2-like response. Significantly less inflammatory joint infiltrates and cartilage breakdown associated with reduced protein concentrations of IL-1 beta (CIA and STIA), IL-17 (CIA), and the monocyte chemoattractant protein-1 (MCP-1) (CIA) were detected in mice receiving LS diet compared to HS diet. However, we did not find a reduced IL-17A expression in CD4+ T cells upon salt restriction in CIA. Analysis of mRNA transcripts and immunoblots revealed a link between LS diet and inhibition of the p38 MAPK (mitogen-activated protein kinase)/NFAT5 (nuclear factor of activated T-cells 5) signaling axis in STIA. Further experiments indicated a decreased leukodiapedesis under LS conditions. In conclusion, dietary salt restriction ameliorates CIA and STIA, indicating a beneficial role of LS diet during both the immunization and effector phase of immune-mediated arthritides by predominantly modulating the humoral immunity and the activation status of myeloid lineage cells. Hence, salt restriction might represent a supportive dietary intervention not only to reduce cardiovascular risk, but also to improve human inflammatory joint diseases like rheumatoid arthritis.


Asunto(s)
Artritis Experimental , Dieta Hiposódica , Inmunidad Adaptativa , Animales , Artritis Experimental/sangre , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/patología , Linfocitos B/inmunología , Citocinas/genética , Citocinas/inmunología , Selectina E/inmunología , Células Endoteliales/inmunología , Articulaciones del Pie/inmunología , Articulaciones del Pie/patología , Inmunidad Innata , Inmunoglobulina G/sangre , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Monocitos/inmunología , Células Progenitoras Mieloides/inmunología , Receptores de Interleucina-1/inmunología
3.
Cell Rep ; 37(4): 109887, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34706233

RESUMEN

In emergency myelopoiesis (EM), expansion of the myeloid progenitor compartment and increased myeloid cell production are observed and often mediated by the pro-inflammatory cytokine interferon gamma (IFN-γ). Interleukin-10 (IL-10) inhibits IFN-γ secretion, but paradoxically, its therapeutic administration to humans causes hematologic changes similar to those observed in EM. In this work, we use different in vivo systems, including a humanized immune system mouse model, to show that IL-10 triggers EM, with a significant expansion of the myeloid progenitor compartment and production of myeloid cells. Hematopoietic progenitors display a prominent IFN-γ transcriptional signature, and we show that IFN-γ mediates IL-10-driven EM. We also find that IL-10, unexpectedly, reprograms CD4 and CD8 T cells toward an activation state that includes IFN-γ production by these T cell subsets in vivo. Therefore, in addition to its established anti-inflammatory properties, IL-10 can induce IFN-γ production and EM, opening additional perspectives for the design of IL-10-based immunotherapies.


Asunto(s)
Interferón gamma/inmunología , Interleucina-10/inmunología , Células Progenitoras Mieloides/inmunología , Mielopoyesis/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Interferón gamma/genética , Interleucina-10/genética , Ratones , Ratones Noqueados , Mielopoyesis/genética
4.
Clin Sci (Lond) ; 135(15): 1859-1871, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34296277

RESUMEN

BACKGROUND AND AIMS: Preterm birth is associated with increased risk of cardiovascular disease (CVD). This may reflect a legacy of inflammatory exposures such as chorioamnionitis which complicate pregnancies delivering preterm, or recurrent early-life infections, which are common in preterm infants. We previously reported that experimental chorioamnionitis followed by postnatal inflammation has additive and deleterious effects on atherosclerosis in ApoE-/- mice. Here, we aimed to investigate whether innate immune training is a contributory inflammatory mechanism in this murine model of atherosclerosis. METHODS: Bone marrow-derived macrophages and peritoneal macrophages were isolated from 13-week-old ApoE-/- mice, previously exposed to prenatal intra-amniotic (experimental choriomanionitis) and/or repeated postnatal (peritoneal) lipopolysaccharide (LPS). Innate immune responses were assessed by cytokine responses following ex vivo stimulation with toll-like receptor (TLR) agonists (LPS, Pam3Cys) and RPMI for 24-h. Bone marrow progenitor populations were studied using flow cytometric analysis. RESULTS: Following postnatal LPS exposure, bone marrow-derived macrophages and peritoneal macrophages produced more pro-inflammatory cytokines following TLR stimulation than those from saline-treated controls, characteristic of a trained phenotype. Cytokine production ex vivo correlated with atherosclerosis severity in vivo. Prenatal LPS did not affect cytokine production capacity. Combined prenatal and postnatal LPS exposure was associated with a reduction in populations of myeloid progenitor cells in the bone marrow. CONCLUSIONS: Postnatal inflammation results in a trained phenotype in atherosclerosis-prone mice that is not enhanced by prenatal inflammation. If analogous mechanisms occur in humans, then there may be novel early life opportunities to reduce CVD risk in infants with early life infections.


Asunto(s)
Aterosclerosis/inmunología , Corioamnionitis/inmunología , Inmunidad Innata , Macrófagos Peritoneales/inmunología , Células Progenitoras Mieloides/inmunología , Peritonitis/inmunología , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Células Cultivadas , Corioamnionitis/inducido químicamente , Corioamnionitis/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/metabolismo , Lipopolisacáridos , Macrófagos Peritoneales/metabolismo , Ratones Noqueados para ApoE , Células Progenitoras Mieloides/metabolismo , Peritonitis/inducido químicamente , Peritonitis/metabolismo , Fenotipo , Embarazo
5.
Nat Immunol ; 22(5): 595-606, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33903766

RESUMEN

Although the pathological significance of tumor-associated macrophage (TAM) heterogeneity is still poorly understood, TAM reprogramming is viewed as a promising anticancer therapy. Here we show that a distinct subset of TAMs (F4/80hiCD115hiC3aRhiCD88hi), endowed with high rates of heme catabolism by the stress-responsive enzyme heme oxygenase-1 (HO-1), plays a critical role in shaping a prometastatic tumor microenvironment favoring immunosuppression, angiogenesis and epithelial-to-mesenchymal transition. This population originates from F4/80+HO-1+ bone marrow (BM) precursors, accumulates in the blood of tumor bearers and preferentially localizes at the invasive margin through a mechanism dependent on the activation of Nrf2 and coordinated by the NF-κB1-CSF1R-C3aR axis. Inhibition of F4/80+HO-1+ TAM recruitment or myeloid-specific deletion of HO-1 blocks metastasis formation and improves anticancer immunotherapy. Relative expression of HO-1 in peripheral monocyte subsets, as well as in tumor lesions, discriminates survival among metastatic melanoma patients. Overall, these results identify a distinct cancer-induced HO-1+ myeloid subgroup as a new antimetastatic target and prognostic blood marker.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Hemo-Oxigenasa 1/metabolismo , Neoplasias Pulmonares/inmunología , Melanoma/inmunología , Neoplasias Cutáneas/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/sangre , Línea Celular Tumoral/trasplante , Quimioterapia Adyuvante/métodos , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/inmunología , Femenino , Hemo/metabolismo , Hemo-Oxigenasa 1/antagonistas & inhibidores , Hemo-Oxigenasa 1/sangre , Hemo-Oxigenasa 1/genética , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Masculino , Melanoma/mortalidad , Melanoma/secundario , Melanoma/terapia , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Escape del Tumor/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Macrófagos Asociados a Tumores/metabolismo
6.
Elife ; 102021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33830019

RESUMEN

Innate immune cellular effectors are actively consumed during systemic inflammation, but the systemic traffic and the mechanisms that support their replenishment remain unknown. Here, we demonstrate that acute systemic inflammation induces the emergent activation of a previously unrecognized system of rapid migration of granulocyte-macrophage progenitors and committed macrophage-dendritic progenitors, but not other progenitors or stem cells, from bone marrow (BM) to regional lymphatic capillaries. The progenitor traffic to the systemic lymphatic circulation is mediated by Ccl19/Ccr7 and is NF-κB independent, Traf6/IκB-kinase/SNAP23 activation dependent, and is responsible for the secretion of pre-stored Ccl19 by a subpopulation of CD205+/CD172a+ conventional dendritic cells type 2 and upregulation of BM myeloid progenitor Ccr7 signaling. Mature myeloid Traf6 signaling is anti-inflammatory and necessary for lymph node myeloid cell development. This report unveils the existence and the mechanistic basis of a very early direct traffic of myeloid progenitors from BM to lymphatics during inflammation.


When the body becomes infected with disease-causing pathogens, such as bacteria, the immune system activates various mechanisms which help to fight off the infection. One of the immune system's first lines of defense is to launch an inflammatory response that helps remove the pathogen and recruit other immune cells. However, this response can become overactivated, leading to severe inflammatory conditions that damage healthy cells and tissues. A second group of cells counteract this over inflammation and are different to the ones involved in the early inflammatory response. Both types of cells ­ inflammatory and anti-inflammatory ­ develop from committed progenitors, which, unlike stem cells, are already destined to become a certain type of cell. These committed progenitors reside in the bone marrow and then rapidly travel to secondary lymphoid organs, such as the lymph nodes, where they mature into functioning immune cells. During this journey, committed progenitors pass from the bone marrow to the lymphatic vessels that connect up the different secondary lymphoid organs, and then spread to all tissues in the body. Yet, it is not fully understood what exact route these cells take and what guides them towards these lymphatic tissues during inflammation. To investigate this, Serrano-Lopez, Hegde et al. used a combination of techniques to examine the migration of progenitor cells in mice that had been treated with lethal doses of a bacterial product that triggers inflammation. This revealed that as early as one to three hours after the onset of infection, progenitor cells were already starting to travel from the bone marrow towards lymphatic vessels. Serrano-Lopez, Hegde et al. found that a chemical released by an "alarm" immune cell already residing in secondary lymphoid organs attracted these progenitor cells towards the lymphatic tissue. Further experiments showed that the progenitor cells travelling to secondary lymphoid organs were already activated by bacterial products. They then follow the chemical released by alarm immune cells ready to respond to the immune challenge and suppress inflammation. These committed progenitors were also found in the inflamed lymph nodes of patients. These findings suggest this rapid circulation of progenitors is a mechanism of defense that contributes to the fight against severe inflammation. Altering how these cells migrate from the bone marrow to secondary lymphoid organs could provide a more effective treatment for inflammatory conditions and severe infections. However, these approaches would need to be tested further in the laboratory and in clinical trials.


Asunto(s)
Médula Ósea/metabolismo , Movimiento Celular , Células Progenitoras de Granulocitos y Macrófagos/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Linfadenopatía/metabolismo , Sistema Linfático/metabolismo , Células Progenitoras Mieloides/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Médula Ósea/inmunología , Médula Ósea/patología , Linaje de la Célula , Células Cultivadas , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Células Progenitoras de Granulocitos y Macrófagos/inmunología , Células Progenitoras de Granulocitos y Macrófagos/patología , Humanos , Inflamación/inmunología , Inflamación/patología , Linfadenopatía/inmunología , Linfadenopatía/patología , Sistema Linfático/inmunología , Sistema Linfático/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/patología , Fenotipo , Transducción de Señal , Factores de Tiempo , Adulto Joven
8.
Nat Immunol ; 22(3): 301-311, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33603226

RESUMEN

The transcription factor IRF8 is essential for the development of monocytes and dendritic cells (DCs), whereas it inhibits neutrophilic differentiation. It is unclear how Irf8 expression is regulated and how this single transcription factor supports the generation of both monocytes and DCs. Here, we identified a RUNX-CBFß-driven enhancer 56 kb downstream of the Irf8 transcription start site. Deletion of this enhancer in vivo significantly decreased Irf8 expression throughout the myeloid lineage from the progenitor stages, thus resulting in loss of common DC progenitors and overproduction of Ly6C+ monocytes. We demonstrated that high, low or null expression of IRF8 in hematopoietic progenitor cells promotes differentiation toward type 1 conventional DCs, Ly6C+ monocytes or neutrophils, respectively, via epigenetic regulation of distinct sets of enhancers in cooperation with other transcription factors. Our results illustrate the mechanism through which IRF8 controls the lineage choice in a dose-dependent manner within the myeloid cell system.


Asunto(s)
Linaje de la Célula , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Células Dendríticas/metabolismo , Elementos de Facilitación Genéticos , Factores Reguladores del Interferón/metabolismo , Monocitos/metabolismo , Células Progenitoras Mieloides/metabolismo , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Células de la Médula Ósea , Células Cultivadas , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/genética , Células Dendríticas/inmunología , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Factores Reguladores del Interferón/deficiencia , Factores Reguladores del Interferón/genética , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Monocitos/inmunología , Células Progenitoras Mieloides/inmunología , Fenotipo , Transducción de Señal
9.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33635312

RESUMEN

Hematopoietic stem cells reside in the bone marrow, where they generate the effector cells that drive immune responses. However, in response to inflammation, some hematopoietic stem and progenitor cells (HSPCs) are recruited to tissue sites and undergo extramedullary hematopoiesis. Contrasting with this paradigm, here we show residence and differentiation of HSPCs in healthy gingiva, a key oral barrier in the absence of overt inflammation. We initially defined a population of gingiva monocytes that could be locally maintained; we subsequently identified not only monocyte progenitors but also diverse HSPCs within the gingiva that could give rise to multiple myeloid lineages. Gingiva HSPCs possessed similar differentiation potentials, reconstitution capabilities, and heterogeneity to bone marrow HSPCs. However, gingival HSPCs responded differently to inflammatory insults, responding to oral but not systemic inflammation. Combined, we highlight a novel pathway of myeloid cell development at a healthy barrier, defining a gingiva-specific HSPC network that supports generation of a proportion of the innate immune cells that police this barrier.


Asunto(s)
Encía/citología , Encía/inmunología , Células Progenitoras Mieloides/citología , Células Progenitoras Mieloides/inmunología , Animales , Médula Ósea/metabolismo , Femenino , Hematopoyesis , Masculino , Ratones , Ratones Endogámicos C57BL , Mucosa Bucal/citología , Mucosa Bucal/inmunología , RNA-Seq/métodos , Análisis de la Célula Individual/métodos
10.
Stem Cell Reports ; 16(2): 324-336, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33482101

RESUMEN

Hemorrhagic shock induces an aberrant immune response characterized by simultaneous induction of a proinflammatory state and impaired host defenses. The objective of this study was to evaluate the impact of conditionally immortalized neutrophil progenitors (NPs) on this aberrant immune response. We employed a mouse model of hemorrhagic shock, followed by the adoptive transfer of NPs and subsequent inoculation of Staphylococcus aureus to induce pneumonia. We observed that transplant of NPs decreases the proportion of host neutrophils that express programmed death ligand 1 and intercellular adhesion molecule 1 in the context of prior hemorrhage. Following hemorrhage, NP transplant decreased proinflammatory cytokines in the lungs, increased neutrophil migration into the airspaces, and enhanced bacterial clearance. Further, hemorrhagic shock improved NP engraftment in the bone marrow. These results suggest that NPs hold the potential for use as a cellular therapy in the treatment and prevention of secondary infection following hemorrhagic shock.


Asunto(s)
Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Neutrófilos/inmunología , Neumonía/inmunología , Choque Hemorrágico/inmunología , Choque Hemorrágico/metabolismo , Staphylococcus aureus/inmunología , Animales , Antígeno B7-H1/metabolismo , Médula Ósea/inmunología , Línea Celular , Movimiento Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inmunidad , Molécula 1 de Adhesión Intercelular/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Neutrófilos/trasplante , Neumonía/microbiología , Choque Hemorrágico/complicaciones
11.
J Immunol ; 206(2): 432-445, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33310871

RESUMEN

Although neutrophils play important roles in immunity and inflammation, their analysis is strongly hindered by their short-lived and terminally differentiated nature. Prior studies reported conditional immortalization of myeloid progenitors using retroviral expression of an estrogen-dependent fusion protein of the HoxB8 transcription factor. This approach allowed the long-term culture of mouse myeloid progenitors (HoxB8 progenitors) in estrogen-containing media, followed by differentiation toward neutrophils upon estrogen withdrawal. Although several reports confirmed the in vitro functional responsiveness of the resulting differentiated cells (HoxB8 neutrophils), little is known about their capacity to perform in vivo neutrophil functions. We have addressed this issue by an in vivo transplantation approach. In vitro-generated HoxB8 neutrophils showed a neutrophil-like phenotype and were able to perform conventional neutrophil functions, like respiratory burst, chemotaxis, and phagocytosis. The i.v. injection of HoxB8 progenitors into lethally irradiated recipients resulted in the appearance of circulating donor-derived HoxB8 neutrophils. In vivo-differentiated HoxB8 neutrophils were able to migrate to the inflamed peritoneum and to phagocytose heat-killed Candida particles. The reverse passive Arthus reaction could be induced in HoxB8 chimeras but not in irradiated, nontransplanted control animals. Repeated injection of HoxB8 progenitors also allowed us to maintain stable circulating HoxB8 neutrophil counts for several days. Injection of arthritogenic K/B×N serum triggered robust arthritis in HoxB8 chimeras, but not in irradiated, nontransplanted control mice. Taken together, our results indicate that HoxB8 progenitor-derived neutrophils are capable of performing various in vivo neutrophil functions, providing a framework for using the HoxB8 system for the in vivo analysis of neutrophil function.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/genética , Proteínas de Homeodominio/metabolismo , Inflamación/inmunología , Células Progenitoras Mieloides/inmunología , Neutrófilos/inmunología , Animales , Diferenciación Celular , Línea Celular Transformada , Quimiotaxis , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Transgenes/genética
12.
Front Immunol ; 11: 1126, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32582203

RESUMEN

Myeloid cells are key components of the tumor microenvironment and critical regulators of disease progression. These innate immune cells are usually short-lived and require constant replenishment. Emerging evidence indicates that tumors alter the host hematopoietic system and induce the biased differentiation of myeloid cells to tip the balance of the systemic immune activities toward tumor-promoting functions. Altered myelopoiesis is not restricted to the bone marrow and also occurs in extramedullary organs. In this review, we outline the recent advances in the field of cancer-associated myelopoiesis, with a focus on the spleen, the major site of extramedullary hematopoiesis in the cancer setting. We discuss the functional specialization, distinct mechanisms, and clinical relevance of cancer-associated myeloid cell generation from early progenitors in the spleen and its potential as a novel therapeutic target.


Asunto(s)
Células Mieloides/inmunología , Células Progenitoras Mieloides/inmunología , Mielopoyesis/inmunología , Neoplasias/inmunología , Bazo/inmunología , Animales , Humanos
13.
Blood ; 136(2): 183-198, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32305041

RESUMEN

Hematopoietic stem cell (HSC) aging correlates with an increasing risk of myeloproliferative disease and immunosenescence. In this study, we show that aging-related inflammation promotes HSC aging through tumor necrosis factor-α (TNF-α)→ERK→ETS1→interleukin27Ra (IL27Ra) pathway. TNF-α, a well-known biomarker of inflammation, increases during aging and induces the expression of IL27Ra on HSCs via ERK-ETS1 signaling. Deletion of IL27Ra rescues the functional decline and myeloid bias of HSCs and also reverses the inhibitory effect of TNF-α on HSCs. Aged IL27Ra-/- mice had a reduced proportion of myeloid-biased HSCs and did not display the biased myeloid differentiation that occurs in aged wild-type mice. IL27Ra+ HSCs exhibit impaired reconstitution capacity and myeloid-bias compared with IL27Ra- HSCs and serve as a myeloid-recovery pool upon inflammatory insult. Inflammation-related genes were enriched in IL27Ra+ HSCs and this enrichment increases with aging. Our study demonstrates that age-induced IL27Ra signaling impairs HSCs and raises the possibility that interfering with IL27Ra signaling can counter the physiologically deleterious effect of aging on hematopoietic capacity.


Asunto(s)
Envejecimiento/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Células Progenitoras Mieloides/inmunología , Receptores de Interleucina/inmunología , Envejecimiento/genética , Envejecimiento/patología , Animales , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Células Progenitoras Mieloides/patología , Receptores de Interleucina/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
14.
J Leukoc Biol ; 107(6): 933-939, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32040234

RESUMEN

Systemic TNF neutralization can be used as a therapy for several autoimmune diseases. To evaluate the effects of cell type-restricted TNF blockade, we previously generated bispecific antibodies that can limit TNF secretion by myeloid cells (myeloid cell-specific TNF inhibitors or MYSTIs). In this study several such variable domain (VH) of a camelid heavy-chain only antibody-based TNF inhibitors were compared in relevant experimental models, both in vitro and in vivo. Pretreatment with MYSTI-2, containing the anti-F4/80 module, can restrict the release of human TNF (hTNF) from LPS-activated bone marrow-derived macrophage (BMDM) cultures of humanized TNF knock-in (mice; hTNFKI) more effectively than MYSTI-3, containing the anti-CD11b module. MYSTI-2 was also superior to MYSTI-3 in providing in vivo protection in acute toxicity model. Finally, MYSTI-2 was at least as effective as Infliximab in preventing collagen antibody-induced arthritis. This study demonstrates that a 33 kDa bispecific mini-antibody that specifically restricts TNF secretion by macrophages is efficient for amelioration of experimental arthritis.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Artritis Experimental/terapia , Antígeno CD11b/antagonistas & inhibidores , Proteínas de Unión al Calcio/antagonistas & inhibidores , Cadenas Pesadas de Inmunoglobulina/farmacología , Células Progenitoras Mieloides/efectos de los fármacos , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Inhibidores del Factor de Necrosis Tumoral/farmacología , Animales , Antirreumáticos/farmacología , Artritis Experimental/genética , Artritis Experimental/inmunología , Artritis Experimental/patología , Antígeno CD11b/genética , Antígeno CD11b/inmunología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/inmunología , Expresión Génica , Humanos , Infliximab/farmacología , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Células Progenitoras Mieloides/inmunología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
15.
J Virol ; 94(7)2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-31915281

RESUMEN

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that encodes many proteins to modulate the host immune response. Extensive efforts have led to the elucidation of multiple strategies employed by HCMV to effectively block NK cell targeting of virus-infected cells and the major histocompatibility complex (MHC) class I-primed CD8+ T cell response. However, viral regulation of the MHC class II-mediated CD4+ T cell response is understudied in endogenous MHC class II-expressing cells, largely because the popular cell culture systems utilized for studying HCMV do not endogenously express MHC class II. Of the many cell types infected by HCMV in the host, myeloid cells, such as monocytes, are of particular importance due to their role in latency and subsequent dissemination throughout the host. We investigated the impact of HCMV infection on MHC class II in Kasumi-3 cells, a myeloid-progenitor cell line that endogenously expresses the MHC class II gene, HLA-DR. We observed a significant reduction in the expression of surface and total HLA-DR at 72 h postinfection (hpi) and 120 hpi in infected cells. The decrease in HLA-DR expression was independent of the expression of previously described viral genes that regulate the MHC class II complex or the unique short (US) region of HCMV, a region expressing many immunomodulatory genes. The altered surface level of HLA-DR was not a result of increased endocytosis and degradation but was a result of a reduction in HLA-DR transcripts due to a decrease in the expression of the class II transactivator (CIITA).IMPORTANCE Human cytomegalovirus (HCMV) is an opportunistic herpesvirus that is asymptomatic for healthy individuals but that can lead to severe pathology in patients with congenital infections and immunosuppressed patients. Thus, it is important to understand the modulation of the immune response by HCMV, which is understudied in the context of endogenous MHC class II regulation. Using Kasumi-3 cells as a myeloid progenitor cell model endogenously expressing MHC class II (HLA-DR), this study shows that HCMV decreases the expression of HLA-DR in infected cells by reducing the transcription of HLA-DR transcripts early during infection independently of the expression of previously implicated genes. This is an important finding, as it highlights a mechanism of immune evasion utilized by HCMV to decrease the expression of MHC class II in a relevant cell system that endogenously expresses the MHC class II complex.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Antígenos HLA-DR/genética , Células Progenitoras Mieloides/inmunología , Proteínas Nucleares/genética , Transactivadores/genética , Activación Transcripcional , Presentación de Antígeno , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Línea Celular , Citomegalovirus , Endocitosis , Fibroblastos/inmunología , Fibroblastos/virología , Regulación de la Expresión Génica , Humanos , Células Progenitoras Mieloides/virología
16.
Front Immunol ; 11: 601494, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33424847

RESUMEN

We recently reported that offspring of mice treated during pregnancy with the microbial-derived immunomodulator OM-85 manifest striking resistance to allergic airways inflammation, and localized the potential treatment target to fetal conventional dendritic cell (cDC) progenitors. Here, we profile maternal OM-85 treatment-associated transcriptomic signatures in fetal bone marrow, and identify a series of immunometabolic pathways which provide essential metabolites for accelerated myelopoiesis. Additionally, the cDC progenitor compartment displayed treatment-associated activation of the XBP1-ERN1 signalling axis which has been shown to be crucial for tissue survival of cDC, particularly within the lungs. Our forerunner studies indicate uniquely rapid turnover of airway mucosal cDCs at baseline, with further large-scale upregulation of population dynamics during aeroallergen and/or pathogen challenge. We suggest that enhanced capacity for XBP1-ERN1-dependent cDC survival within the airway mucosal tissue microenvironment may be a crucial element of OM-85-mediated transplacental innate immune training which results in postnatal resistance to airway inflammatory disease.


Asunto(s)
Extractos Celulares/farmacología , Células Dendríticas/efectos de los fármacos , Endorribonucleasas/metabolismo , Inmunidad Innata/efectos de los fármacos , Intercambio Materno-Fetal/efectos de los fármacos , Células Progenitoras Mieloides/efectos de los fármacos , Placenta/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína 1 de Unión a la X-Box/metabolismo , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Endorribonucleasas/genética , Femenino , Redes Reguladoras de Genes , Ratones Endogámicos BALB C , Células Progenitoras Mieloides/inmunología , Células Progenitoras Mieloides/metabolismo , Mielopoyesis/efectos de los fármacos , Placenta/inmunología , Placenta/metabolismo , Embarazo , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Transcriptoma , Respuesta de Proteína Desplegada , Proteína 1 de Unión a la X-Box/genética
17.
Adv Immunol ; 143: 99-119, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31607369

RESUMEN

Rapid advances have been made to uncover the mechanisms that regulate dendritic cell (DC) development, and in turn, how models of development can be employed to define dendritic cell function. Models of DC development have been used to define the unique functions of DC subsets during immune responses to distinct pathogens. More recently, models of DC function have expanded to include their homeostatic and inflammatory physiology, modes of communication with various innate and adaptive immune lineages, and specialized functions across different lymphoid organs. New models of DC development call for revisions of previously accepted paradigms with respect to the ontogeny of plasmacytoid DC (pDC) and classical DC (cDC) subsets. By far, development of the cDC1 subset is best understood, and models have now been developed that can separate deficiencies in development from deficiencies in function. Such models are lacking for pDCs and cDC2s, limiting the depth of our understanding of their unique and essential roles during immune responses. If novel immunotherapies aim to harness the functions of human DCs, understanding of DC development will be essential to develop models DC function. Here we review emerging models of DC development and function.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/inmunología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Mieloides/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Citocinas/metabolismo , Células Dendríticas/citología , Humanos , Proteína 2 Inhibidora de la Diferenciación/genética , Proteína 2 Inhibidora de la Diferenciación/metabolismo , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Células Progenitoras Linfoides/metabolismo , Ratones , Modelos Inmunológicos , Células Progenitoras Mieloides/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo
18.
Mol Immunol ; 114: 314-322, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31442915

RESUMEN

Hematopoietic development occurs in the bone marrow, and this process begins with hematopoietic stem cells (HSCs). Ubc9 is a unique E2-conjugating enzyme required for SUMOylation, an evolutionarily conserved post-translational modification system. We herein show that a conditional Ubc9 deletion in the hematopoietic system caused decreased thymus weight and reduced lymphocyte to myeloid cell ratio. Importantly, Ubc9 deletion in the hematopoietic system only selectively impaired the development of common lymphoid progenitors (CLPs) in the bone marrow and perturbed their potential to differentiate into lymphocytes, thereby decreasing the number of T/B cells in the periphery. Ubc9 was found to be required for CLP viability, and therefore, Ubc9 deficiency rendered CLPs to undergo apoptosis and attenuated their proliferation. Thus, Ubc9 plays a critical role in the regulation of CLP function during hematopoietic development in the bone marrow.


Asunto(s)
Médula Ósea/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología , Enzimas Ubiquitina-Conjugadoras/deficiencia , Enzimas Ubiquitina-Conjugadoras/inmunología , Animales , Apoptosis/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Masculino , Ratones , Células Progenitoras Mieloides/inmunología , Linfocitos T/inmunología
19.
Nat Commun ; 10(1): 3817, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31444334

RESUMEN

Acquired resistance to targeted cancer therapy is a significant clinical challenge. In parallel with clinical trials combining CDK4/6 inhibitors to treat HER2+ breast cancer, we sought to prospectively model tumor evolution in response to this regimen in vivo and identify a clinically actionable strategy to combat drug resistance. Despite a promising initial response, acquired resistance emerges rapidly to the combination of anti-HER2/neu antibody and CDK4/6 inhibitor Palbociclib. Using high-throughput single-cell profiling over the course of treatments, we reveal a distinct immunosuppressive immature myeloid cell (IMC) population to infiltrate the resistant tumors. Guided by single-cell transcriptome analysis, we demonstrate that combination of IMC-targeting tyrosine kinase inhibitor cabozantinib and immune checkpoint blockade enhances anti-tumor immunity, and overcomes the resistance. Furthermore, sequential combinatorial immunotherapy enables a sustained control of the fast-evolving CDK4/6 inhibitor-resistant tumors. Our study demonstrates a translational framework for treating rapidly evolving tumors through preclinical modeling and single-cell analyses.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Modelos Biológicos , Células Progenitoras Mieloides/efectos de los fármacos , Células Progenitoras Mieloides/inmunología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Análisis de la Célula Individual , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Curr Opin Lipidol ; 30(5): 395-400, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31335332

RESUMEN

PURPOSE OF REVIEW: The two major challenges in cardiovascular medicine are to refine risk prediction and to improve pharmacological prevention and treatment. The concept of innate immune memory, which is called trained immunity, has the potential to improve clinical practice in these regards. RECENT FINDINGS: Monocytes and macrophages have the capability to develop a long-term proinflammatory and proatherogenic phenotype after brief exposure to inflammatory stimuli, such as oxidized low-density lipoprotein particles. This innate immune memory develops because of rewiring of intracellular metabolic pathways and epigenetic reprogramming of histone modifications. The persistence of circulating hyperresponsive monocytes in vivo is explained by the fact that training occurs in myeloid progenitor cells in the bone marrow. Several recent studies reported the presence of monocytes with a trained immune phenotype in patients with established atherosclerosis, and in patients with an increased risk for atherosclerosis because of dyslipoproteinemia. SUMMARY: In monocytes and their bone marrow progenitors, metabolic and epigenetic reprogramming can induce trained immunity, which might contribute to the persistent nonresolving inflammation that characterizes atherosclerosis. These pathways offer exciting novel drug targets to improve the prevention and treatment of cardiovascular disease.


Asunto(s)
Aterosclerosis/inmunología , Enfermedades Cardiovasculares/inmunología , Inflamación/inmunología , Lipoproteínas LDL/genética , Aterosclerosis/genética , Células de la Médula Ósea/inmunología , Enfermedades Cardiovasculares/genética , Epigénesis Genética/inmunología , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inflamación/genética , Lipoproteínas LDL/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Células Progenitoras Mieloides/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA