Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Blood ; 139(22): 3303-3313, 2022 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-35313334

RESUMEN

Pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL) outcome has improved in the last decades, but leukemic relapses are still one of the main problems of this disease. Bone morphogenetic protein 4 (BMP4) was investigated as a new candidate biomarker with potential prognostic relevance, and its pathogenic role was assessed in the development of disease. A retrospective study was performed with 115 pediatric patients with BCP-ALL, and BMP4 expression was analyzed by quantitative reverse transcription polymerase chain reaction in leukemic blasts at the time of diagnosis. BMP4 mRNA expression levels in the third (upper) quartile were associated with a higher cumulative incidence of relapse as well as a worse 5-year event-free survival and central nervous system (CNS) involvement. Importantly, this association was also evident among children classified as having a nonhigh risk of relapse. A validation cohort of 236 patients with BCP-ALL supported these data. Furthermore, high BMP4 expression promoted engraftment and rapid disease progression in an NSG mouse xenograft model with CNS involvement. Pharmacological blockade of the canonical BMP signaling pathway significantly decreased CNS infiltration and consistently resulted in amelioration of clinical parameters, including neurological score. Mechanistically, BMP4 favored chemoresistance, enhanced adhesion and migration through brain vascular endothelial cells, and promoted a proinflammatory microenvironment and CNS angiogenesis. These data provide evidence that BMP4 expression levels in leukemic cells could be a useful biomarker to identify children with poor outcomes in the low-/intermediate-risk groups of BCP-ALL and that BMP4 could be a new therapeutic target to blockade leukemic CNS disease.


Asunto(s)
Linfoma de Burkitt , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Proteína Morfogenética Ósea 4/genética , Niño , Células Endoteliales/metabolismo , Humanos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Recurrencia , Estudios Retrospectivos , Microambiente Tumoral
2.
Exp Dermatol ; 33(8): e15146, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39075828

RESUMEN

Patients with recessive dystrophic epidermolysis bullosa (RDEB) experience numerous complications, which are exacerbated by inflammatory dysregulation and infection. Understanding the immunological mechanisms is crucial for selecting medications that balance inflammation control and immunocompetence. In this cross-sectional study, aiming to identify potential immunotherapeutic targets and inflammatory biomarkers, we delved into the interrelationship between clinical severity and systemic inflammatory parameters in a representative RDEB cohort. Encompassing 84 patients aged 1-67 and spanning all three Epidermolysis Bullosa Disease Activity and Scarring Index (EBDASI) severity categories, we analysed the interrelationship of infection history, standard inflammatory markers, systemic cytokines and Ig levels to elucidate their roles in RDEB pathophysiology. Our findings identify C-reactive protein as an excellent biomarker for disease severity in RDEB. A type 2 inflammatory profile prevails among moderate and severe RDEB patients, correlating with dysregulated circulating IgA and IgG. These results underscore the IL4/IL13 pathways as potential evidence-based therapeutic targets. Moreover, the complete inflammatory scenario aligns with Staphylococcus aureus virulence mechanisms. Concurrently, abnormalities in IgG, IgE and IgM levels suggest an immunodeficiency state in a substantial number of the cohort's patients. Our results provide new insights into the interplay of infection and immunological factors in the pathogenesis of RDEB.


Asunto(s)
Biomarcadores , Proteína C-Reactiva , Epidermólisis Ampollosa Distrófica , Interleucina-13 , Interleucina-4 , Índice de Severidad de la Enfermedad , Humanos , Estudios Transversales , Biomarcadores/sangre , Niño , Preescolar , Interleucina-4/sangre , Adolescente , Proteína C-Reactiva/metabolismo , Adulto , Adulto Joven , Femenino , Masculino , Lactante , Persona de Mediana Edad , Interleucina-13/sangre , Interleucina-13/metabolismo , Anciano
3.
J Pathol ; 252(2): 189-200, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32686161

RESUMEN

Despite current central nervous system-directed therapies for childhood B-cell precursor acute lymphoblastic leukaemia, relapse at this anatomical site still remains a challenging issue. Few reports have addressed the study of the specific cellular microenvironments which can promote the survival, quiescence, and therefore chemoresistance of B-cell precursor acute lymphoblastic leukaemia cells in the central nervous system. Herein, we showed by immunofluorescence and electron microscopy that in xenotransplanted mice, leukaemic cells infiltrate the connective tissue stroma of the choroid plexus, the brain structure responsible for the production of cerebrospinal fluid. The ultrastructural study also showed that leukaemia cells are able to migrate through blood vessels located in the choroid plexus stroma. In short-term co-cultures, leukaemic cells established strong interactions with human choroid plexus fibroblasts, mediated by an increased expression of ITGA4 (VLA-4)/ITGAL (LFA-1) and their ligands VCAM1/ICAM1. Upon contact with leukaemia cells, human choroid plexus fibroblasts acquired a cancer-associated fibroblast phenotype, with an increased expression of α-SMA and vimentin as well as pro-inflammatory factors. Human choroid plexus fibroblasts also have the capacity to reduce the proliferative index of leukaemic blasts and promote their survival and chemoresistance to methotrexate and cytarabine. The inhibition of VLA-4/VCAM-1 interactions using anti-VLA-4 antibodies, and the blockade of Notch signalling pathway by using a γ-secretase inhibitor partially restored chemotherapy sensitivity of leukaemia cells. We propose that the choroid plexus stroma constitutes a sanctuary for B-cell precursor acute lymphoblastic leukaemia cells in the central nervous system. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Plexo Coroideo/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Animales , Niño , Resistencia a Antineoplásicos/fisiología , Fibroblastos/patología , Xenoinjertos , Humanos , Ratones
4.
Int J Mol Sci ; 22(20)2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34681929

RESUMEN

Extracellular vesicles (EVs) derived from mesenchymal stromal cells (MSCs) have therapeutic potential in the treatment of several immune disorders, including ulcerative colitis, owing to their regenerative and immunosuppressive properties. We recently showed that MSCs engineered to overexpress hypoxia-inducible factor 1-alpha and telomerase (MSC-T-HIF) and conditioned with pro-inflammatory stimuli release EVs (EVMSC-T-HIFC) with potent immunomodulatory activity. We tested the efficacy of EVMSC-T-HIFC to repolarize M1 macrophages (Mφ1) to M2-like macrophages (Mφ2-like) by analyzing surface markers and cytokines and performing functional assays in co-culture, including efferocytosis and T-cell proliferation. We also studied the capacity of EVMSC-T-HIFC to dampen the inflammatory response of activated endothelium and modulate fibrosis. Finally, we tested the therapeutic capacity of EVMSC-T-HIFC in an acute colitis model. EVMSC-T-HIFc induced the repolarization of monocytes from Mφ1 to an Mφ2-like phenotype, which was accompanied by reduced inflammatory cytokine release. EVMSC-T-HIFc-treated Mφ1 had similar effects of immunosuppression on activated peripheral blood mononuclear cells (PBMC) as Mφ2, and reduced the adhesion of PBMCs to activated endothelium. EVMSC-T-HIFc also prevented myofibroblast differentiation of TGF-ß-treated fibroblasts. Finally, administration of EVMSC-T-HIFc promoted healing in a TNBS-induced mouse colitis model in terms of preserving colon length and intestinal mucosa architecture and altering the ratio of Mφ1/ Mφ2 infiltration. In conclusion, EVMSC-T-HIFC have effective anti-inflammatory properties, making them potential therapeutic agents in cell free-based therapies for the treatment of Crohn's disease and likely other immune-mediated inflammatory diseases.


Asunto(s)
Enfermedad de Crohn/terapia , Vesículas Extracelulares/trasplante , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Mesenquimatosas/metabolismo , Animales , Adhesión Celular , Polaridad Celular , Enfermedad de Crohn/inducido químicamente , Enfermedad de Crohn/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Vesículas Extracelulares/genética , Vesículas Extracelulares/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Telomerasa/metabolismo , Ácido Trinitrobencenosulfónico/efectos adversos , Adulto Joven
6.
Cytotherapy ; 19(5): 640-653, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28262465

RESUMEN

Fibroblastic reticular cells (FRCs) are essential players during adaptive immune responses not only as a structural support for the encounter of antigen-presenting cells and naive T lymphocytes but also as a source of modulatory signals. However, little is known about this cell population in humans. To address the phenotypical and functional analysis of human FRCs here we established splenic (SP) and mesenteric lymph node (LN) CD45-CD31-CD90+podoplanin+ myofibroblastic cell cultures. They shared the phenotypical characteristics distinctive of FRCs, including the expression of immunomodulatory factors and peripheral tissue antigens. Nevertheless, human FRCs also showed particular features, some differing from mouse FRCs, like the lack of nitric oxide synthase (NOS2) expression after interferon (IFN)γstimulation. Interestingly, SP-FRCs expressed higher levels of interleukin (IL)-6, BMP4, CCL2, CXCL12 and Notch molecules, and strongly adapted their functional profile to lipopolysaccharide (LPS), polyinosinic:polycytidylic acid (Poly I:C) and IFNγ stimulation. In contrast, we found higher expression of transforming growth factor (TGF)ß and Activin A in LN-FRCs that barely responded via Toll-Like Receptor (TLR)3 and constitutively expressed retinaldehyde dehydrogenase 1 enzyme, absent in SP-FRCs. This study reveals human FRCs can be valuable models to increase our knowledge about the physiology of human secondary lymphoid organs in health and disease and to explore the therapeutic options of FRCs.


Asunto(s)
Fibroblastos/citología , Inmunoterapia , Inmunidad Adaptativa , Animales , Proteínas de Unión al ADN , Femenino , Fibroblastos/metabolismo , Humanos , Inmunomodulación , Terapia de Inmunosupresión , Inflamación/patología , Ganglios Linfáticos/citología , Masculino , Ratones , Miofibroblastos/citología , Miofibroblastos/metabolismo , Proteínas Nucleares/metabolismo , Fenotipo , Bazo/citología , Linfocitos T/citología , Linfocitos T/inmunología , Factores de Transcripción
7.
Cytotherapy ; 18(10): 1297-311, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27637760

RESUMEN

BACKGROUND AIMS: The immunomodulatory properties of mesenchymal stromal cells (MSCs), together with their tissue regenerative potential, make them interesting candidates for clinical application. METHODS: In the current study, we analyzed the in vitro immunomodulatory effects of MSCs derived from bone marrow (BM-MSCs) and from adipose tissue (AT-MSCs) obtained from the same donor on both innate and acquired immunity cells. BM-MSCs and AT-MSCs were expanded to fourth or fifth passage and co-cultured with T cells, monocytes or natural killer (NK) cells isolated from human peripheral blood and stimulated in vitro. The possible differing impact of MSCs obtained from distinct sources on phenotype, cell proliferation and differentiation, cytokine production and function of these immune cells was comparatively analyzed. RESULTS: BM-MSCs and AT-MSCs induced a similar decrease in NK-cell proliferation, cytokine secretion and expression of both activating receptors and cytotoxic molecules. However, only BM-MSCs significantly reduced NK-cell cytotoxic activity, although both MSC populations showed the same susceptibility to NK-cell-mediated lysis. AT-MSCs were more potent in inhibiting dendritic-cell (DC) differentiation than BM-MSC, but both MSC populations similarly reduced the ability of DCs to induce CD4(+) T-cell proliferation and cytokine production. BM-MSCs and AT-MSCs induced a similar decrease in T-cell proliferation and production of inflammatory cytokines after activation. CONCLUSIONS: AT-MSCs and BM-MSCs from the same donor had similar immunomodulatory capacity on both innate and acquired immunity cells. Thus, other variables, such as accessibility of samples or the frequency of MSCs in the tissue should be considered to select the source of MSC for cell therapy.


Asunto(s)
Tejido Adiposo/citología , Células de la Médula Ósea/fisiología , Inmunomodulación/fisiología , Células Madre Mesenquimatosas/fisiología , Linfocitos T/inmunología , Adulto , Anciano , Células de la Médula Ósea/citología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Femenino , Humanos , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Masculino , Células Madre Mesenquimatosas/citología , Persona de Mediana Edad , Donantes de Tejidos
8.
Eur J Immunol ; 44(4): 1031-8, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24532425

RESUMEN

Bone morphogenetic proteins (BMPs) are multifunctional growth factors regulating differentiation and proliferation in numerous systems including the immune system. Previously, we described that the BMP signaling pathway is functional in human monocyte-derived dendritic cells (MoDCs), which were found to express both the specific receptors and the Smad proteins required for signal transduction. In this study, we provide evidence that human MoDCs produce BMP-4 and that this production is increased over the maturation process as is BMP signal transduction. When DCs are matured in the presence of an inhibitor of the BMP pathway, the expression of the maturation markers PD-L1 and PD-L2 is reduced, while cytokine production is not affected. As a result, these mature DCs present an augmented ability to stimulate both T cells and NK cells. Eventually, the inhibition of BMP signaling during maturation causes a reduced expression of IRF-1, a transcription factor that positively regulates the expression of PD-L1 and PD-L2. The present study indicates that the BMP signaling pathway regulates PD-L1 and PD-L2 expression in human MoDCs during the maturation process, probably through the IRF-1 transcription factor, and also points out that the manipulation of BMP signaling might considerably improve the immunogenicity of MoDCs used in immunotherapy.


Asunto(s)
Comunicación Autocrina/inmunología , Antígeno B7-H1/inmunología , Proteína Morfogenética Ósea 4/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Transducción de Señal/inmunología , Comunicación Autocrina/genética , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Citotoxicidad Inmunológica/genética , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Expresión Génica/inmunología , Humanos , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Factor 1 Regulador del Interferón/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
9.
Immunol Cell Biol ; 93(7): 673-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25753268

RESUMEN

Human thymus contains two major subpopulations of dendritic cells (DCs), conventional DCs (cDCs) and plasmacytoid DCs (pDCs), which are mainly involved in central tolerance and also in protecting the thymus against infections. In blood and peripheral organs cDCs include the subpopulation of BDCA3(hi) DCs, considered as equivalents to mouse CD8α(+) DCs. In this study we describe in human thymus the presence of a discrete population of BDCA3(hi) DCs that, like their peripheral counterparts, express CD13, low-intermediate levels of CD11c, CLEC9A, high levels of XCR1, IRF8 and TLR3, and mostly lack the expression of CD11b, CD14 and TLR7. Thymic BDCA3(hi) DCs display immature features with a low expression of costimulatory molecules and HLA-DR, and a low allostimulatory capacity. Also, BDCA3(hi) DCs exhibit a strong response to TLR3 stimulation, producing high levels of interferon (IFN)-λ1 and CXCL10, which indicates that, similarly to thymic pDCs, BDCA3(hi) DCs can have an important role in thymus protection against viral infections.


Asunto(s)
Antígenos de Superficie/análisis , Células Dendríticas/citología , Interleucinas/análisis , Timo/citología , Antígenos de Diferenciación/análisis , Apoptosis , Células Cultivadas , Quimiocina CXCL10/análisis , Preescolar , Técnicas de Cocultivo , Células Dendríticas/química , Células Dendríticas/clasificación , Antígenos HLA-DR/análisis , Humanos , Lactante , Recién Nacido , Interferones , Interleucinas/biosíntesis , Interleucinas/genética , Lectinas Tipo C/análisis , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Acoplados a Proteínas G/análisis , Receptores Mitogénicos/análisis , Trombomodulina , Timo/inmunología , Receptor Toll-Like 3/análisis
10.
Blood ; 119(8): 1861-71, 2012 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-22210872

RESUMEN

The bone morphogenetic protein (BMP) signaling pathway regulates survival, proliferation, and differentiation of several cell types in multiple tissues, including the thymus. Previous reports have shown that BMP signaling negatively regulates T-cell development. Here, we study the subpopulation of early human intrathymic progenitors expressing the type IA BMP receptor (BMPRIA) and provide evidence that CD34(+)CD1a(-)BMPRIA(+) precursor cells mostly express surface cell markers and transcription factors typically associated with NK cell lineage. These CD34(+) cells mostly differentiate into functional CD56(+) natural killer (NK) cells when they are cocultured with thymic stromal cells in chimeric human-mouse fetal thymic organ cultures and also in the presence of SCF and IL-15. Moreover, autocrine BMP signaling can promote the differentiation of thymic NK cells by regulating the expression of key transcription factors required for NK cell lineage (eg, Id3 and Nfil3) as well as one of the components of IL-15 receptor, CD122. Subsequently, the resulting population of IL-15-responsive NK cell precursors can be expanded by IL-15, whose action is mediated by BMP signaling during the last steps of thymic NK cell differentiation. Our results strongly suggest that BMPRIA expression identifies human thymic NK cell precursors and that BMP signaling is relevant for NK cell differentiation in the human thymus.


Asunto(s)
Proteína Morfogenética Ósea 4/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/metabolismo , Células Asesinas Naturales/metabolismo , Transducción de Señal , Timocitos/metabolismo , Animales , Antígenos CD34/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Antígeno CD56/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Células Cultivadas , Preescolar , Técnicas de Cocultivo , Citometría de Flujo , Expresión Génica , Humanos , Células Híbridas/metabolismo , Células Híbridas/ultraestructura , Inmunofenotipificación , Lactante , Interleucina-15/farmacología , Ratones , Ratones SCID , Microscopía Electrónica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timo/citología , Timo/embriología
11.
J Histochem Cytochem ; 72(5): 289-307, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38725414

RESUMEN

Several types of cytotoxic insults disrupt endoplasmic reticulum (ER) homeostasis, cause ER stress, and activate the unfolded protein response (UPR). The role of ER stress and UPR activation in hypersensitivity pneumonitis (HP) has not been described. HP is an immune-mediated interstitial lung disease that develops following repeated inhalation of various antigens in susceptible and sensitized individuals. The aim of this study was to investigate the lung expression and localization of the key effectors of the UPR, BiP/GRP78, CHOP, and sXBP1 in HP patients compared with control subjects. Furthermore, we developed a mouse model of HP to determine whether ER stress and UPR pathway are induced during this pathogenesis. In human control lungs, we observed weak positive staining for BiP in some epithelial cells and macrophages, while sXBP1 and CHOP were negative. Conversely, strong BiP, sXBP1- and CHOP-positive alveolar and bronchial epithelial, and inflammatory cells were identified in HP lungs. We also found apoptosis and autophagy markers colocalization with UPR proteins in HP lungs. Similar results were obtained in lungs from an HP mouse model. Our findings suggest that the UPR pathway is associated with the pathogenesis of HP.


Asunto(s)
Alveolitis Alérgica Extrínseca , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Células Epiteliales , Proteínas de Choque Térmico , Factor de Transcripción CHOP , Respuesta de Proteína Desplegada , Proteína 1 de Unión a la X-Box , Animales , Alveolitis Alérgica Extrínseca/patología , Alveolitis Alérgica Extrínseca/inmunología , Alveolitis Alérgica Extrínseca/metabolismo , Humanos , Ratones , Proteína 1 de Unión a la X-Box/metabolismo , Proteína 1 de Unión a la X-Box/genética , Proteínas de Choque Térmico/metabolismo , Factor de Transcripción CHOP/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Masculino , Pulmón/patología , Pulmón/inmunología , Pulmón/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción del Factor Regulador X/metabolismo , Factores de Transcripción/metabolismo , Modelos Animales de Enfermedad , Persona de Mediana Edad , Ratones Endogámicos C57BL , Adulto , Inflamación/patología , Inflamación/metabolismo , Inflamación/inmunología
12.
Front Immunol ; 15: 1365015, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39391311

RESUMEN

Inflammation plays a pivotal role in the pathogenesis of primary and post-essential thrombocythemia or post-polycythemia vera myelofibrosis (MF) in close cooperation with the underlying molecular drivers. This inflammatory state is induced by a dynamic spectrum of inflammatory cytokines, although recent evidence points to the participation of additional soluble inflammatory mediators. Damage-associated molecular patterns (DAMPs) represent endogenous signals released upon cell death or damage which trigger a potent innate immune response. We assessed the contribution of two prototypical DAMPs, HMGB1 and S100A8/A9, to MF inflammation. Circulating HMGB1 and S100A8/A9 were elevated in MF patients in parallel to the degree of systemic inflammation and levels increased progressively during advanced disease stages. Patients with elevated DAMPs had higher frequency of adverse clinical features, such as anemia, and inferior survival, suggesting their contribution to disease progression. Monocytes, which are key players in MF inflammation, were identified as a source of S100A8/A9 but not HMGB1 release, while both DAMPs correlated with cell death parameters, such as serum LDH and cell-free DNA, indicating that passive release is an additional mechanism leading to increased DAMPs. HMGB1 and S100A8/A9 promote inflammation through binding to Toll-like receptor (TLR) 4, whereas the former also binds TLR2. Monocytes from MF patients were shown to be hyperactivated at baseline, as reflected by higher CD11b and tissue factor exposure and increased expression levels of proinflammatory cytokines IL-1ß and IL-6. Patient monocytes showed preserved TLR4 and TLR2 expression and were able to mount normal or even exacerbated functional responses and cytokine upregulation following stimulation of TLR4 and TLR2. Elevated levels of endogenous TLR ligands HMGB1 and S100A8/A9 coupled to the finding of preserved or hyperreactive TLR-triggered responses indicate that DAMPs may promote monocyte activation and cytokine production in MF, fueling inflammation. Plasma IL-1ß and IL-6 were elevated in MF and correlated with DAMPs levels, raising the possibility that DAMPs could contribute to cytokine generation in vivo. In conclusion, this study highlights that, in cooperation with classic proinflammatory cytokines, DAMPs represent additional inflammatory mediators that may participate in the generation of MF inflammatory state, potentially providing novel biomarkers of disease progression and new therapeutic targets.


Asunto(s)
Alarminas , Calgranulina A , Calgranulina B , Proteína HMGB1 , Inflamación , Monocitos , Mielofibrosis Primaria , Humanos , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Calgranulina A/sangre , Calgranulina B/sangre , Masculino , Femenino , Monocitos/inmunología , Monocitos/metabolismo , Anciano , Persona de Mediana Edad , Alarminas/metabolismo , Alarminas/inmunología , Inflamación/inmunología , Mielofibrosis Primaria/inmunología , Mielofibrosis Primaria/metabolismo , Anciano de 80 o más Años , Receptores Toll-Like/metabolismo , Citocinas/metabolismo , Adulto , Receptor Toll-Like 4/metabolismo , Biomarcadores
13.
Haematologica ; 103(10): e458-e461, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29748439
14.
J Immunol ; 187(8): 4129-39, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21918189

RESUMEN

Dendritic cells (DCs) are critical regulators of immune responses that integrate signals from the innate and adaptive immune system and orchestrate T cell responses toward either immunity or tolerance. Growing evidence points to the Wnt signaling pathway as a pivotal piece in the immune balance and focuses on DCs as a direct target for their immunoregulatory role. Our results show that the increase in Wnt5a signaling during the differentiation of human DCs from monocytes alters their phenotype and compromises their subsequent capacity to mature in response to TLR-dependent stimuli. These Wnt5a-DCs produce scant amounts of IL-12p70 and TNF-α but increased levels of IL-10. Consequently, these Wnt5a-DCs have a reduced capacity to induce Th1 responses that promote IL-10 secretion by CD4 T cells. Changes in the transcriptional profile of Wnt5a-DCs correlate with their unconventional phenotype caused presumably by increased IL-6/IL-10 signaling during the process of DC differentiation. The effect of Wnt5a is not a consequence of ß-catenin accumulation but is dependent on noncanonical Ca(2+)/calmodulin-dependent protein kinase II/NF-κB signaling. Our results therefore suggest that under high levels of Wnt5a, typical of the inflammatory state and sepsis, monocytes could differentiate into unconventional DCs with tolerogenic features.


Asunto(s)
Diferenciación Celular/inmunología , Células Dendríticas/citología , Tolerancia Inmunológica/inmunología , Monocitos/citología , Proteínas Proto-Oncogénicas/inmunología , Proteínas Wnt/inmunología , Separación Celular , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Activación de Linfocitos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Monocitos/inmunología , Monocitos/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Proteínas Wnt/metabolismo , Proteína Wnt-5a
15.
Immunol Cell Biol ; 89(5): 610-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21102536

RESUMEN

Bone morphogenetic proteins (BMPs), members of the transforming growth factor-ß superfamily, are multifunctional polypeptides regulating a broad spectrum of functions in embryonic and adult tissues. Recent reports have demonstrated that BMPs regulate the survival, proliferation and differentiation of several cell types in the immune system. In this study, we investigate the effects of BMP signaling activation on the capacity of human dendritic cells (DCs) to stimulate immune responses. Human DCs express type I and type II BMP receptors (BMPRIA, BMPRIB, type IA activin receptor, BMPRII) and BMP signal transduction molecules (Smad1, 5, and 8, as well as Smad4). On BMP stimulation, Id1-3 (inhibitor of differentiation 1-3/DNA binding) mRNA expression is upregulated and this effect can be blocked with the inhibitor dorsomorphin, showing that the canonical BMP signal transduction pathway is functionally active in DCs. BMP signaling activation promotes the phenotypic maturation of human DCs by increasing the expression of co-stimulatory molecules and also CD83, programmed cell death ligand 1 (PD-L1) and PD-L2, and stimulates cytokine secretion, mainly interleukin-8 and tumor necrosis factor-α. Accordingly, BMP-treated DCs exhibit an enhanced T-cell stimulatory capacity. BMP signaling also enhances the survival of human DCs increasing the Bcl-2/Bax ratio. Finally, the expression of Runx transcription factors is increased in mature DCs, and the mRNA levels of Runx1-3 are upregulated in response to BMP stimulation, indicating that Runx transcription factor family may mediate the effects of BMP signaling in human DC maturation.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Monocitos/citología , Proteínas Morfogenéticas Óseas/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Subunidades alfa del Factor de Unión al Sitio Principal/genética , Citocinas/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunización , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Fenotipo , ARN Mensajero/genética , Transducción de Señal/efectos de los fármacos
16.
Neuroimmunomodulation ; 17(3): 217-20, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20134207

RESUMEN

CXCL12 is an important CXC chemokine involved in numerous biological processes. We had previously demonstrated the synergistic participation of CXCL12 and IL-7 in the control of both survival and proliferation of CD34(+) human thymic lymphoid progenitors. On this basis, we hypothesize a presumptive role for CXCL12 and its receptor, CXCR4, in the thymus involution. In this respect, in the current report we describe the expression of both molecules in the human thymus during aging. Our results demonstrate that, despite the profound alterations observed in the thymic epithelial microenvironment of aged thymuses, the proportions of different CD4/CD8 thymocyte subsets do not undergo significant variations. Remarkably, a strong CXCL12 expression was found in older thymuses, which appeared in the same locations as in younger thymuses: the subcapsulary and medullary areas. The proportions of CXCR4(+) cells, most of them belonging to the CD3(-) compartment, showed no important variations in the older thymuses. However, within the CD34(+) cell population, a significant reduction in the expression of CXCR4 molecules was observed.


Asunto(s)
Envejecimiento/inmunología , Quimiocina CXCL12/metabolismo , Receptores CXCR4/metabolismo , Linfocitos T/inmunología , Timo/inmunología , Adolescente , Adulto , Anciano , Antígenos CD34/metabolismo , Atrofia/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Homeostasis/inmunología , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Células Madre/citología , Células Madre/inmunología , Linfocitos T/citología , Timo/citología , Adulto Joven
17.
Front Immunol ; 11: 567391, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329530

RESUMEN

Recent clinical observations indicate that bacterial vaccines induce cross-protection against infections produced by different microorganisms. MV130, a polyvalent bacterial sublingual preparation designed to prevent recurrent respiratory infectious diseases, reduces the infection rate in patients with recurrent respiratory tract infections. On the other hand, mesenchymal stem cells (MSCs) are key cell components that contribute to the maintenance of tissue homeostasis and exert both immunostimulatory and immunosuppressive functions. Herein, we study the effects of MV130 in human MSC functionality as a potential mechanism that contributes to its clinical benefits. We provide evidence that during MV130 sublingual immunization of mice, resident oral mucosa MSCs can take up MV130 components and their numbers remain unchanged after vaccination, in contrast to granulocytes that are recruited from extramucosal tissues. MSCs treated in vitro with MV130 show an increased viability without affecting their differentiation potential. In the short-term, MSC treatment with MV130 induces higher leukocyte recruitment and T cell expansion. In contrast, once T-cell activation is initiated, MV130 stimulation induces an up-regulated expression of immunosuppressor factors in MSCs. Accordingly, MV130-primed MSCs reduce T lymphocyte proliferation, induce the differentiation of dendritic cells with immunosuppressive features and favor M2-like macrophage polarization, thus counterbalancing the immune response. In addition, MSCs trained with MV130 undergo functional changes, enhancing their immunomodulatory response to a secondary stimulus. Finally, we show that MSCs are able to uptake, process and retain a reservoir of the TLR ligands derived from MV130 digestion which can be subsequently transferred to dendritic cells, an additional feature that also may be associated to trained immunity.


Asunto(s)
Vacunas Bacterianas/inmunología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Mucosa Bucal/inmunología , Mucosa Bucal/metabolismo , Administración Sublingual , Animales , Vacunas Bacterianas/administración & dosificación , Citocinas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunización , Memoria Inmunológica , Inmunomodulación , Inmunofenotipificación , Inflamación/etiología , Inflamación/metabolismo , Inflamación/terapia , Leucocitos/inmunología , Leucocitos/metabolismo , Activación de Linfocitos/inmunología , Células Madre Mesenquimatosas/citología , Ratones , Receptores de Reconocimiento de Patrones/metabolismo
18.
Front Immunol ; 11: 705, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32425934

RESUMEN

Essential thrombocythemia (ET) is comprised among chronic myeloproliferative neoplasms (MPN) and is caused by driver mutations in JAK2, CALR, and MPL, which lead to megakaryocyte proliferation and prominent thrombocytosis. Thrombosis remains the main cause of morbidity in ET and is driven by the interplay between blood cells, the endothelium, the clotting cascade, and host-derived inflammatory mediators. Platelet activation plays a key role in the thrombotic predisposition, although the underlying mechanisms remain poorly defined. In addition to their role in hemostasis, platelets participate in innate immunity and inflammation owing to the expression of toll-like receptors (TLR), which recognize inflammatory signals, triggering platelet functional responses. Considering the impact of inflammation on ET procoagulant state, we assessed the contribution of TLR2 and TLR4 to platelet hemostatic and inflammatory properties in ET patients, by using Pam3CSK4 and lipopolysaccharide (LPS) as specific TLR2 and TLR4 ligands, respectively. TLR2 ligation induced increased surface translocation of α-granule-derived P-selectin and CD40L, which mediate platelet interaction with leukocytes and endothelial cells, respectively, and higher levels of dense granule-derived CD63 in patients, whereas PAC-1 binding was not increased and LPS had no effect on these platelet responses. Platelet-neutrophil aggregate formation was elevated in ET at baseline and after stimulation of both TLR2 and TLR4. In addition, ET patients displayed higher TLR2- and TLR4-triggered platelet secretion of the chemokine RANTES (CCL5), whereas von Willebrand factor release was not enhanced, revealing a differential releasate pattern for α-granule-stored inflammatory molecules. TLR-mediated hyperresponsiveness contrasted with impaired or preserved responses to classic platelet hemostatic agonists, such as TRAP-6 and thrombin. TLR2 and TLR4 expression on the platelet surface was normal, whereas phosphorylation of downstream effector ERK1/2 was higher in patients at baseline and after incubation with Pam3CSK4, which may partly explain the enhanced TLR2 response. In conclusion, exacerbated response to TLR stimulation may promote platelet activation in ET, boosting platelet/leukocyte/endothelial interactions and secretion of inflammatory mediators, overall reinforcing the thromboinflammatory state. These findings highlight the role of platelets as inflammatory sentinels in MPN prothrombotic scenario and provide additional evidence for the close intertwining between thrombosis and inflammation in this setting.


Asunto(s)
Plaquetas/fisiología , Inflamación/etiología , Trombocitemia Esencial/complicaciones , Trombosis/etiología , Receptores Toll-Like/fisiología , Adulto , Anciano , Quimiocina CCL5/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neutrófilos/fisiología , Fosforilación , Activación Plaquetaria , Trombocitemia Esencial/inmunología
19.
Front Med (Lausanne) ; 7: 576558, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33324660

RESUMEN

Recessive dystrophic epidermolysis bullosa (RDEB) is an incurable inherited mucocutaneous fragility disorder characterized by recurrent blisters, erosions, and wounds. Continuous blistering triggers overlapping cycles of never-ending healing and scarring commonly evolving to chronic systemic inflammation and fibrosis. The systemic treatment with allogeneic mesenchymal cells (MSC) from bone marrow has previously shown benefits in RDEB. MSC from adipose tissue (ADMSC) are easier to isolate. This is the first report on the use of systemic allogeneic ADMSC, correlating the clinical, inflammatory, and immunologic outcomes in RDEB indicating long-lasting benefits. We present the case of an RDEB patient harboring heterozygous biallelic COL7A1 gene mutations and with a diminished expression of C7. The patient presented with long-lasting refractory and painful oral ulcers distressing her quality of life. Histamine receptor antagonists, opioid analgesics, proton-pump inhibitors, and low-dose tricyclic antidepressants barely improved gastric symptoms, pain, and pruritus. Concomitantly, allogeneic ADMSC were provided as three separate intravenous injections of 106 cells/kg every 21 days. ADMSC treatment was well-tolerated. Improvements in wound healing, itch, pain and quality of life were observed, maximally at 6-9 months post-treatment, with the relief of symptoms still noticeable for up to 2 years. Remarkably, significant modifications in PBL participating in both the innate and adaptive responses, alongside regulation of levels of profibrotic factors, MCP-1/CCL2 and TGF-ß, correlated with the health improvement. This treatment might represent an alternative for non-responding patients to conventional management. It seems critical to elucidate the paracrine modulation of the immune system by MSC for their rational use in regenerative/immunoregulatory therapies.

20.
J Leukoc Biol ; 83(6): 1476-83, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18334540

RESUMEN

The Hedgehog (Hh) family of signaling molecules functions in the development of numerous tissues during embryogenesis and has also been involved in adult self-renewing tissues. Recent results have demonstrated that the different components of the Hh signaling pathway are expressed in the human thymus. In this study, we investigate whether thymic dendritic cells (DCs) are cell targets for Hh signaling. Both components of the Hh receptor, Patched and Smoothened, as well as other Hh-binding proteins with modulating functions, are expressed by human thymic DCs. The expression of Gli1, Gli2, and Gli3 transcription factors suggests that the Hh signaling pathway is active in thymic DCs, and approximately one-half of thymic DCs produces Sonic Hh (Shh). The culture of thymic DCs with Shh protects them from apoptosis [similarly to CD40 ligand (CD40L)], and these antiapoptotic effects are related to an up-regulation of Bcl-2 and Bcl-X(L) protein expression. The addition of the Hh pathway inhibitor, cyclopamine, decreases DC viability and impairs their allostimulatory function in vitro. In addition, the blockade of the Hh signaling pathway by cyclopamine treatment abrogates the up-regulation of HLA-DR, CD86, CD80, and CD83 expression induced by CD40L on thymic DCs. Finally, we also show that after activation with CD40L thymic DCs down-regulate the expression of Hh receptor components as well as Shh production. Taken together, these results suggest that the survival and function of thymic DCs are regulated by an autocrine Hh signaling.


Asunto(s)
Células Dendríticas/fisiología , Proteínas Hedgehog/fisiología , Transducción de Señal/fisiología , Timo/citología , Antígenos CD40/fisiología , Supervivencia Celular , Células Cultivadas , Humanos , Receptores Patched , Receptores de Superficie Celular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA