Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Cell Mol Life Sci ; 78(17-18): 6087-6104, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34296319

RESUMEN

Different types of multinucleated giant cells (MGCs) of myeloid origin have been described; osteoclasts are the most extensively studied because of their importance in bone homeostasis. MGCs are formed by cell-to-cell fusion, and most types have been observed in pathological conditions, especially in infectious and non-infectious chronic inflammatory contexts. The precise role of the different MGCs and the mechanisms that govern their formation remain poorly understood, likely due to their heterogeneity. First, we will introduce the main populations of MGCs derived from the monocyte/macrophage lineage. We will then discuss the known molecular actors mediating the early stages of fusion, focusing on cell-surface receptors involved in the cell-to-cell adhesion steps that ultimately lead to multinucleation. Given that cell-to-cell fusion is a complex and well-coordinated process, we will also describe what is currently known about the evolution of F-actin-based structures involved in macrophage fusion, i.e., podosomes, zipper-like structures, and tunneling nanotubes (TNT). Finally, the localization and potential role of the key fusion mediators related to the formation of these F-actin structures will be discussed. This review intends to present the current status of knowledge of the molecular and cellular mechanisms supporting multinucleation of myeloid cells, highlighting the gaps still existing, and contributing to the proposition of potential disease-specific MGC markers and/or therapeutic targets.


Asunto(s)
Adhesión Celular , Células Gigantes/metabolismo , Células Mieloides/metabolismo , Podosomas/metabolismo , Células Gigantes/citología , Humanos , Integrinas/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Células Mieloides/citología , Células Mieloides/ultraestructura , Osteoclastos/citología , Osteoclastos/metabolismo , Osteogénesis , Receptores Inmunológicos/metabolismo
2.
Int J Mol Sci ; 22(8)2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33921064

RESUMEN

Myelodysplastic syndromes (MDS) comprise a heterogeneous group of clonal hematopoietic stem (HSCs) and/or progenitor cells disorders. The established dependence of MDS progenitors on the hypoxic bone marrow (BM) microenvironment turned scientific interests to the transcription factor hypoxia-inducible factor 1 (HIF-1). HIF-1 facilitates quiescence maintenance and regulates differentiation by manipulating HSCs metabolism, being thus an appealing research target. Therefore, we examine the aberrant HIF-1 stabilization in BMs from MDS patients and controls (CTRLs). Using a nitroimidazole-indocyanine conjugate, we show that HIF-1 aberrant expression and transcription activity is oxygen independent, establishing the phenomenon of pseudohypoxia in MDS BM. Next, we examine mitochondrial quality and quantity along with levels of autophagy in the differentiating myeloid lineage isolated from fresh BM MDS and CTRL aspirates given that both phenomena are HIF-1 dependent. We show that the mitophagy of abnormal mitochondria and autophagic death are prominently featured in the MDS myeloid lineage, their severity increasing with intra-BM blast counts. Finally, we use in vitro cultured CD34+ HSCs isolated from fresh human BM aspirates to manipulate HIF-1 expression and examine its potential as a therapeutic target. We find that despite being cultured under 21% FiO2, HIF-1 remained aberrantly stable in all MDS cultures. Inhibition of the HIF-1α subunit had a variable beneficial effect in all <5%-intra-BM blasts-MDS, while it had no effect in CTRLs or in ≥5%-intra-BM blasts-MDS that uniformly died within 3 days of culture. We conclude that HIF-1 and pseudohypoxia are prominently featured in MDS pathobiology, and their manipulation has some potential in the therapeutics of benign MDS.


Asunto(s)
Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/metabolismo , Hipoxia/fisiopatología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/fisiopatología , Anciano , Anciano de 80 o más Años , Antígenos CD34/metabolismo , Autofagia/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitofagia/efectos de los fármacos , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Células Mieloides/ultraestructura , Nitroimidazoles/farmacología , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos
4.
Biochim Biophys Acta Gene Regul Mech ; 1863(11): 194628, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32979559

RESUMEN

Macrophages (MΦ) and dendritic cells (DC) play a fundamental role in shaping immune responses by sensing a plethora of Pathogen Associated Molecular Patterns (PAMPs), phagocytosis and antigen presentation to T lymphocytes. These important biological processes require efficient cell movement and an intact cellular morphology for dynamic interaction. The role of microRNAs (miRs) in this regard, however, is not well understood. In the present study, we show that miR-30b and miR-142-3p regulate migration and morphology of MΦ and DC. Transient overexpression of miR-30b and miR-142-3p attenuates migration and these cells display unique morphological deformities observed under electron microscopy. In addition, miR-142-3p overexpression in MΦ impaired phagocytosis of FITC-conjugated latex beads using live microscopy imaging. Interestingly, live cell imaging and F-actin staining revealed marked changes in the cell polarity and actin polymerization status, respectively. To identify miR-142-3p regulated pathways, we profiled global transcriptome changes in miR-142-3p or control mimic transfected DC. Expression of several genes were differentially altered by miR-142-3p and were associated with pathways related to cell movement, cell adhesion, and cytoskeletal rearrangement. Bioinformatics analysis identified a significant subset of downregulated genes with one or more predicted miR-142-3p binding sites in their 3'UTR strongly suggesting direct post-transcriptional impact of these miRNAs on multiple transcripts. Using dual luciferase assays, novel miR-142-3p binding sites were validated for three genes (Vinculin, Dab2 and Skap2) directly associated with cytoskeletal rearrangement and cell movement. In summary, our results show that miR-30b and miR-142-3p are regulators of myeloid cell cytoskeletal homeostasis and morphology.


Asunto(s)
Movimiento Celular/genética , Expresión Génica , MicroARNs/genética , Células Mieloides/inmunología , Células Mieloides/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Citoesqueleto , Regulación de la Expresión Génica , Genes Reporteros , Homeostasis , Humanos , Modelos Biológicos , Monocitos/inmunología , Monocitos/metabolismo , Células Mieloides/ultraestructura , Fagocitosis/genética , Fagocitosis/inmunología , Interferencia de ARN , Transducción de Señal , Transcriptoma
5.
Cell Rep ; 29(11): 3385-3393.e6, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31825823

RESUMEN

p21-Activated kinase 4 (PAK4), a serine/threonine kinase, is purported to localize to podosomes: transient adhesive structures that degrade the extracellular matrix to facilitate rapid myeloid cell migration. We find that treatment of transforming growth factor ß (TGF-ß)-differentiated monocytic (THP-1) cells with a PAK4-targeted inhibitor significantly reduces podosome formation and induces the formation of focal adhesions. This switch in adhesions confers a diminution of matrix degradation and reduced cell migration. Furthermore, reduced PAK4 expression causes a significant reduction in podosome number that cannot be rescued by kinase-dead PAK4, supporting a kinase-dependent role. Concomitant with PAK4 depletion, phosphorylation of Akt is perturbed, whereas a specific phospho-Akt signal is detected within the podosomes. Using superresolution analysis, we find that PAK4 specifically localizes in the podosome ring, nearer to the actin core than other ring proteins. We propose PAK4 kinase activity intersects with the Akt pathway at the podosome ring:core interface to drive regulation of macrophage podosome turnover.


Asunto(s)
Células Mieloides/metabolismo , Podosomas/metabolismo , Quinasas p21 Activadas/metabolismo , Células Cultivadas , Disulfuros/farmacología , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Células HEK293 , Humanos , Células Mieloides/efectos de los fármacos , Células Mieloides/ultraestructura , Naftoles/farmacología , Fosforilación , Podosomas/ultraestructura , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Células THP-1 , Quinasas p21 Activadas/antagonistas & inhibidores
6.
Proc Natl Acad Sci U S A ; 116(33): 16497-16506, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31346084

RESUMEN

Host inflammatory responses must be tightly regulated to ensure effective immunity while limiting tissue injury. IFN gamma (IFNγ) primes macrophages to mount robust inflammatory responses. However, IFNγ also induces cell death, and the pathways that regulate IFNγ-induced cell death are incompletely understood. Using genome-wide CRISPR/Cas9 screening, we identified autophagy genes as central mediators of myeloid cell survival during the IFNγ response. Hypersensitivity of autophagy gene-deficient cells to IFNγ was mediated by tumor necrosis factor (TNF) signaling via receptor interacting protein kinase 1 (RIPK1)- and caspase 8-mediated cell death. Mice with myeloid cell-specific autophagy gene deficiency exhibited marked hypersensitivity to fatal systemic TNF administration. This increased mortality in myeloid autophagy gene-deficient mice required the IFNγ receptor, and mortality was completely reversed by pharmacologic inhibition of RIPK1 kinase activity. These findings provide insight into the mechanism of IFNγ-induced cell death via TNF, demonstrate a critical function of autophagy genes in promoting cell viability in the presence of inflammatory cytokines, and implicate this cell survival function in protection against mortality during the systemic inflammatory response.


Asunto(s)
Autofagia/genética , Interferón gamma/toxicidad , Células Mieloides/patología , Factor de Necrosis Tumoral alfa/toxicidad , Animales , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia/metabolismo , Sistemas CRISPR-Cas/genética , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Citoprotección/efectos de los fármacos , Genoma , Ratones Noqueados , Células Mieloides/efectos de los fármacos , Células Mieloides/metabolismo , Células Mieloides/ultraestructura , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Transcriptoma/genética
7.
J Neuroinflammation ; 16(1): 9, 2019 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-30651101

RESUMEN

BACKGROUND: Phagocytosis is a key function of myeloid cells and is highly involved in brain ischemic injury. It has been scarcely studied in vivo, thus preventing a deep knowledge of the processes occurring in the ischemic environment. Structured illumination microscopy (SIM) is a superresolution technique which helps study phagocytosis, a process involving the recruitment of vesicles sized below the resolution limits of standard confocal microscopy. METHODS: Mice underwent permanent occlusion of the middle cerebral artery and were sacrificed at 48 h or 7 days after insult. Immunofluorescence for CD11b, myeloid cell membrane marker, and CD68, lysosomal marker was done in the ischemic area. Images were acquired using a SIM system and verified with SIM check. Lysosomal distribution was measured in the ischemic area by the gray level co-occurrence matrix (GLCM). SIM dataset was compared with transmission electron microscopy images of macrophages in the ischemic tissue at the same time points. Cultured microglia were stimulated with LPS to uptake 100 nm fluorescent beads and imaged by time-lapse SIM. GLCM was used to analyze bead distribution over the cytoplasm. RESULTS: SIM images reached a resolution of 130 nm and passed the quality control diagnose, ruling out possible artifacts. After ischemia, GLCM applied to the CD68 images showed that myeloid cells at 48 h had higher angular second moment (ASM), inverse difference moment (IDM), and lower entropy than myeloid cells at 7 days indicating higher lysosomal clustering at 48 h. At this time point, lysosomal clustering was proximal (< 700 nm) to the cell membrane indicating active target internalization, while at 7 days, it was perinuclear, consistent with final stages of phagocytosis or autophagy. Electron microscopy images indicated a similar pattern of lysosomal distribution thus validating the SIM dataset. GLCM on time-lapse SIM from phagocytic microglia cultures revealed a temporal decrease in ASM and IDM and increase in entropy, as beads were uptaken, indicating that GLCM informs on the progression of phagocytosis. CONCLUSIONS: GLCM analysis on SIM dataset quantitatively described different phases of macrophage phagocytic behavior revealing the dynamics of lysosomal movements in the ischemic brain indicating initial active internalization vs. final digestion/autophagy.


Asunto(s)
Encéfalo/diagnóstico por imagen , Infarto de la Arteria Cerebral Media/diagnóstico por imagen , Infarto de la Arteria Cerebral Media/patología , Células Mieloides/fisiología , Imagen Óptica/métodos , Fagocitosis/fisiología , Animales , Animales Recién Nacidos , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígeno CD11b/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Lipopolisacáridos/farmacología , Lisosomas/patología , Lisosomas/ultraestructura , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Células Mieloides/ultraestructura , Médula Espinal/citología , Factores de Tiempo
8.
Nat Neurosci ; 21(9): 1209-1217, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30150661

RESUMEN

Innate immune cells recruited to inflammatory sites have short life spans and originate from the marrow, which is distributed throughout the long and flat bones. While bone marrow production and release of leukocyte increases after stroke, it is currently unknown whether its activity rises homogeneously throughout the entire hematopoietic system. To address this question, we employed spectrally resolved in vivo cell labeling in the murine skull and tibia. We show that in murine models of stroke and aseptic meningitis, skull bone marrow-derived neutrophils are more likely to migrate to the adjacent brain tissue than cells that reside in the tibia. Confocal microscopy of the skull-dura interface revealed myeloid cell migration through microscopic vascular channels crossing the inner skull cortex. These observations point to a direct local interaction between the brain and the skull bone marrow through the meninges.


Asunto(s)
Médula Ósea/fisiología , Movimiento Celular/fisiología , Células Mieloides/fisiología , Cráneo/fisiología , Adulto , Animales , Médula Ósea/ultraestructura , Femenino , Humanos , Inflamación/patología , Masculino , Meningitis Aséptica/patología , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células Mieloides/ultraestructura , Neutrófilos , Cráneo/citología , Cráneo/ultraestructura , Accidente Cerebrovascular/patología , Tibia/fisiología , Tibia/ultraestructura , Tomografía Computarizada por Rayos X
9.
J Neuroinflammation ; 13(1): 196, 2016 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-27562117

RESUMEN

BACKGROUND: Alphaviruses can cause fatal encephalitis in humans. Natural infections occur via the bite of infected mosquitos, but aerosol transmissibility makes some of these viruses potential bioterrorism agents. Central nervous system (CNS) host responses contribute to alphavirus pathogenesis in experimental models and are logical therapeutic targets. We investigated whether reactive oxygen species (ROS) generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) activity within the CNS contributes to fatal alphavirus encephalitis in mice. METHODS: Infected animals were treated systemically with the angiotensin receptor-blocking drug, telmisartan, given its ability to cross the blood-brain barrier, selectively block type-1 angiotensin receptors (AT1R), and inhibit Nox-derived ROS production in vascular smooth muscle and other extraneural tissues. Clinical, virological, biochemical, and histopathological outcomes were followed over time. RESULTS: The importance of the angiotensin II (Ang II)/AT1R axis in disease pathogenesis was confirmed by demonstrating increased Ang II levels in the CNS following infection, enhanced disease survival when CNS Ang II production was suppressed, increased AT1R expression on microglia and tissue-infiltrating myeloid cells, and enhanced disease survival in AT1R-deficient mice compared to wild-type (WT) controls. Systemic administration of telmisartan protected WT mice from lethal encephalitis caused by two different alphaviruses in a dose-dependent manner without altering virus replication or exerting any anti-inflammatory effects in the CNS. Infection triggered up-regulation of multiple Nox subunits in the CNS, while drug treatment inhibited local Nox activity, ROS production, and oxidative neuronal damage. Telmisartan proved ineffective in Nox-deficient mice, demonstrating that this enzyme is its main target in this experimental setting. CONCLUSIONS: Nox-derived ROS, likely arising from CNS myeloid cells triggered by AT1R signaling, are pathogenic during fatal alphavirus encephalitis in mice. Systemically administered telmisartan at non-hypotensive doses targets Nox activity in the CNS to exert a neuroprotective effect. Disruption of this pathway may have broader implications for the treatment of related infections as well as for other CNS diseases driven by oxidative injury.


Asunto(s)
Sistema Nervioso Central/patología , Encefalomielitis Equina/patología , Células Mieloides/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal/fisiología , ATPasas Asociadas con Actividades Celulares Diversas , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Animales , Bencimidazoles/farmacología , Benzoatos/farmacología , Receptor 1 de Quimiocinas CX3C , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/virología , ADN Helicasas/genética , ADN Helicasas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Equina/tratamiento farmacológico , Encefalomielitis Equina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Mieloides/clasificación , Células Mieloides/ultraestructura , Células Mieloides/virología , Neuronas/patología , Neuronas/ultraestructura , Consumo de Oxígeno/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Telmisartán
10.
J Neuroinflammation ; 12: 161, 2015 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-26338007

RESUMEN

BACKGROUND: The NG2 proteoglycan is expressed by several cell types in demyelinated lesions and has important effects on the biology of these cells. Here we determine the cell-type-specific roles of NG2 in the oligodendrocyte progenitor cell (OPC) and myeloid cell contributions to demyelination and remyelination. METHODS: We have used Cre-Lox technology to dissect the cell-type-specific contributions of NG2 to myelin damage and repair. Demyelination is induced by microinjection of 1 % lysolecithin into the spinal cord white matter of control, OPC-specific NG2-null (OPC-NG2ko), and myeloid-specific NG2-null (My-NG2ko) mice. The status of OPCs, myeloid cells, axons, and myelin is assessed by light, immunofluorescence, confocal, and electron microscopy. RESULTS: In OPC-NG2ko mice 1 week after lysolecithin injection, the OPC mitotic index is reduced by 40 %, resulting in 25 % fewer OPCs at 1 week and a 28 % decrease in mature oligodendrocytes at 6 weeks post-injury. The initial demyelinated lesion size is not affected in OPC-NG2ko mice, but lesion repair is delayed by reduced production of oligodendrocytes. In contrast, both the initial extent of demyelination and the kinetics of lesion repair are decreased in My-NG2ko mice. Surprisingly, the OPC mitotic index at 1 week post-injury is also reduced (by 48 %) in My-NG2ko mice, leading to a 35 % decrease in OPCs at 1 week and a subsequent 34 % reduction in mature oligodendrocytes at 6 weeks post-injury. Clearance of myelin debris is also reduced by 40 % in My-NG2ko mice. Deficits in myelination detected by immunostaining for myelin basic protein are confirmed by toluidine blue staining and by electron microscopy. In addition to reduced myelin repair, fewer axons are found in 6-week lesions in both OPC-NG2ko and My-NG2ko mice, emphasizing the importance of myelination for neuron survival. CONCLUSIONS: Reduced generation of OPCs and oligodendrocytes in OPC-NG2ko mice correlates with reduced myelin repair. Diminished demyelination in My-NG2ko mice may stem from a reduction (approximately 70 %) in myeloid cell recruitment to lesions. Reduced macrophage/microglia numbers may then result in decreased myelin repair via diminished clearance of myelin debris and reduced stimulatory effects on OPCs.


Asunto(s)
Antígenos/metabolismo , Enfermedades Autoinmunes Desmielinizantes SNC/patología , Células Mieloides/patología , Oligodendroglía/patología , Proteoglicanos/metabolismo , Recuperación de la Función/fisiología , Médula Espinal/patología , Animales , Antígenos/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Trasplante de Médula Ósea , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/fisiología , Enfermedades Autoinmunes Desmielinizantes SNC/inducido químicamente , Enfermedades Autoinmunes Desmielinizantes SNC/cirugía , Modelos Animales de Enfermedad , Lisofosfatidilcolinas/toxicidad , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Vaina de Mielina/ultraestructura , Células Mieloides/ultraestructura , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Fagocitosis/fisiología , Proteoglicanos/genética , Células Madre/metabolismo , Células Madre/ultraestructura
11.
J Leukoc Biol ; 97(4): 737-49, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25673294

RESUMEN

DCs are the first immune cells to be exposed to allergens, including chemical sensitizers, such as nickel, a human TLR4 agonist that induces DC maturation. In ACD, DCs can interact with PMNs that are recruited and activated, leading, in particular, to ectosome release. The objective of this work was to characterize the effects of PMN-Ect on DC functions in an ACD context. We first developed a standardized protocol to produce, characterize, and quantify ectosomes by use of human PLB-985 cells, differentiated into mature PMN (PLB-Ect). We then studied the in vitro effects of these purified ectosomes on human moDC functions in response to NiSO4 and to LPS, another TLR4 agonist. Confocal fluorescence microscopy showed that PLB-Ect was internalized by moDCs and localized in the lysosomal compartment. We then showed that PLB-Ect down-regulated NiSO4-induced moDC maturation, as witnessed by decreased expression of CD40, CD80, CD83, CD86, PDL-1, and HLA-DR and by decreased levels of IL-1ß, IL-6, TNF-α, and IL-12p40 mRNAs. These effects were related to p38MAPK and NF-κB down-regulation. However, no increase in pan-regulatory DC marker genes (GILZ, CATC, C1QA) was observed; rather, levels of effector DC markers (Mx1, NMES1) were increased. Finally, when these PLB-Ect + NiSO4-treated moDCs were cocultured with CD4(+) T cells, a Th2 cytokine profile seemed to be induced, as shown, in particular, by enhanced IL-13 production. Together, these results suggest that the PMN-Ect can modulate DC maturation in response to nickel, a common chemical sensitizer responsible for ADC.


Asunto(s)
Alérgenos/inmunología , Antígenos CD/biosíntesis , Micropartículas Derivadas de Células/fisiología , Células Dendríticas/inmunología , Dermatitis Alérgica por Contacto/inmunología , Regulación de la Expresión Génica/inmunología , Linfocinas/biosíntesis , Células Mieloides/inmunología , Neutrófilos/inmunología , Níquel/inmunología , Células Th2/citología , Alérgenos/farmacología , Antígenos CD/genética , Antígeno B7-H1/biosíntesis , Antígeno B7-H1/genética , Diferenciación Celular , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Dermatitis Alérgica por Contacto/etiología , Antígenos HLA-DR/biosíntesis , Antígenos HLA-DR/genética , Humanos , Leucemia Mieloide Aguda/patología , Liposomas , Linfocinas/genética , Monocitos/citología , Células Mieloides/ultraestructura , Neutrófilos/ultraestructura , Níquel/farmacología , Receptor Toll-Like 4/agonistas , Receptor Toll-Like 4/inmunología
12.
J Cutan Pathol ; 41(2): 123-33, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24117956

RESUMEN

Clear cell tumors of the skin are observed in a wide variety of benign and malignant conditions with different histogenesis, sharing the presence of cells with abundant clear cytoplasm. Herein, we report the clinicopathologic features of a healthy young patient affected by asymptomatic, eruptive and disseminated, benign clear cell dermal tumors since early infancy. Neither family history nor genetic testing and counseling provided further useful information. The lesions were mostly confined to the face and lower left extremity with pink teleangiectatic papules and small nodules. Over a 4-year period, a total of 16 different cutaneous lesions were biopsied and histopathologic and immunohistochemical studies carried out; an additional lesion was also removed for electron microscopy examination. Histopathology evidenced multiple perivascular growths of spindle to oval and round cells intermingled with clear/granular cells throughout the dermis, with prominent desmoplasia and numerous capillary-like vessels with focal hemangiopericytoma-like features. Immunohistochemical neoplastic cells were uniformly positive for h-caldesmon and focally smooth muscle α-actin and CD13 indicating myoid differentiation whereas the consistent diffuse cytoplasmic staining for lysosome antigen, such as CD68PG-M1 and NKI/C3 along with the ultrastructural findings supported the view of a lysosome-mediated apoptotic process. The differential diagnosis with other clear cell cutaneous neoplasms is discussed.


Asunto(s)
Apoptosis , Diferenciación Celular , Lisosomas , Células Mieloides , Neoplasias Cutáneas , Actinas , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos CD13/metabolismo , Niño , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Lisosomas/metabolismo , Lisosomas/ultraestructura , Masculino , Microscopía Electrónica de Transmisión , Células Mieloides/metabolismo , Células Mieloides/ultraestructura , Proteínas de Neoplasias/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/ultraestructura
13.
Nat Methods ; 10(8): 795-803, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23749299

RESUMEN

Investigation of immune-cell differentiation and function is limited by shortcomings of suitable and scalable experimental systems. Here we show that retroviral delivery of an estrogen-regulated form of Hoxb8 into mouse bone marrow cells can be used along with Flt3 ligand to conditionally immortalize early hematopoietic progenitor cells (Hoxb8-FL cells). Hoxb8-FL cells have lost self-renewal capacity and potential to differentiate into megakaryocytes and erythrocytes but retain the potential to differentiate into myeloid and lymphoid cells. They differentiate in vitro and in vivo into macrophages, granulocytes, dendritic cells, B lymphocytes and T lymphocytes that are phenotypically and functionally indistinguishable from their primary counterparts. Quantitative in vitro assays indicate that myeloid and B-cell potential of Hoxb8-FL cells is comparable to that of primary lymphoid-primed multipotent progenitors, whereas T-cell potential is diminished. The simplicity of this system and the unlimited proliferative capacity of Hoxb8-FL cells will enable studies of immune-cell differentiation and function.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Hematopoyéticas/citología , Proteínas de Homeodominio/metabolismo , Linfocitos/citología , Células Mieloides/citología , Animales , Diferenciación Celular/fisiología , Linaje de la Célula , Femenino , Citometría de Flujo , Linfocitos/ultraestructura , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Mieloides/ultraestructura , Análisis de Componente Principal , Proteómica
14.
Infect Immun ; 79(11): 4696-707, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21844238

RESUMEN

Anaplasma phagocytophilum is an obligate intracellular bacterium that invades neutrophils to cause the emerging infectious disease human granulocytic anaplasmosis. A. phagocytophilum undergoes a biphasic developmental cycle, transitioning between an infectious dense-cored cell (DC) and a noninfectious reticulate cell (RC). To gain insights into the organism's biology and pathogenesis during human myeloid cell infection, we conducted proteomic analyses on A. phagocytophilum organisms purified from HL-60 cells. A total of 324 proteins were unambiguously identified, thereby verifying 23.7% of the predicted A. phagocytophilum proteome. Fifty-three identified proteins had been previously annotated as hypothetical or conserved hypothetical. The second most abundant gene product, after the well-studied major surface protein 2 (P44), was the hitherto hypothetical protein APH_1235. APH_1235 homologs are found in other Anaplasma and Ehrlichia species but not in other bacteria. The aph_1235 RNA level is increased 70-fold in the DC form relative to that in the RC form. Transcriptional upregulation of and our ability to detect APH_1235 correlate with RC to DC transition, DC exit from host cells, and subsequent DC binding and entry during the next round of infection. Immunoelectron microscopy pronouncedly detects APH_1235 on DC organisms, while detection on RC bacteria minimally, at best, exceeds background. This work represents an extensive study of the A. phagocytophilum proteome, discerns the complement of proteins that is generated during survival within human myeloid cells, and identifies APH_1235 as the first known protein that is pronouncedly upregulated on the infectious DC form.


Asunto(s)
Anaplasma phagocytophilum/metabolismo , Proteínas Bacterianas/metabolismo , Perfilación de la Expresión Génica , Regulación Bacteriana de la Expresión Génica/fisiología , Células Mieloides/microbiología , Secuencia de Aminoácidos , Anaplasma phagocytophilum/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Centrifugación por Gradiente de Densidad , Cromatografía Liquida , Ehrlichia/genética , Ehrlichia/metabolismo , Células HL-60 , Humanos , Anotación de Secuencia Molecular , Células Mieloides/ultraestructura , Proteómica , Especificidad de la Especie , Espectrometría de Masas en Tándem , Regulación hacia Arriba
15.
J Biotechnol ; 152(1-2): 43-8, 2011 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-21262283

RESUMEN

Growing tumours have acquired several mechanisms to resist to immune recognition. Among these strategies, myeloid-derived suppressor cells (MDSCs) contribute to tumour escape by suppressing T-cell specific anti-tumoural functions. The development of therapies that could specifically inhibit MDSC maturation, recruitment, accumulation and immunosuppressive functions is thus of great interest. This requires the identification of valuable biomarkers of MDSC behaviour in vitro. As for immune cells, whose energetic state is known as a biomarker of their functionality, we have characterized in vitro the metabolic and energetic behaviour of MSC-1 cells, an immortalized cell line derived from mouse MDSCs and used as model cell line. Combined results from in vitro(31)P-NMR with living cells and HPLC-MS analyses from cell extracts allowed to identify two distinct bioenergetic steady-states that coincided with exponential and stationary growth phases. While the adenylate energy charge remained constant throughout the culture duration, both the percentage of total pyrimidines, the UTP-to-ATP and PME (phosphomonoesters)-to-NTP ratios were higher at the exponential growth phase compared to the plateau phase, suggesting metabolically active cells and the production of growth-related molecules. Conversely, the NTP ratio increased at the entry of the stationary phase revealing the deterioration of the global bioenergetic status and the arrest of anabolic processes.


Asunto(s)
Células Mieloides/citología , Células Mieloides/metabolismo , Animales , Reactores Biológicos , Línea Celular , Proliferación Celular , Cromatografía Líquida de Alta Presión , Metabolismo Energético , Humanos , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Ratones , Microscopía Electrónica de Rastreo , Células Mieloides/ultraestructura , Óxidos de Nitrógeno/metabolismo , Fosfolípidos/metabolismo
16.
J Nucl Med ; 51(5): 782-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20395332

RESUMEN

UNLABELLED: Nanobodies are single-domain antigen-binding fragments derived from heavy-chain antibodies that are devoid of light chains and occur naturally in Camelidae. We have shown before that their small size and high affinity and specificity for their target antigen make Nanobodies ideal probes for in vivo tumor imaging. In the present study, we have evaluated the use of Nanobodies as a generic method for imaging the in vivo biodistribution of specific immune cell types, using myeloid cells as an example. METHODS: The cellular specificity of Nanobodies raised against murine bone marrow-derived dendritic cells was verified using flow cytometry on a range of myeloid and nonmyeloid cell types. The Nanobodies were then labeled with (99m)Tc and their biodistribution was analyzed using SPECT. The biodistribution was also assessed by measuring radioactivity in various organs and tissues. To verify whether the observed biodistribution was due to specific targeting through the antigen-binding loops, rather than retention in organs because of effects of the framework regions, we genetically grafted the antigen-binding loops of the Nanobodies onto the framework region of a Nanobody scaffold that by itself showed low background retention in the periphery. The cellular specificity and biodistribution of these grafted Nanobodies were determined as before. RESULTS: Nb-DC2.1, which recognizes a wide range of myeloid cells, targets most strongly to the liver, spleen, and lungs. Nb-DC1.8, which recognizes immature bone marrow-derived dendritic cells in vitro, gives a much smaller signal in the liver and spleen than does Nb-DC2.1 but mainly targets to the lungs and gives a pronounced signal in the skin. Grafting of the antigen-binding loops of Nb-DC1.8 or Nb-DC2.1 to the scaffold of Nb-BCII10 alters the observed biodistribution of the Nanobodies to resemble that of the Nanobody from which the antigen-binding loops have been derived. CONCLUSION: The observed in vivo biodistribution of the Nanobodies reflects the main in vivo locations of the cells recognized by the Nanobodies and is determined by the antigen-binding loops of the Nanobodies. Thus, Nanobodies represent elegant targeting probes for imaging the in vivo biodistribution of specific immune cell types.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/ultraestructura , Células Mieloides/inmunología , Células Mieloides/ultraestructura , Nanopartículas , Secuencia de Aminoácidos , Animales , Médula Ósea/diagnóstico por imagen , Escherichia coli/metabolismo , Femenino , Citometría de Flujo , Colorantes Fluorescentes , Fluorobencenos , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Distribución Tisular , Tomografía Computarizada de Emisión , Tomografía Computarizada de Emisión de Fotón Único
17.
Folia Morphol (Warsz) ; 69(1): 1-14, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20235044

RESUMEN

Mammary gland epithelium is composed of an inner layer of secretory cells (luminal) and an outer layer of myoepithelial cells (MEC) bordering the basal lamina which separates the epithelial layer from the extracellular matrix. Mature MECs morphologically resemble smooth muscle cells; however, they exhibit features typical for epithelial cells, such as the presence of specific cytokeratin filaments. During lactation, secretory cells synthesize milk components, which are collected in alveoli and duct lumen, and transported to the nipple as a result of MEC contraction. Although the induction of MEC contraction results from oxytocin action, also other, still unknown auto/paracrine mechanisms participate in the regulation of this process. As well as milk ejection, MECs are involved in mammary gland morphogenesis in all developmental stages, modulating proliferation and differentiation of luminal cells. They take part in the formation of extracellular matrix, synthesizing its components and secreting proteinases and their inhibitors. In addition, MECs are regarded as natural cancer suppressors, stabilizing the normal structure of the mammary gland, they secrete suppressor proteins (e.g. maspin) limiting cancer growth, invasiveness, and neoangiogenesis. The majority of malignant breast cancers are derived from luminal cells, whereas neoplasms of MEC origin are the most seldom and usually benign form of breast tumours. MECs are markedly resistant to malignant transformation and they are able to suppress the transformation of neighboring luminal cells. Therefore, a deeper insight into the role of MECs in the physiology and pathology of mammary glands would allow a better understanding of cancerogenesis mechanisms and possible application of specific MEC markers in the diagnosis and therapy of breast cancer.


Asunto(s)
Neoplasias de la Mama , Glándulas Mamarias Animales , Glándulas Mamarias Humanas , Neoplasias Mamarias Animales , Células Mieloides , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/ultraestructura , Femenino , Humanos , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/ultraestructura , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/ultraestructura , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/ultraestructura , Células Mieloides/metabolismo , Células Mieloides/ultraestructura
19.
Cell ; 129(7): 1287-98, 2007 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-17604718

RESUMEN

M. tuberculosis and M. leprae are considered to be prototypical intracellular pathogens that have evolved strategies to enable growth in the intracellular phagosomes. In contrast, we show that lysosomes rapidly fuse with the virulent M. tuberculosis- and M. leprae-containing phagosomes of human monocyte-derived dendritic cells and macrophages. After 2 days, M. tuberculosis progressively translocates from phagolysosomes into the cytosol in nonapoptotic cells. Cytosolic entry is also observed for M. leprae but not for vaccine strains such as M. bovis BCG or in heat-killed mycobacteria and is dependent upon secretion of the mycobacterial gene products CFP-10 and ESAT-6. The cytosolic bacterial localization and replication are pathogenic features of virulent mycobacteria, causing significant cell death within a week. This may also reveal a mechanism for MHC-based antigen presentation that is lacking in current vaccine strains.


Asunto(s)
Citosol/fisiología , Lisosomas/fisiología , Mycobacterium/fisiología , Células Mieloides/microbiología , Fagosomas/fisiología , Presentación de Antígeno/fisiología , Biomarcadores/metabolismo , Compartimento Celular/fisiología , Muerte Celular/fisiología , División Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Células Cultivadas , Citosol/ultraestructura , Regulación Bacteriana de la Expresión Génica/fisiología , Interacciones Huésped-Parásitos/fisiología , Humanos , Inmunohistoquímica , Membranas Intracelulares/fisiología , Membranas Intracelulares/ultraestructura , Lisosomas/ultraestructura , Proteínas de la Membrana/metabolismo , Microscopía Electrónica de Transmisión , Mycobacterium/genética , Mycobacterium/ultraestructura , Mycobacterium leprae/genética , Mycobacterium leprae/fisiología , Mycobacterium leprae/ultraestructura , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/fisiología , Mycobacterium tuberculosis/ultraestructura , Células Mieloides/fisiología , Células Mieloides/ultraestructura , Fagosomas/ultraestructura
20.
Clin Chim Acta ; 371(1-2): 32-6, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16777084

RESUMEN

Advanced glycation endproducts (AGEs) are chemical modifications of proteins by carbohydrates including those metabolic intermediates formed during the Maillard reaction. The generation of AGEs is an inevitable process in vivo. AGEs constitute a heterogeneous class of compounds characterized by brown color, fluorescence and a tendency to polymerize. These unique compounds are specifically recognized by AGE receptors (RAGE) present on different cell types. A remarkable feature of AGE-mediated cross-linked proteins is decreased solubility and resistance to proteolytic digestion. This effect results in altered biomechanical properties in affected tissues including increased stiffness and rigidity. The AGE-RAGE interaction additionally induces activation of nuclear factor kB (NF-kB) in RAGE bearing cells (e.g., cells participating in bone turnover). This interaction results e.g. in increased expression of cytokines, growth factors and adhesion molecules. Recent findings provide important evidence that bone proteins are also affected by AGE modification. Investigations conducted by other groups, as well as ours, support the hypothesis that bone protein glycation influences osteoclasts (bone resorption) and osteoblasts (bone formation).


Asunto(s)
Productos Finales de Glicación Avanzada/metabolismo , Osteoporosis/metabolismo , Remodelación Ósea/fisiología , Resorción Ósea/fisiopatología , Humanos , Células Mieloides/fisiología , Células Mieloides/ultraestructura , Osteoblastos/patología , Osteoblastos/fisiología , Osteoclastos/patología , Osteoclastos/fisiología , Osteoporosis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA