Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cells ; 12(21)2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37947594

RESUMO

Neuroblastoma (NB) is the most common extracranial solid tumor during infancy, causing up to 10% of mortality in children; thus, identifying novel early and accurate diagnostic and prognostic biomarkers is mandatory. NB-derived exosomes carry proteins (Exo-prots) reflecting the status of the tumor cell of origin. The purpose of this study was to characterize, for the first time, the Exo-prots specifically expressed in NB patients associated with tumor phenotype and disease stage. We isolated exosomes from plasma specimens of 24 HR-NB patients and 24 low-risk (LR-NB) patients at diagnosis and of 24 age-matched healthy controls (CTRL). Exo-prot expression was measured by liquid chromatography-mass spectrometry. The data are available via ProteomeXchange (PXD042422). The NB patients had a different Exo-prot expression profile compared to the CTRL. The deregulated Exo-prots in the NB specimens acted mainly in the tumor-associated pathways. The HR-NB patients showed a different Exo-prot expression profile compared to the LR-NB patients, with the modulation of proteins involved in cell migration, proliferation and metastasis. NCAM, NCL, LUM and VASP demonstrated a diagnostic value in discriminating the NB patients from the CTRL; meanwhile, MYH9, FN1, CALR, AKAP12 and LTBP1 were able to differentiate between the HR-NB and LR-NB patients with high accuracy. Therefore, Exo-prots contribute to NB tumor development and to the aggressive metastatic NB phenotype.


Assuntos
Exossomos , Neuroblastoma , Criança , Humanos , Exossomos/metabolismo , Prognóstico , Neuroblastoma/genética , Fenótipo , Biomarcadores/metabolismo
2.
Redox Biol ; 36: 101618, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32863220

RESUMO

Fanconi Anemia (FA) is a disease characterized by bone marrow (BM) failure and aplastic anemia. In addition to a defective DNA repair system, other mechanisms are involved in its pathogenesis, such as defective mitochondrial metabolism, accumulation of lipids, and increment of oxidative stress production. To better understand the role of these metabolic alterations in the process of HSC maturation in FA, we evaluated several biochemical and cellular parameters on mononuclear cells isolated from the bone marrow of FA patients or healthy donors. To mimic the cellular residence in the BM niche or their exit from the BM niche to the bloodstream, cells have been grown in hypoxic or normoxic conditions, respectively. The data show that, in normoxic conditions, a switch from anaerobic to aerobic metabolism occurs both in healthy and in pathological samples. However, in FA cells this change is associated with altered oxidative phosphorylation, the increment of oxidative stress production, no activation of the endogenous antioxidant defenses and arrest in the G2M phase of the cell cycle. By contrast, FA cells grown in hypoxic conditions do not show cell cycle and metabolic alterations in comparison to the healthy control, maintaining both an anaerobic flux. The data reported herein suggests that the passage from the BM niche to the bloodstream represents a crucial point in the FA pathogenesis associated with mitochondrial dysfunction.


Assuntos
Anemia de Fanconi , Medula Óssea/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Humanos , Mitocôndrias/metabolismo , Fosforilação Oxidativa , Estresse Oxidativo
3.
Cancers (Basel) ; 12(5)2020 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-32456204

RESUMO

Inflammatory cells are major players in the onset of cancer. The degree of inflammation and type of inflammatory cells in the tumor microenvironment (TME) are responsible for tilting the balance between tumor progression and regression. Cancer-related inflammation has also been shown to influence the efficacy of conventional therapy. Mononuclear phagocytes (MPs) represent a major component of the inflammatory circuit that promotes tumor progression. Despite their potential to activate immunosurveillance and exert anti-tumor responses, MPs are subverted by the tumor to support its growth, immune evasion, and spread. MP responses in the TME are dictated by a network of stimuli integrated through the cross-talk between activatory and inhibitory receptors. Alterations in receptor expression/signaling can create excessive inflammation and, when chronic, promote tumorigenesis. Research advances have led to the development of new therapeutic strategies aimed at receptor targeting to induce a tumor-infiltrating MP switch from a cancer-supportive toward an anti-tumor phenotype, demonstrating efficacy in different human cancers. This review provides an overview of the role of MP receptors in inflammation-mediated carcinogenesis and discusses the most recent updates regarding their targeting for immunotherapeutic purposes. We focus in particular on the TREM-1 receptor, a major amplifier of MP inflammatory responses, highlighting its relevance in the development and progression of several types of inflammation-associated malignancies and the promises of its inhibition for cancer immunotherapy.

4.
J Transl Med ; 18(1): 21, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31924244

RESUMO

BACKGROUND: Tetralogy of Fallot (ToF) and Atrial Septal Defects (ASD) are the most common types of congenital heart diseases and a major cause of childhood morbidity and mortality. Cardiopulmonary bypass (CPB) is used during corrective cardiac surgery to support circulation and heart stabilization. However, this procedure triggers systemic inflammatory and stress response and consequent increased risk of postoperative complications. The aim of this study was to define the molecular bases of ToF and ASD pathogenesis and response to CPB and identify new potential biomarkers. METHODS: Comparative transcriptome analysis of right atrium specimens collected from 10 ToF and 10 ASD patients was conducted before (Pre-CPB) and after (Post-CPB) corrective surgery. Total RNA isolated from each sample was individually hybridized on Affymetrix HG-U133 Plus Array Strips containing 38,500 unique human genes. Differences in the gene expression profiles and functional enrichment/network analyses were assessed using bioinformatic tools. qRT-PCR analysis was used to validate gene modulation. RESULTS: Pre-CPB samples showed significant differential expression of a total of 72 genes, 28 of which were overexpressed in ToF and 44 in ASD. According to Gene Ontology annotation, the mostly enriched biological processes were represented by matrix organization and cell adhesion in ToF and by muscle development and contractility in ASD specimens. GSEA highlighted the specific enrichment of hypoxia gene sets in ToF samples, pointing to a role for hypoxia in disease pathogenesis. The post-CPB myocardium exhibited significant alterations in the expression profile of genes related to transcription regulation, growth/apoptosis, inflammation, adhesion/matrix organization, and oxidative stress. Among them, only 70 were common to the two disease groups, whereas 110 and 24 were unique in ToF and ASD, respectively. Multiple functional interactions among differentially expressed gene products were predicted by network analysis. Interestingly, gene expression changes in ASD samples followed a consensus hypoxia profile. CONCLUSION: Our results provide a comprehensive view of gene reprogramming in right atrium tissues of ToF and ASD patients before and after CPB, defining specific molecular pathways underlying disease pathophysiology and myocardium response to CPB. These findings have potential translational value because they identify new candidate prognostic markers and targets for tailored cardioprotective post-surgical therapies.


Assuntos
Comunicação Interatrial , Miocárdio , Tetralogia de Fallot , Ponte Cardiopulmonar , Criança , Perfilação da Expressão Gênica , Comunicação Interatrial/genética , Humanos , Miocárdio/metabolismo
5.
Cancers (Basel) ; 11(10)2019 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-31575060

RESUMO

Despite intensive treatment, 50% of children with high-risk neuroblastoma (HR-NB) succumb to their disease. Progression through current trials evaluating the efficacy of new treatments for children with HR disease usually depends on an inadequate response to induction chemotherapy, assessed using imaging modalities. In this study, we sought to identify circulating biomarkers that might be detected in a simple blood sample to predict patient response to induction chemotherapy. Since exosomes released by tumor cells can drive tumor growth and chemoresistance, we tested the hypothesis that exosomal microRNA (exo-miRNAs) in blood might predict response to induction chemotherapy. The exo-miRNAs expression profile in plasma samples collected from children treated in HR-NBL-1/SIOPEN before and after induction chemotherapy was compared to identify a three exo-miRs signature that could discriminate between poor and good responders. Exo-miRNAs expression also provided a chemoresistance index predicting the good or poor prognosis of HR-NB patients.

6.
J Proteome Res ; 18(7): 2965-2978, 2019 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-31173686

RESUMO

Glycogen storage disease type 1a (GSD-1a) is a rare genetic disease caused by mutations in the catalytic subunit of the enzyme glucose-6-phosphatase-alpha (G6Pase-α). The majority of patients develop long-term complications including renal failure and hepatocellular adenoma/carcinoma. The purpose of this study was to ascertain the proteomic changes in the liver of LS- G6pc-/- mice, a murine model of GSD-1a, in comparison with wild type mice to identify potential biomarkers of the pathophysiology of the affected liver. We used liquid chromatography-tandem mass spectrometry (LC-MS/MS) to analyze liver lysates from a total of 20 LS- G6pc-/- and 18 wild type (WT) mice. We compared the proteomic expression profile of LS- G6pc-/- and WT mice. We identified 4138 significantly expressed proteins, 1243 of which were differentially represented. Network and pathway analyses indicate that LS- G6pc-/- livers display an age-dependent modulation of the expression of proteins involved in specific biological processes associated with increased progression of liver disease. Moreover, we found upregulation of proteins involved in the process of tissue inflammation and macrophage polarization toward the M2 phenotype in LS- G6pc-/- mice with adenomas. Our results identify a metabolic reprogramming of glucose-6-P and a pathologic environment in the liver compatible with tumor development and progression.


Assuntos
Doença de Depósito de Glicogênio Tipo I/metabolismo , Fígado/química , Proteômica , Animais , Cromatografia Líquida , Modelos Animais de Doenças , Glucose-6-Fosfatase/genética , Doença de Depósito de Glicogênio Tipo I/patologia , Inflamação , Fígado/metabolismo , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Proteínas/análise , Espectrometria de Massas em Tandem
7.
Front Immunol ; 9: 2358, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30459756

RESUMO

Hypoxia, which characterizes most tumor tissues, can alter the function of different immune cell types, favoring tumor escape mechanisms. In this study, we show that hypoxia profoundly acts on NK cells by influencing their transcriptome, affecting their immunoregulatory functions, and changing the chemotactic responses of different NK cell subsets. Exposure of human peripheral blood NK cells to hypoxia for 16 or 96 h caused significant changes in the expression of 729 or 1,100 genes, respectively. Gene Set Enrichment Analysis demonstrated that these changes followed a consensus hypoxia transcriptional profile. As assessed by Gene Ontology annotation, hypoxia-targeted genes were implicated in several biological processes: metabolism, cell cycle, differentiation, apoptosis, cell stress, and cytoskeleton organization. The hypoxic transcriptome also showed changes in genes with immunological relevance including those coding for proinflammatory cytokines, chemokines, and chemokine-receptors. Quantitative RT-PCR analysis confirmed the modulation of several immune-related genes, prompting further immunophenotypic and functional studies. Multiplex ELISA demonstrated that hypoxia could variably reduce NK cell ability to release IFNγ, TNFα, GM-CSF, CCL3, and CCL5 following PMA+Ionomycin or IL15+IL18 stimulation, while it poorly affected the response to IL12+IL18. Cytofluorimetric analysis showed that hypoxia could influence NK chemokine receptor pattern by sustaining the expression of CCR7 and CXCR4. Remarkably, this effect occurred selectively (CCR7) or preferentially (CXCR4) on CD56bright NK cells, which indeed showed higher chemotaxis to CCL19, CCL21, or CXCL12. Collectively, our data suggest that the hypoxic environment may profoundly influence the nature of the NK cell infiltrate and its effects on immune-mediated responses within tumor tissues.


Assuntos
Hipóxia/genética , Hipóxia/metabolismo , Imunomodulação/genética , Células Matadoras Naturais/metabolismo , Transcriptoma , Diferenciação Celular , Movimento Celular/genética , Quimiotaxia/genética , Quimiotaxia/imunologia , Citocinas/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia
8.
J Inherit Metab Dis ; 41(6): 1015-1025, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29967951

RESUMO

BACKGROUND AND AIMS: Glycogen storage disease type Ib (GSD1b) is a rare metabolic and immune disorder caused by a deficiency in the glucose-6-phosphate transporter (G6PT) and characterized by impaired glucose homeostasis, myeloid dysfunction, and long-term risk of hepatocellular adenomas. Despite maximal therapy, based on a strict diet and on granulocyte colony-stimulating factor treatment, long-term severe complications still develop. Understanding the pathophysiology of GSD1b is a prerequisite to develop new therapeutic strategies and depends on the availability of animal models. The G6PT-KO mouse mimics the human disease but is very fragile and rarely survives weaning. We generated a conditional G6PT-deficient mouse as an alternative model for studying the long-term pathophysiology of the disease. We utilized this conditional mouse to develop an inducible G6PT-KO model to allow temporally regulated G6PT deletion by the administration of tamoxifen (TM). METHODS: We generated a conditional G6PT-deficient mouse utilizing the CRElox strategy. Histology, histochemistry, and phenotype analyses were performed at different times after TM-induced G6PT inactivation. Neutrophils and monocytes were isolated and analyzed for functional activity with standard techniques. RESULTS: The G6PT-inducible KO mice display the expected disturbances of G6P metabolism and myeloid dysfunctions of the human disorder, even though with a milder intensity. CONCLUSIONS: TM-induced inactivation of G6PT in these mice leads to a phenotype which mimics that of human GSD1b patients. The conditional mice we have generated represent an excellent tool to study the tissue-specific role of the G6PT gene and the mechanism of long-term complications in GSD1b.


Assuntos
Antiporters/deficiência , Modelos Animais de Doenças , Glucose/metabolismo , Doença de Depósito de Glicogênio Tipo I/genética , Homeostase , Proteínas de Transporte de Monossacarídeos/deficiência , Animais , Antiporters/genética , Doença de Depósito de Glicogênio Tipo I/etiologia , Doença de Depósito de Glicogênio Tipo I/patologia , Camundongos , Camundongos Knockout , Proteínas de Transporte de Monossacarídeos/genética , Neutropenia/etiologia , Tamoxifeno/administração & dosagem
9.
Front Immunol ; 8: 1097, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28936211

RESUMO

Macrophages (Mf) are a heterogeneous population of tissue-resident professional phagocytes and a major component of the leukocyte infiltrate at sites of inflammation, infection, and tumor growth. They can undergo diverse forms of activation in response to environmental factors, polarizing into specialized functional subsets. A common hallmark of the pathologic environment is represented by hypoxia. The impact of hypoxia on human Mf polarization has not been fully established. The objective of this study was to elucidate the effects of a hypoxic environment reflecting that occurring in vivo in diseased tissues on the ability of human Mf to polarize into classically activated (proinflammatory M1) and alternatively activated (anti-inflammatory M2) subsets. We present data showing that hypoxia hinders Mf polarization toward the M1 phenotype by decreasing the expression of T cell costimulatory molecules and chemokine homing receptors and the production of proinflammatory, Th1-priming cytokines typical of classical activation, while promoting their acquisition of phenotypic and secretory features of alternative activation. Furthermore, we identify the triggering receptor expressed on myeloid cells (TREM)-1, a member of the Ig-like immunoregulatory receptor family, as a hypoxia-inducible gene in Mf and demonstrate that its engagement by an agonist Ab reverses the M2-polarizing effect of hypoxia imparting a M1-skewed phenotype to Mf. Finally, we provide evidence that Mf infiltrating the inflamed hypoxic joints of children affected by oligoarticular juvenile idiopatic arthritis express high surface levels of TREM-1 associated with predominant M1 polarization and suggest the potential of this molecule in driving M1 proinflammatory reprogramming in the hypoxic synovial environment.

10.
JAMA Ophthalmol ; 134(10): 1125-1133, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27532663

RESUMO

IMPORTANCE: Chromosome 6p amplification is associated with more benign behavior for uveal melanomas (UMs) with an otherwise high risk of metastasis conferred by chromosome 3 monosomy. Chromosome 6p contains several members of the B7 family of immune regulator genes, including butyrophilin-like 2 (BTNL2; OMIM, 606000), which is associated with prostate cancer risk and autoimmune diseases. OBJECTIVE: To investigate the expression and variant allele frequencies of BTNL2, a candidate gene for chromosome 6 amplification, in patients with UM. DESIGN, SETTING, AND PARTICIPANTS: In this case-control study, we analyzed the expression of BTNL2 in UM cell lines and human macrophages in patients with UM. Variants of BTNL2 were analyzed using probes for polymerase chain reaction and high-resolution melting. The association of missense variants rs28362679 and rs41441651 with tumor risk was analyzed in 209 patients with UM and 116 matched control patients as well as 12 UM and 64 other tumor cell lines. Genes that were differentially expressed in M1- and M2-polarized macrophages were identified by microarray analysis of 111 patients with UM, and the association of the expression of these genes with disease-free survival was analyzed by Cox regression analysis. Data were collected from September 2013 to November 2015. MAIN OUTCOMES AND MEASURES: Butyrophilin-like 2 single-nucleotide variants were associated with UM risk; M1 and M2 macrophage-specific gene expression was associated with disease-free survival. RESULTS: We genotyped a total of 325 patients. Of the 209 patients with UM, 124 (59.3%) were male, 114 (54.5%) were Italian, and 95 (45.5%) were German; the mean (range) age was 65 (27-94) years. Of the 116 Italian control patients, 67 (57.8%) were female, and the mean (range) age was 39 (21-88) years. Butyrophilin-like 2 is expressed in patients with UM and macrophages. The frequency of the rs28362679 variant was higher in patients with UM (16 of 209 [7.7%]; 95% CI, 4.7-12.2) than frequencies from European Variation Archive and Exome Aggregation Consortium data (2134 of 118 564 [1.8%]; 95% CI, 1.7-1.9) and Exome Sequencing Project data (100 of 4540 [2.2%]; 95% CI, 1.8-2.7) but were not higher compared with Italian control patients (10 of 116 [8.6%]; 95% CI, 4.6-15.4). The rs41441651 variant was present in 5 patients with UM (2.4%; 95% CI, 0.9-5.7), 2 Italian control patients (1.7%; 95% CI, 0.1-6.5), 2846 patients from European Variation Archive and Exome Aggregation Consortium data (2.4%; 95% CI, 2.3-2.5), and 23 patients from Exome Sequencing Project data (0.5%; 95% CI, 0.3-0.8). Human UM cells express M1 and M2 macrophage-specific genes, whose expression is associated with disease-free survival. CONCLUSIONS AND RELEVANCE: Butyrophilin-like 2, expressed at various levels by UM cells and macrophages, might interfere with the immune control of the tumor. Butyrophilin-like 2 variants showed highly variable frequencies among ethnically related cohorts. There was no enrichment of BTNL2 variants in patients with UM compared with control patients.


Assuntos
Butirofilinas/genética , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Polimorfismo de Nucleotídeo Único , Neoplasias Uveais/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Butirofilinas/biossíntese , Linhagem Celular Tumoral , Feminino , Frequência do Gene , Genótipo , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Melanoma/diagnóstico , Melanoma/metabolismo , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Estudos Retrospectivos , Neoplasias Uveais/diagnóstico , Neoplasias Uveais/metabolismo
11.
Curr Pharm Des ; 22(41): 6209-6233, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27568730

RESUMO

The triggering receptor expressed on myeloid cells (TREM)-1 is a member of the Ig-like immunoregulatory receptor family and a major amplifier of innate immune responses. TREM- 1 has been implicated in the development and perpetuation of a number of inflammatory disorders, and soluble TREM-1 levels are a clinically valuable diagnostic and prognostic biomarker in patients with sepsis and other types of acute and chronic inflammation- associated diseases, easily detectable in biological fluids. High TREM-1 expression in macrophages infiltrating human tumors and increased concentrations of soluble TREM-1 also correlate with aggressive tumor behavior and recurrence and are a relevant independent predictor of poor patient survival. Pharmacological inhibition of TREM-1 has proven effective in preclinical mouse models of infectious and non-infectious inflammatory disorders and malignancies, conferring survival advantages and protecting from organ damage or tumor growth by attenuating inflammatory responses. This review aims at providing a comprehensive overview of the state of the art on TREM-1 research. We review the literature addressing TREM-1 role in the amplification of myeloid cell inflammatory responses at pathologic sites and its relevance in the development, severity, and progression of inflammatory diseases and cancer. Furthermore, we discuss recent advances in the pharmacological use of TREM-1 inhibitors in mouse preclinical models, emphasizing their potential in new strategies for the treatment of acute and chronic inflammatory conditions and for therapeutic intervention in cancer. This information will be of value to investigators in the field of pharmacology, drawing attention to novel therapeutic opportunities to complement current treatment approaches.


Assuntos
Inflamação/tratamento farmacológico , Neoplasias/tratamento farmacológico , Receptor Gatilho 1 Expresso em Células Mieloides/antagonistas & inibidores , Animais , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Neoplasias/imunologia , Neoplasias/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides/imunologia , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo
12.
Innate Immun ; 20(7): 721-34, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24131792

RESUMO

Peripheral blood monocytes are recruited to inflammatory and tumor lesions where they undergo terminal differentiation into macrophages. Monocytes/macrophages integrate stimulatory and inhibitory signals present in the pathologic microenvironment through a defined repertoire of cell surface receptors, and deregulated expression of these molecules may result in amplification of inflammation or establishment of immune escape mechanisms. Characterization of the expression and function of these receptors is required for a better understanding of the regulation of monocyte/macrophage activity at pathologic sites. Hypoxia is a common feature of many pathological situations and an important regulator of monocyte/macrophage pro-inflammatory responses. In this study, we identify the leukocyte membrane antigen, CD300a, a member of the CD300 superfamily of immunoregulatory receptors, as a new hypoxia-inducible gene in primary human monocytes and monocyte-derived macrophages. CD300a mRNA up-regulation by hypoxia was rapid and reversible, paralleled by increased surface protein expression, and mediated by hypoxia-inducible factor-1α. CD300a induction was also triggered by the hypoxia-mimetic agent, desferrioxamine. CD300a exhibited both activating and inhibitory potential, differentially regulating CCL20 and vascular endothelial growth factor pro-inflammatory cytokine production by monocytes/macrophages upon triggering by an agonist Ab. These results suggest that CD300a induction by the hypoxic environment represents a mechanism of regulation of monocyte/macrophage pro-inflammatory responses at pathologic sites.


Assuntos
Antígenos CD/genética , Quimiocina CCL20/biossíntese , Hipóxia/genética , Hipóxia/metabolismo , Macrófagos/metabolismo , Monócitos/metabolismo , Receptores Imunológicos/genética , Fator A de Crescimento do Endotélio Vascular/biossíntese , Citometria de Fluxo , Humanos , Técnicas In Vitro , Quelantes de Ferro/farmacologia , Macrófagos/efeitos dos fármacos , Monócitos/efeitos dos fármacos , Cultura Primária de Células
13.
Eur J Immunol ; 43(10): 2756-64, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23913266

RESUMO

In certain infection sites or tumor tissues, the disruption of homeostasis can give rise to a hypoxic microenvironment, which, in turn, can alter the function of different immune cell types and favor the progression of the disease. Natural killer (NK) cells are directly involved in the elimination of virus-infected or transformed cells, however it is unknown whether their function is affected by hypoxia or not. In this study, we show that NK cells adapt to a hypoxic environment by upregulating the hypoxia-inducible factor 1α. However, NK cells lose their ability to upregulate the surface expression of the major activating NK-cell receptors (NKp46, NKp30, NKp44, and NKG2D) in response to IL-2 (or other activating cytokines, including IL-15, IL-12, and IL-21). These altered phenotypic features correlate with reduced responses to triggering signals resulting in impaired capability of killing infected or tumor target cells. Remarkably, hypoxia does not significantly alter the surface density and the triggering function of the Fc-γ receptor CD16, thus allowing NK cells to maintain their capability of killing target cells via antibody-dependent cellular cytotoxicity. This finding offers an important clue for exploitation of NK cell in antibody-based immunotherapy of cancer.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/imunologia , Células Matadoras Naturais/imunologia , Antígenos de Neoplasias/imunologia , Células Cultivadas , Microambiente Celular , Citocinas/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Ativação Linfocitária , Receptores Desencadeadores da Citotoxicidade Natural/genética , Receptores Desencadeadores da Citotoxicidade Natural/metabolismo
14.
Immunobiology ; 218(1): 76-89, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22465745

RESUMO

Myeloid dendritic cells (DCs) are professional antigen-presenting cells critical for the orchestration of immunity and maintenance of self-tolerance. DC development and functions are tightly regulated by a complex network of inhibitory and activating signals present in the tissue microenvironment, and dysregulated DC responses may result in amplification of inflammation, loss of tolerance, or establishment of immune escape mechanisms. Generation of mature (m)DCs from monocytic precursors recruited at pathological sites occurs under condition of low partial oxygen pressure (pO(2)). However, the way in which the hypoxic microenvironment modulates the functions of these cells is still not clear. We demonstrate that chronic hypoxia (4 days, 1% O(2)) promotes the onset of a highly proinflammatory gene expression profile in mDCs generated from primary human monocytes, characterized by the modulation of a significant cluster of genes coding for proinflammatory chemokines/cytokines and/or their receptors. Within the chemokine system, strong upregulation of genes encoding proteins chemotactic for neutrophils, such as CXCL2, CXCL3, CXCL5, CXCL6, and CXCL8, and for activated/memory T lymphocytes, monocytes, and immature (i) DCs, e.g. CCL20, CCL3 and CCL5, was observed, concomitant with decreased expression of genes coding for naive/resting T cells chemoattractants, CCL18 and CCL23. Other hypoxia-inducible genes coded for cytokines with a primary role in inflammation and angiogenesis, including osteopontin, vascular endothelial growth factor, and IL-1ß. mRNA modulation was paralleled by protein secretion. These results suggest that conditions of reduced O(2) availability reprograms mDCs toward a proinflammatory direction by tuning the cytokine/chemokine repertoire, thus affecting their ability to regulate leukocyte trafficking and activation at pathological sites, with potential implications for the pathogenesis of chronic inflammatory diseases.


Assuntos
Hipóxia Celular/imunologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Dendríticas/imunologia , Osteopontina/metabolismo , Diferenciação Celular , Movimento Celular , Células Cultivadas , Microambiente Celular/imunologia , Quimiocinas/genética , Citocinas/genética , Regulação da Expressão Gênica/imunologia , Humanos , Imunidade Celular/genética , Mediadores da Inflamação/metabolismo , Monócitos/imunologia , Neovascularização Patológica/genética , Osteogênese/genética , Osteopontina/genética , Transcriptoma , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Mol Immunol ; 47(4): 685-93, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19939449

RESUMO

Alterations in iron availability can trigger pro-inflammatory signals in various cell types. We demonstrate that desferrioxamine (DFX), an iron chelator used in clinics for the treatment of iron overload, neoplasias, and Alzheimer disease, stimulates the expression and secretion of CCL20, a chemoattractant for immature dendritic cells, activated/memory T lymphocytes, and naive B cells, in primary human monocytes and monocyte-derived macrophages. Iron chelation was part of the mechanism by which DFX induced CCL20, because addition of iron sulfate counteracted its stimulatory effects. Functional studies of the CCL20 promoter, using a series of 5'-deleted and mutated reporter constructs, demonstrated that CCL20 mRNA induction was dependent on gene transcription activation and mediated by the NF-kappaB pathway. The NF-kappaB element located at position -92/-82 of the CCL20 promoter was required for gene transactivation by DFX because: (i) transcription was abrogated by a 3bp mutation of the NF-kappaB-binding motif; (ii) treatment with DFX increased specific NF-kappaB binding to this sequence. Nuclear translocation of both NF-kappaBp65 and NF-kappaBp50 family members was increased in response to DFX and associated with I-kappaBalpha degradation, suggesting a role for these subunits in CCL20 promoter transactivation. In conclusion, this study provides the first evidence that iron chelation can transcriptionally induce CCL20 in mononuclear phagocytes and identify the NF-kappaB binding site as a regulatory sequence of the CCL20 promoter that is activated by iron deprivation. These results add new insights into our understanding of the mechanisms by which iron perturbations affect mononuclear phagocyte immune functions and regulate inflammatory responses.


Assuntos
Quimiocina CCL20/genética , Desferroxamina/farmacologia , Quelantes de Ferro/farmacologia , NF-kappa B/metabolismo , Fagócitos/efeitos dos fármacos , Fagócitos/metabolismo , Transcrição Gênica/efeitos dos fármacos , Animais , Sítios de Ligação , Hipóxia Celular/efeitos dos fármacos , Quimiocina CCL20/metabolismo , DNA/metabolismo , Humanos , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
16.
Neurotox Res ; 15(2): 138-54, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19384577

RESUMO

Several in vitro and in vivo studies addressed the identification of molecular determinants of the neuronal death induced by PrP(Sc) or related peptides. We developed an experimental model to assess PrP(Sc) neurotoxicity using a recombinant polypeptide encompassing amino acids 90-231 of human PrP (hPrP90-231) that corresponds to the protease-resistant core of PrP(Sc) identified in prion-infected brains. By means of mild thermal denaturation, we can convert hPrP90-231 from a PrP(C)-like conformation into a PrP(Sc)-like structure. In virtue of these structural changes, hPrP90-231 powerfully affected the survival of SH-SY5Y cells, inducing caspase 3 and p38-dependent apoptosis, while in the native alpha-helix-rich conformation, hPrP90-231 did not induce cell toxicity. The aim of this study was to identify drugs able to block hPrP90-231 neurotoxic effects, focusing on minocycline, a tetracycline with known neuroprotective activity. hPrP90-231 caused a caspase 3-dependent apoptosis via the blockade of ERK1/2 activation and the subsequent activation of p38 MAP kinase. We propose that hPrP90-231-induced apoptosis is dependent on the inhibition of ERK1/2 responsiveness to neurotrophic factors, removing a tonic inhibition of p38 activity and resulting in caspase 3 activation. Minocycline prevented hPrP90-231-induced toxicity interfering with this mechanism: the pretreatment with this tetracycline restored ERK1/2 activity and reverted p38 and caspase 3 activities. The effects of minocycline were not mediated by the prevention of hPrP90-231 structural changes or cell internalization (differently from Congo Red). In conclusion, minocycline elicits anti-apoptotic effects against the neurotoxic activity of hPrP90-231 and these effects are mediated by opposite modulation of ERK1/2 and p38 MAP kinase activities.


Assuntos
Apoptose/efeitos dos fármacos , Minociclina/farmacologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurotoxinas/toxicidade , Fragmentos de Peptídeos/toxicidade , Príons/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Interações Medicamentosas , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neuroblastoma/patologia , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA