Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Int J Mol Sci ; 20(20)2019 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-31615111

RESUMO

Natural occurring polymers, or biopolymers, represent a huge part of our planet biomass. They are formed by long chains of monomers of the same type or a combination of different ones. Polysaccharides are biopolymers characterized by complex secondary structures performing several roles in plants, animals, and microorganisms. Because of their versatility and biodegradability, some of them are extensively used for packaging, food, pharmaceutical, and biomedical industries as sustainable and renewable materials. In the recent years, their manipulation at the nanometric scale enormously increased the range of potential applications, boosting an interdisciplinary research attempt to exploit all the potential advantages of nanostructured polysaccharides. Biomedical investigation mainly focused on nano-objects aimed at drug delivery, tissue repair, and vaccine adjuvants. The achievement of all these applications requires the deep knowledge of polysaccharide nanomaterials' interactions with the immune system, which orchestrates the biological response to any foreign substance entering the body. In the present manuscript we focused on natural polysaccharides of high commercial importance, namely, starch, cellulose, chitin, and its deacetylated form chitosan, as well as the seaweed-derived carrageenan and alginate. We reviewed the available information on their biocompatibility, highlighting the importance of their physicochemical feature at the nanoscale for the modulation of the immune system.


Assuntos
Materiais Biocompatíveis/química , Biopolímeros/química , Sistemas de Liberação de Medicamentos , Polissacarídeos/química , Adjuvantes Imunológicos/química , Alginatos/química , Carragenina/química , Celulose/química , Quitina/química , Quitosana/química , Humanos , Nanoestruturas/química , Amido/química
2.
Small ; 14(24): e1800227, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29756263

RESUMO

The biotransformation and biological impact of few layer graphene (FLG) and graphene oxide (GO) are studied, following ingestion as exposure route. An in vitro digestion assay based on a standardized operating procedure (SOP) is exploited. The assay simulates the human ingestion of nanomaterials during their dynamic passage through the different environments of the gastrointestinal tract (salivary, gastric, intestinal). Physical-chemical changes of FLG and GO during digestion are assessed by Raman spectroscopy. Moreover, the effect of chronic exposure to digested nanomaterials on integrity and functionality of an in vitro model of intestinal barrier is also determined according to a second SOP. These results show a modulation of the aggregation state of FLG and GO nanoflakes after experiencing the complex environments of the different digestive compartments. In particular, chemical doping effects are observed due to FLG and GO interaction with digestive juice components. No structural changes/degradation of the nanomaterials are detected, suggesting that they are biopersistent when administered by oral route. Chronic exposure to digested graphene does not affect intestinal barrier integrity and is not associated with inflammation and cytotoxicity, though possible long-term adverse effects cannot be ruled out.


Assuntos
Grafite/administração & dosagem , Grafite/farmacologia , Administração Oral , Biotransformação , Células CACO-2 , Proteínas Filagrinas , Humanos , Inflamação/patologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Nanopartículas/química , Nanopartículas/ultraestrutura , Análise Espectral Raman
3.
Chem Soc Rev ; 46(16): 4951-4975, 2017 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-28696452

RESUMO

Oxidative stress-dependent inflammatory diseases represent a major concern for the population's health worldwide. Biocompatible nanomaterials with enzymatic properties could play a crucial role in the treatment of such pathologies. In this respect, platinum nanoparticles (PtNPs) are promising candidates, showing remarkable catalytic activity, able to reduce the intracellular reactive oxygen species (ROS) levels and impair the downstream pathways leading to inflammation. This review reports a critical overview of the growing evidence revealing the anti-inflammatory ability of PtNPs and their potential applications in nanomedicine. It provides a detailed description of the wide variety of synthetic methods recently developed, with particular attention to the aspects influencing biocompatibility. Special attention has been paid to the studies describing the toxicological profile of PtNPs with an attempt to draw critical conclusions. The emerging picture suggests that the material per se is not causing cytotoxicity, while other physicochemical features related to the synthesis and surface functionalization may play a crucial role in determining the observed impairment of cellular functions. The enzymatic activity of PtNPs is also summarized, analyzing their action against ROS produced by pathological conditions within the cells. In particular, we extensively discuss the potential of these properties in nanomedicine to down-regulate inflammatory pathways or to be employed as diagnostic tools with colorimetric readout. A brief overview of other biomedical applications of nanoplatinum is also presented.


Assuntos
Nanopartículas Metálicas/química , Nanomedicina , Platina/química , Humanos , Tamanho da Partícula
4.
J Immunol ; 192(11): 5305-13, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24778447

RESUMO

The cross-regulation of G protein-coupled receptors (GPCRs) plays an important role in the immune response. Studies from several laboratories have suggested that a hierarchy of sensitivities to cross-desensitization exists for the chemoattractant GPCRs. We carried out experiments to study the capacity of the formyl peptide receptor-1 (FPR1) to desensitize chemokine receptors CCR1 and CCR2. Our results show that activation of FPR1 resulted in the desensitization and partial internalization of CCR1, but not CCR2, in both primary human monocytes and HEK293 cells coexpressing CCR1, CCR2, and FPR1 (HR1R2F cells). The desensitization of CCR1 by FPR1 stimulation was not due to the simple depletion of the Ca(2+) stores, but was dependent on activation of protein kinase C. Furthermore, we found that the cross-desensitization of CCR1 by FPR1 was associated with CCR1 phosphorylation and moderate reduction of CCR1 cell-surface expression. In contrast, CCR2 was not phosphorylated or internalized after FPR1 activation. Additional studies showed that optimal cross talk between FPR1 and CCR1 was dependent on the functional activity of protein kinase Cß. These results provide a mechanistic basis for the capacity of certain GPCR ligands to exert rapid and selective cross-inactivation of other chemoattractant receptors, and suggest that FPR1 is able to exert "traffic control" in the migration of inflammatory cells by rapidly inhibiting the cell responses to potentially "low-priority" chemoattractants such as CCR1 agonists without inhibiting the response to "higher priority" CCR2 chemoattractants.


Assuntos
Monócitos/imunologia , Receptores CCR1/imunologia , Receptores CCR2/imunologia , Receptores de Formil Peptídeo/imunologia , Cálcio/imunologia , Regulação da Expressão Gênica/imunologia , Células HEK293 , Humanos , Monócitos/citologia , Proteína Quinase C beta/imunologia
5.
Cell Mol Life Sci ; 71(1): 1-20, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23508806

RESUMO

RNA interference has been envisaged as a powerful tool for molecular and clinical investigation with a great potential for clinical applications. In recent years, increased understanding of cancer biology and stem cell biology has dramatically accelerated the development of technology for cell and gene therapy in these areas. This paper is a review of the most recent report of innovative use of siRNA to benefit several central nervous system diseases. Furthermore, a description is made of innovative strategies of delivery into the brain by means of viral and non-viral vectors with high potential for translation into clinical use. Problems are also highlighted that might hamper the transition from bench to bed, analyzing the lack of reliable preclinical models with predictive validity and the lack of effective delivery systems, which are able to overcome biological barriers and specifically reach the brain site of action.


Assuntos
Doenças do Sistema Nervoso/terapia , RNA Interferente Pequeno/administração & dosagem , Barreira Hematoencefálica/metabolismo , Portadores de Fármacos/química , Vetores Genéticos/metabolismo , Humanos , Nanopartículas/química , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/patologia , Interferência de RNA , Pesquisa Translacional Biomédica , Vírus/genética
6.
Nanomedicine ; 11(3): 731-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25546848

RESUMO

Toxicity of silver nanoparticles (AgNPs) is supported by many observations in literature, but no mechanism details have been proved yet. Here we confirm and quantify the toxic potential of fully characterized AgNPs in HeLa and A549 cells. Notably, through a specific fluorescent probe, we demonstrate the intracellular release of Ag(+) ions in living cells after nanoparticle internalization, showing that in-situ particle degradation is promoted by the acidic lysosomal environment. The activation of metallothioneins in response to AgNPs and the possibility to reverse the main toxic pathway by Ag(+) chelating agents demonstrate a cause/effect relationship between ions and cell death. We propose that endocytosed AgNPs are degraded in the lysosomes and the release of Ag(+) ions in the cytosol induces cell damages, while ions released in the cell culture medium play a negligible effect. These findings will be useful to develop safer-by-design nanoparticles and proper regulatory guidelines of AgNPs. From the clinical editor: The authors describe the toxic potential of silver nanoparticles (AgNP) in human cancer cell lines. Cell death following the application of AgNPs is dose-dependent, and it is mostly due to Ag+ ions. Further in vivo studies should be performed to gain a comprehensive picture of AgNP-toxicity in mammals.


Assuntos
Citosol/metabolismo , Nanopartículas Metálicas/química , Prata , Cátions Monovalentes/farmacocinética , Células HeLa , Humanos , Lisossomos/metabolismo , Prata/química , Prata/farmacocinética , Prata/farmacologia
7.
Proc Natl Acad Sci U S A ; 108(27): 10952-7, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690348

RESUMO

Stroke is the second cause of death worldwide with ischemic stroke accounting for 80% of all stroke insults. Caspase-3 activation contributes to brain tissue loss and downstream biochemical events that lead to programmed cell death after traumatic brain injury. Alleviation of symptoms following ischemic neuronal injury can be potentially achieved by either genetic disruption or pharmacological inhibition of caspases. Here, we studied whether silencing of Caspase-3 using carbon nanotube-mediated in vivo RNA interference (RNAi) could offer a therapeutic opportunity against stroke. Effective delivery of siRNA directly to the CNS has been shown to normalize phenotypes in animal models of several neurological diseases. It is shown here that peri-lesional stereotactic administration of a Caspase-3 siRNA (siCas 3) delivered by functionalized carbon nanotubes (f-CNT) reduced neurodegeneration and promoted functional preservation before and after focal ischemic damage of the rodent motor cortex using an endothelin-1 induced stroke model. These observations illustrate the opportunity offered by carbon nanotube-mediated siRNA delivery and gene silencing of neuronal tissue applicable to a variety of different neuropathological conditions where intervention at well localized brain foci may offer therapeutic and functional benefits.


Assuntos
Isquemia Encefálica/terapia , Caspase 3/genética , Inibidores de Caspase , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Apoptose/fisiologia , Sequência de Bases , Isquemia Encefálica/enzimologia , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatologia , Linhagem Celular , Células Cultivadas , Endotelina-1/toxicidade , Feminino , Terapia Genética , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Nanomedicina , Nanotubos de Carbono , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Desempenho Psicomotor , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , Ratos , Ratos Sprague-Dawley
8.
Mol Pharm ; 10(1): 249-60, 2013 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-23163881

RESUMO

Dendrimers have been described as one of the most tunable and therefore potentially applicable nanoparticles both for diagnostics and therapy. Recently, in order to realize drug delivery agents, most of the effort has been dedicated to the development of dendrimers that could internalize into the cells and target specific intracellular compartments in vitro and in vivo. Here, we describe cell internalization properties and diffusion of G4 and G4-C12 modified PAMAM dendrimers in primary neuronal cultures and in the CNS of live animals. Confocal imaging on primary neurons reveals that dendrimers are able to cross the cell membrane and reach intracellular localization following endocytosis. Moreover, functionalization of PAMAMs has a dramatic effect on their ability to diffuse in the CNS tissue in vivo and penetrate living neurons as shown by intraparenchymal or intraventricular injections. 100 nM G4-C12 PAMAM dendrimer already induces dramatic apoptotic cell death of neurons in vitro. On the contrary, G4 PAMAM does not induce apoptotic cell death of neural cells in the sub-micromolar range of concentration and induces low microglia activation in brain tissue after a week. Our detailed description of dendrimer distribution patterns in the CNS will facilitate the design of tailored nanomaterials in light of future clinical applications.


Assuntos
Apoptose/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Dendrímeros/química , Dendrímeros/farmacocinética , Nylons/química , Nylons/farmacocinética , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Dendrímeros/toxicidade , Difusão , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Nylons/toxicidade , Córtex Visual/efeitos dos fármacos , Córtex Visual/metabolismo
9.
Vaccines (Basel) ; 11(8)2023 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-37631901

RESUMO

The mucosal barrier constitutes a huge surface area, close to 40 m2 in humans, located mostly in the respiratory, gastrointestinal and urogenital tracts and ocular cavities. It plays a crucial role in tissue interactions with the microbiome, dietary antigens and other environmental materials. Effective vaccinations to achieve highly protective mucosal immunity are evolving strategies to counteract several serious diseases including tuberculosis, diphtheria, influenzae B, severe acute respiratory syndrome, Human Papilloma Virus infection and Acquired Immune Deficiency Syndrome. Interestingly, one of the reasons behind the rapid spread of severe acute respiratory syndrome coronavirus 2 variants has been the weakness of local immunization at the level of the respiratory mucosa. Mucosal vaccines can outperform parenteral vaccination as they specifically elicit protective mucosal immune responses blocking infection and transmission. In this scenario, chitosan-based nanovaccines are promising adjuvants-carrier systems that rely on the ability of chitosan to cross tight junctions and enhance particle uptake due to chitosan-specific mucoadhesive properties. Indeed, chitosan not only improves the adhesion of antigens to the mucosa promoting their absorption but also shows intrinsic immunostimulant abilities. Furthermore, by finely tuning the colloidal properties of chitosan, it can provide sustained antigen release to strongly activate the humoral defense. In the present review, we agnostically discuss the potential reasons why chitosan-based vaccine carriers, that efficiently elicit strong immune responses in experimental setups and in some pre-clinical/clinical studies, are still poorly considered for therapeutic formulations.

10.
Nanoscale ; 15(35): 14284-14300, 2023 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-37584343

RESUMO

Inflammation is a complex process of the body in response to pathogen infections or dysregulated metabolism, involving the recruitment and activation of immune system components. Repeated dangerous stimuli or uncontrolled immune effector mechanisms can result in tissue injury. Reactive Oxygen Species (ROS) play key roles in physiological cell signaling as well as in the destruction of internalized pathogens. However, aberrant ROS production and release have deleterious effects on the surrounding environment, making ROS regulation a priority to reduce inflammation. Most of the current anti-inflammatory therapies rely on drugs that impair the release of pro-inflammatory mediators. Nevertheless, increasing the enzymatic activity to reduce ROS levels could be an alternative or complementary therapeutic approach to decrease inflammation. Nanozymes are nanomaterials with high catalytic activity that mimic natural enzymes, allowing biochemical reactions to take place. Such functional particles typically show different and regenerable oxidation states or catalytically reactive surfaces offering long-term activity and stability. In this scenario, platinum-based nanozymes (PtNZs) exhibit broad and efficient catalytic functionalities and can reduce inflammation mainly through ROS scavenging, e.g. by catalase and superoxide dismutase reactions. Dose-dependent biocompatibility and immune compatibility of PtNZs have been shown in different cells and tissues, both in vitro and in vivo. Size/shape/surface engineering of the nanozymes could also potentiate their efficacy to act at different sites and/or steps of the inflammation process, such as cytokine removal or specific targeting of activated leukocytes. In the present review, we analyze key inflammation triggering processes and the effects of platinum nanozymes under exemplificative inflammatory conditions. We further discuss potential platinum nanozyme design and improvements to modulate and expand their anti-inflammatory action.


Assuntos
Nanoestruturas , Platina , Humanos , Espécies Reativas de Oxigênio/metabolismo , Inflamação/tratamento farmacológico , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico
11.
J Biol Chem ; 286(23): 20354-65, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21454526

RESUMO

We have previously shown that the µ-opioid receptor (MOR) is capable of mediating cross-desensitization of several chemokine receptors including CCR5, but the biochemical mechanism of this process has not been fully elucidated. We have carried out a series of functional and biochemical studies and found that the mechanism of MOR-induced cross-desensitization of CCR5 involves the activation of PKCζ. Inhibition of PKCζ by its pseudosubstrate inhibitor, or its siRNA, or dominant negative mutants suppresses the cross-desensitization of CCR5. Our results further indicate that the activation of PKCζ is mediated through a pathway involving phosphoinositol-dependent kinase-1 (PDK1). In addition, activation of MOR elevates the phosphorylation level and kinase activity of PKCζ. The phosphorylation of PKCζ can be suppressed by a dominant negative mutant of PDK1. We observed that following MOR activation, the interaction between PKCζ and PDK1 is immediately increased based on the analysis of fluorescent resonance energy transfer in cells with the expression of PKCζ-YFP and PDK1-CFP. In addition, cells expressing PKCζ kinase motif mutants (Lys-281, Thr-410, Thr-560) fail to exhibit full MOR-induced desensitization of CCR5 activity. Taken together, we propose that upon DAMGO treatment, MOR activates PKCζ through a PDK1-dependent signaling pathway to induce CCR5 phosphorylation and desensitization. Because CCR5 is a highly proinflammatory receptor, and a critical coreceptor for HIV-1, these results may provide a novel approach for the development of specific therapeutic agents to treat patients with certain inflammatory diseases or AIDS.


Assuntos
Proteína Quinase C/metabolismo , Receptores Opioides mu/metabolismo , Transdução de Sinais/fisiologia , Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Síndrome da Imunodeficiência Adquirida/genética , Síndrome da Imunodeficiência Adquirida/metabolismo , Motivos de Aminoácidos , Analgésicos Opioides/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , HIV-1 , Humanos , Camundongos , Mutação , Fragmentos de Peptídeos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil , Receptores CCR5 , Receptores Opioides mu/genética , Transdução de Sinais/efeitos dos fármacos
12.
Biomacromolecules ; 13(12): 4089-97, 2012 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-23140570

RESUMO

Hybrid dendritic-linear block copolymers based on a 4-arm poly(ethylene glycol) (PEG) core were synthesized using an accelerated AB2/CD2 dendritic growth approach through orthogonal amine/epoxy and thiol-yne chemistries. The biological activity of these 4-arm and the corresponding 2-arm hybrid dendrimers revealed an enhanced, dendritic effect with an exponential increase in cell internalization concomitant with increasing amine end groups and low cytotoxicity. Furthermore, the ability of these hybrid dendrimers to induce endosomal escape combined with their facile and efficient synthesis makes them attractive platforms for gene transfection. The 4-arm-based dendrimer showed significantly improved DNA binding and gene transfection capabilities in comparison with the 2-arm derivative. These results combined with the MD simulation indicate a significant effect of both the topology of the PEG core and the multivalency of these hybrid macromolecules on their DNA binding and delivery capablities.


Assuntos
Cátions/química , DNA/química , Dendrímeros/química , Polietilenoglicóis/química , Sobrevivência Celular , Dendrímeros/farmacocinética , Etídio/análise , Técnicas de Transferência de Genes , Células HeLa , Humanos , Espectroscopia de Ressonância Magnética , Microscopia Confocal , Simulação de Dinâmica Molecular , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Transfecção
13.
Nanomaterials (Basel) ; 12(12)2022 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-35745372

RESUMO

This is the second Special Issue on the topic "Immune Responses to Nanomaterials for Biomedical Applications" [...].

14.
Nanomaterials (Basel) ; 12(20)2022 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-36296750

RESUMO

Specific cell targeting to deliver nanoparticles can be achieved by tailored modifications of the material surface with chemical moieties. The selection of the cell targets can be optimized by covering the nanoparticle with molecules, the receptor expression of which is restricted to particular cell subsets. Chemokines perform their biological action through 7-TM Gi-protein-coupled receptors differently expressed in all tissues. We decorated the surface of biocompatible polymer nanoparticles with full-length CCL5, an inflammatory chemokine that attracts leukocytes by binding CCR5, which is highly expressed in blood-circulating monocytes. Our observations showed that CCL5 functionalization does not affect the nanoparticle biocompatibility. Notably, CCL5 NPs delivered to PBMCs are selectively internalized by CCR5+ monocytes but not by CCR5- lymphocytes. The efficacy of PBMC subpopulation targeting by chemokine-decorated nanoparticles establishes an easy-to-use functionalization for specific leukocyte delivery.

15.
Nanomaterials (Basel) ; 11(5)2021 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-34066755

RESUMO

The present Special Issue hosts six research papers and five review articles regarding different aspects of nanotechnologies for therapeutic and diagnostic applications [...].

16.
Nanomedicine (Lond) ; 16(26): 2377-2387, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34632802

RESUMO

As the current COVID-19 pandemic illustrates, vaccination is the most powerful method of disease prevention and public confidence in vaccines depends on their safety and efficacy. The information gathered in the current pandemic is growing at an accelerated pace. Both the key vital protein DNA/RNA messengers and the delivery carriers are the elements of a puzzle including their interactions with the immune system to suppress SARS-CoV-2 infection. A new nano-era is beginning in the vaccine development field and an array of side applications for diagnostic and antiviral tools will likely emerge. This review focuses on the evolution of vaccine carriers up to COVID-19-aimed nanoparticles and the immune-related adverse effects imposed by these nanocarriers.


Assuntos
COVID-19 , Vacinas , Vacinas contra COVID-19 , Humanos , Pandemias , SARS-CoV-2
17.
J Exp Med ; 196(6): 753-63, 2002 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-12235209

RESUMO

Mice lacking the p110delta catalytic subunit of phosphatidylinositol 3-kinase have reduced numbers of B1 and marginal zone B cells, reduced levels of serum immunoglobulins, respond poorly to immunization with type II thymus-independent antigen, and are defective in their primary and secondary responses to thymus-dependent antigen. p110delta(-/-) B cells proliferate poorly in response to B cell receptor (BCR) or CD40 signals in vitro, fail to activate protein kinase B, and are prone to apoptosis. p110delta function is required for BCR-mediated calcium flux, activation of phosphlipaseCgamma2, and Bruton's tyrosine kinase. Thus, p110delta plays a critical role in B cell homeostasis and function.


Assuntos
Linfócitos B/fisiologia , Ativação Linfocitária , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Serina-Treonina Quinases , Tirosina Quinase da Agamaglobulinemia , Animais , Formação de Anticorpos , Cálcio/metabolismo , Isoenzimas/fisiologia , Masculino , Camundongos , Camundongos Knockout , Fosfatos de Fosfatidilinositol/biossíntese , Fosfolipase C gama , Subunidades Proteicas , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , Receptores de Antígenos de Linfócitos B/fisiologia , Fosfolipases Tipo C/fisiologia
18.
BMC Biol ; 7: 87, 2009 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-20017911

RESUMO

BACKGROUND: Obesity is a chronic low inflammatory state. In the obesity condition the white adipose tissue (WAT) is massively infiltrated with monocytes/macrophages, and the nature of the signals recruiting these inflammatory cells has yet to be fully elucidated. Haptoglobin (Hp) is an inflammatory marker and its expression is induced in the WAT of obese subjects. In an effort to elucidate the biological significance of Hp presence in the WAT and of its upregulation in obesity we formulated the hypothesis that Hp may serve as a macrophage chemoattractant. RESULTS: We demonstrated by chemotaxis assay that Hp is able to attract chemokine (C-C motif) receptor 2 (CCR2)-transfected pre-B lymphocytes and monocytes in a dose-dependent manner. Moreover, Hp-mediated migration of monocytes is impaired by CCR2-specific inhibition or previous cell exposure to monocyte chemoattractant protein 1 (MCP1) (also known as CCR2 ligand or chemokine (C-C motif) ligand 2 (CCL2)). Downstream effects of Hp/CCR2 interaction were also investigated: flow cytometry proved that monocytes treated with Hp show reduced CCR2 expression on their surface; Hp interaction induces calcium release that is reduced upon pretreatment with CCR2 antagonist; extracellular signal-regulated kinase (ERK)1/2, a signal transducer activated by CCR2, is phosphorylated following Hp treatment and this phosphorylation is reduced when cells are pretreated with a specific CCR2 inhibitor. Consistently, blocking the ERK1/2 pathway with U0126, the selective inhibitor of the ERK upstream mitogen-activated protein (MAP)-ERK kinase (MEK), results in a dramatic reduction (by almost 100%) of the capability of Hp to induce monocyte migration. CONCLUSIONS: Our data show that Hp is a novel monocyte chemoattractant and that its chemotactic potential is mediated, at least in part. by its interaction with CCR2.


Assuntos
Quimiotaxia/fisiologia , Haptoglobinas/metabolismo , Monócitos/fisiologia , Receptores CCR2/metabolismo , Adulto , Linfócitos B/efeitos dos fármacos , Linfócitos B/fisiologia , Butadienos/farmacologia , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Quimiocina CCL2/metabolismo , Quimiotaxia/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Monócitos/efeitos dos fármacos , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Receptores CCR2/agonistas , Receptores CCR2/antagonistas & inibidores , Células U937 , Adulto Jovem
19.
Vaccines (Basel) ; 8(3)2020 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-32751995

RESUMO

The ideal vaccine should trigger a specific response against pathogens and induce the immune system memory to be prepared for eventual following infections. Although different approaches to develop new vaccines are currently taken, several of the features of natural pathogens that allow a tailored immune reaction are difficult to mimic. The viral capsids are the physical interface between a virus and the host defense machinery which recognizes specific patterns of the viral supramolecular complexes. Therefore, empty viral particles deprived of their genomes represent optimal targets to induce immune reactions with several advantages for vaccination and adjuvant realization.

20.
Nanomaterials (Basel) ; 10(11)2020 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-33233846

RESUMO

Chemokine-induced chemotaxis mediates physiological and pathological immune cell trafficking, as well as several processes involving cell migration. Among them, the role of CXCL12/CXCR4 signaling in cancer and metastasis is well known, and CXCR4 has been often targeted with small molecule-antagonists or short CXCL12-derived peptides to limit the pathological processes of cell migration and invasion. To reduce CXCR4-mediated chemotaxis, we adopted a different approach. We manufactured poly(lactic acid-co-glycolic acid) (PLGA)/Pluronic F127 nanoparticles through microfluidics-assisted nanoprecipitation and functionalized them with streptavidin to docking a biotinylated CXCL12 to be exposed on the nanoparticle surface. Our results show that CXCL12-decorated nanoparticles are non-toxic and do not induce inflammatory cytokine release in THP-1 monocytes cultured in fetal bovine and human serum-supplemented media. The cell internalization of our chemokine receptor-targeting particles increases in accordance with CXCR4 expression in FBS/medium. We demonstrated that CXCL12-decorated nanoparticles do not induce cell migration on their own, but their pre-incubation with THP-1 significantly decreases CXCR4+-cell migration, thereby antagonizing the chemotactic action of CXCL12. The use of biodegradable and immune-compatible chemokine-mimetic nanoparticles to reduce cell migration opens the way to novel antagonists with potential application in cancer treatments and inflammation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA