Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Neuroinflammation ; 18(1): 75, 2021 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-33736657

RESUMO

BACKGROUND: Aging and age-related diseases are strong risk factors for the development of neurodegenerative diseases. Neuroinflammation (NIF), as the brain's immune response, plays an important role in aged associated degeneration of central nervous system (CNS). There is a need for well characterized animal models that will allow the scientific community to understand and modulate this process. METHODS: We have analyzed aging-phenotypical and inflammatory changes of brain myeloid cells (bMyC) in a senescent accelerated prone aged (SAMP8) mouse model, and compared with their senescence resistant control mice (SAMR1). We have performed morphometric methods to evaluate the architecture of cellular prolongations and determined the appearance of Iba1+ clustered cells with aging. To analyze specific constant brain areas, we have performed stereology measurements of Iba1+ cells in the hippocampal formation. We have isolated bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch), and analyzed their response to systemic lipopolysaccharide (LPS)-driven inflammation. RESULTS: Aged 10 months old SAMP8 mice present many of the hallmarks of aging-dependent neuroinflammation when compared with their SAMR1 control, i.e., increase of protein aggregates, presence of Iba1+ clusters, but not an increase in the number of Iba1+ cells. We have further observed an increase of main inflammatory mediator IL-1ß, and an augment of border MHCII+Iba1+ cells. Isolated CD45+ bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch) have been analyzed, showing that there is not a significant increase of CD45+ cells from the periphery. Our data support that aged-driven pro-inflammatory cytokine interleukin 1 beta (IL-1ß) transcription is enhanced in CD45+BP cells. Furthermore, LPS-driven systemic inflammation produces inflammatory cytokines mainly in border bMyC, sensed to a lesser extent by the BP bMyC, showing that IL-1ß expression is further augmented in aged SAMP8 compared to control SAMR1. CONCLUSION: Our data validate the SAMP8 model to study age-associated neuroinflammatory events, but careful controls for age and strain are required. These animals show morphological changes in their bMyC cell repertoires associated to age, corresponding to an increase in the production of pro-inflammatory cytokines such as IL-1ß, which predispose the brain to an enhanced inflammatory response after LPS-systemic challenge.


Assuntos
Senilidade Prematura/genética , Envelhecimento/patologia , Encefalite/genética , Encefalite/patologia , Animais , Encéfalo/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Plexo Corióideo/metabolismo , Plexo Corióideo/patologia , Modelos Animais de Doenças , Encefalite/induzido quimicamente , Hipocampo/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos , Meninges/metabolismo , Meninges/patologia , Camundongos , Proteínas dos Microfilamentos/metabolismo
2.
Haematologica ; 104(9): 1853-1865, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30573502

RESUMO

Embryonic megakaryopoiesis starts in the yolk sac on gestational day 7.5 as part of the primitive wave of hematopoiesis, and it continues in the fetal liver when this organ is colonized by hematopoietic progenitors between day 9.5 and 10.5, as the definitive hematopoiesis wave. We characterized the precise phenotype of embryo megakaryocytes in the liver at gestational day 11.5, identifying them as CD41++CD45-CD9++CD61+MPL+CD42c+ tetraploid cells that express megakaryocyte-specific transcripts and display differential traits when compared to those present in the yolk sac at the same age. In contrast to megakaryocytes from adult bone marrow, embryo megakaryocytes are CD45- until day 13.5 of gestation, as are both the megakaryocyte progenitors and megakaryocyte/erythroid-committed progenitors. At gestational day 11.5, liver and yolk sac also contain CD41+CD45+ and CD41+CD45- cells. These populations, and that of CD41++CD45-CD42c+ cells, isolated from liver, differentiate in culture into CD41++CD45-CD42c+ proplatelet-bearing megakaryocytes. Also present at this time are CD41-CD45++CD11b+ cells, which produce low numbers of CD41++CD45-CD42c+ megakaryocytes in vitro, as do fetal liver cells expressing the macrophage-specific Csf receptor-1 (Csf1r/CD115) from MaFIA transgenic mice, which give rise poorly to CD41++CD45-CD42c+ embryo megakaryocytes both in vivo and in vitro In contrast, around 30% of adult megakaryocytes (CD41++CD45++CD9++CD42c+) from C57BL/6 and MaFIA mice express CD115. We propose that differential pathways operating in the mouse embryo liver at gestational day 11.5 beget CD41++CD45-CD42c+ embryo megakaryocytes that can be produced from CD41+CD45- or from CD41+CD45+ cells, at difference from those from bone marrow.


Assuntos
Linhagem da Célula/genética , Embrião de Mamíferos/metabolismo , Antígenos Comuns de Leucócito/genética , Células Progenitoras de Megacariócitos/metabolismo , Megacariócitos/metabolismo , Animais , Antígenos CD/classificação , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Diferenciação Celular , Embrião de Mamíferos/citologia , Citometria de Fluxo , Expressão Gênica , Hematopoese/genética , Imunofenotipagem/métodos , Antígenos Comuns de Leucócito/metabolismo , Fígado/citologia , Fígado/metabolismo , Células Progenitoras de Megacariócitos/classificação , Células Progenitoras de Megacariócitos/citologia , Megacariócitos/classificação , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Cultura Primária de Células , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Tetraploidia
3.
Biomacromolecules ; 20(10): 4015-4025, 2019 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-31449398

RESUMO

Immune response to biomaterials can produce chronic inflammation and fibrosis leading to implant failure, which is related to the surface properties of the biomaterials. This work describes the preparation and characterization of polyelectrolyte multilayer (PEM) coatings that combine the anti-inflammatory activity of heparin as polyanion with the potential release of Naproxen, a nonsteroidal anti-inflammatory drug from polymeric nanoparticles (NP) with cationic surface charge. The polyelectrolyte multilayers were characterized by physical methods to estimate multilayer growth, thickness, zeta potential, and topography. It was found that multilayers with NP had negative zeta potentials and expressed a viscoelastic behavior, while studies of topography showed that nanoparticles formed continuous surface coatings. THP-1-derived macrophages were used to study short-term anti-inflammatory activity (time scale 48 h), showing that PEM that contained heparin reduced cell adhesion and IL1-ß secretion, when compared to those with polystyrenesulfonate, used as alternative polyanion in multilayer formation. On the other hand, the presence of NP in PEM was related to a reduced foreign body giant cell formation after 15 days, when compared to PEM that contained chitosan as alternative polycation, which suggests a long-term anti-inflammatory effect of Naproxen-containing nanoparticles. It was also shown that macrophages were able to take up NP from multilayers, which indicates a release of Naproxen by digestion of NP in the lysosomal compartment. These findings indicate that surface coatings composed of heparin and Naproxen-based NP on implants such as biosensors have the potential to attenuate foreign body reaction after implantation, which may improve the long-term functionality of implants.


Assuntos
Anti-Inflamatórios/química , Heparina/química , Nanopartículas/química , Naproxeno/química , Polieletrólitos/química , Anti-Inflamatórios/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Quitosana/química , Materiais Revestidos Biocompatíveis/química , Heparina/farmacologia , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Naproxeno/farmacologia , Polímeros/química , Poliestirenos/química , Propriedades de Superfície/efeitos dos fármacos
4.
Glia ; 63(6): 987-1004, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25731131

RESUMO

The study of factors that regulate the survival, proliferation, and differentiation of neural precursor cells (NPCs) is essential to understand neural development as well as brain regeneration. The Nuclear Factor of Activated T Cells (NFAT) is a family of transcription factors that can affect these processes besides playing key roles during development, such as stimulating axonal growth in neurons, maturation of immune system cells, heart valve formation, and differentiation of skeletal muscle and bone. Interestingly, NFAT signaling can also promote cell differentiation in adults, participating in tissue regeneration. The goal of the present study is to evaluate the expression of NFAT isoforms in NPCs, and to investigate its possible role in NPC survival, proliferation, migration, and differentiation. Our findings indicate that NFAT proteins are active not only in neurogenic brain regions such as hippocampus and subventricular zone (SVZ), but also in cultured NPCs. The inhibition of NFAT activation with the peptide VIVIT reduced neurosphere size and cell density in NPC cultures by decreasing proliferation and increasing cell death. VIVIT also decreased NPC migration and differentiation of astrocytes and neurons from NPCs. In addition, we identified NFATc3 as a predominant NFAT isoform in NPC cultures, finding that a constitutively-active form of NFATc3 expressed by adenoviral infection reduces NPC proliferation, stimulates migration, and is a potent inducer of NPC differentiation into astrocytes and neurons. In summary, our work uncovers active roles for NFAT signaling in NPC survival, proliferation and differentiation, and highlights its therapeutic potential for tissue regeneration.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Fatores de Transcrição NFATC/metabolismo , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiologia , Adesão Celular/fisiologia , Morte Celular/fisiologia , Células Cultivadas , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Transdução de Sinais , Nicho de Células-Tronco/fisiologia
5.
Glia ; 63(12): 2231-48, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26184558

RESUMO

The role and different origin of brain myeloid cells in the brain is central to understanding how the central nervous system (CNS) responds to injury. C-type lectin receptor family 9, member A (DNGR-1/CLEC9A) is a marker of specific DC subsets that share functional similarities, such as CD8α(+) DCs in lymphoid tissues and CD103(+) CD11b(low) DCs in peripheral tissues. Here, we analyzed the presence of DNGR-1 in DCs present in the mouse brain (bDCs). Dngr-1/Clec9a mRNA is expressed mainly in the meningeal membranes and choroid plexus (m/Ch), and its expression is enhanced by fms-like tyrosine kinase 3 ligand (Flt3L), a cytokine involved in DC homeostasis. Using Clec9a(egfp/egfp) mice, we show that Flt3L induces accumulation of DNGR-1-EGFP(+) cells in the brain m/Ch. Most of these cells also express major histocompatibility complex class II (MHCII) molecules. We also observed an increase in specific markers of cDC CD8α+ cells such as Batf-3 and Irf-8, but not of costimulatory molecules such as Cd80 and Cd86, indicating an immature phenotype for these bDCs in the noninjured brain. The presence of DNGR-1 in the brain provides a potential marker for the study of this specific brain cell subset. Knowledge and targeting of brain antigen presenting cells (APCs) has implications for the fight against brain diseases such as neuroinflammation-based neurodegenerative diseases, microbe-induced encephalitis, and brain tumors such as gliomas.


Assuntos
Plexo Corióideo/citologia , Células Dendríticas/citologia , Lectinas Tipo C/metabolismo , Meninges/citologia , Receptores Imunológicos/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Plexo Corióideo/metabolismo , Células Dendríticas/metabolismo , Genes MHC da Classe II/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fatores Reguladores de Interferon/metabolismo , Lectinas Tipo C/genética , Antígenos Comuns de Leucócito/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Meninges/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Receptores Imunológicos/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
6.
Glia ; 61(7): 1052-66, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23625833

RESUMO

Increase in intracellular calcium ([Ca(2+) ]i ) is a key mediator of astrocyte signaling, important for activation of the calcineurin (CN)/nuclear factor of activated T cells (NFAT) pathway, a central mediator of inflammatory events. We analyzed the expression of matrix metalloproteinase 3 (Mmp3) in response to increases in [Ca(2+) ]i and the role of the CN/NFAT pathway in this regulation. Astrocyte Mmp3 expression was induced by overexpression of a constitutively active form of NFATc3, whereas other MMPs and tissue inhibitor of metalloproteinases (TIMP) were unaffected. Mmp3 mRNA and protein expression was also induced by calcium ionophore (Io) and 2'(3')-O-(4-benzoylbenzoyl) adenosine 5'-triphosphate (Bz-ATP) and Mmp3 upregulation was prevented by the CN inhibitor cyclosporin A (CsA). Ca(2+) -dependent astrocyte Mmp3 expression was also inhibited by actinomycin D, and a Mmp3 promoter luciferase reporter was efficiently activated by increased [Ca(2+) ]i , indicating regulation at the transcriptional level. Furthermore, Ca(2+) /CN/NFAT dependent Mmp3 expression was confirmed in pure astrocyte cultures derived from neural stem cells (Ast-NSC), demonstrating that the induced Mmp3 expression occurs in astrocytes, and not microglial cells. In an in vivo stab-wound model of brain injury, MMP3 expression was detected in NFATc3-positive scar-forming astrocytes. Because [Ca(2+) ]i increase is an early event in most brain injuries, these data support an important role for Ca(2+) /CN/NFAT-induced astrocyte MMP3 expression in the early neuroinflammatory response. Understanding the molecular pathways involved in this regulation could provide novel therapeutic targets and approaches to promoting recovery of the injured brain.


Assuntos
Astrócitos/metabolismo , Cálcio/metabolismo , Regulação da Expressão Gênica/fisiologia , Metaloproteinase 3 da Matriz/metabolismo , Fatores de Transcrição NFATC/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Encéfalo/citologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Metaloproteinase 3 da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/genética , Dibutirato de 12,13-Forbol/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Tempo , Ferimentos e Lesões/patologia
7.
Biomater Adv ; 145: 213247, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36527961

RESUMO

Skin is the most extensive organ within our body. It is continually subjected to stress factors, among which ultraviolet irradiation, a key factor responsible in skin aging since it leads to reactive oxygen species production. In order to fight against these oxidative species, the human body has an innate robust antioxidant mechanism composed of several different substances, one of which is coenzyme Q10. Its capacity to increase cellular energy production and excellent antioxidant properties have been proved, as well as its antiaging properties being able to attenuate cellular damage induced by ultraviolet irradiation in human dermal fibroblasts. However, its high hydrophobicity and photolability hampers its therapeutic potential. In this context, the objective of this work consists of the preparation of chitosan-rosmarinic acid conjugate-based nanoparticles to encapsulate coenzyme Q10 with high encapsulation efficiencies in order to improve its bioavailability and broaden its therapeutic use in skin applications. Hyaluronic acid coating was performed giving stable nanoparticles at physiological pH with 382 ± 3 nm of hydrodynamic diameter (0.04 ± 0.02 polydispersity) and - 18 ± 3 mV of surface charge. Release kinetics studies showed a maximum of 82 % mass release of coenzyme Q10 after 40 min, and radical scavenger activity assay confirmed the antioxidant character of chitosan-rosmarinic acid nanoparticles. Hyaluronic acid-coated chitosan-rosmarinic acid nanoparticles loaded with coenzyme Q10 were biocompatible in human dermal fibroblasts and exhibited interesting photoprotective properties in ultraviolet irradiated cells. In addition, nanoparticles hindered the production of reactive oxygen species, interleukin-6 and metalloproteinase-1, as well as caspase-9 activation maintaining high viability values upon irradiation of dermal fibroblasts. Overall results envision a great potential of these nanovehicles for application in skin disorders or antiaging treatments.


Assuntos
Quitosana , Nanopartículas , Humanos , Antioxidantes/farmacologia , Ubiquinona/farmacologia , Ubiquinona/química , Espécies Reativas de Oxigênio , Quitosana/farmacologia , Quitosana/química , Ácido Hialurônico , Nanopartículas/química , Ácido Rosmarínico
8.
J Funct Biomater ; 14(3)2023 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-36976084

RESUMO

Current limitations of wound dressings for treating chronic wounds require the development of novel approaches. One of these is the immune-centered approach, which aims to restore the pro-regenerative and anti-inflammatory properties of macrophages. Under inflammatory conditions, ketoprofen nanoparticles (KT NPs) can reduce pro-inflammatory markers of macrophages and increase anti-inflammatory cytokines. To assess their suitability as part of wound dressings, these NPs were combined with hyaluronan (HA)/collagen-based hydro- (HGs) and cryogels (CGs). Different HA and NP concentrations and loading techniques for NP incorporation were used. The NP release, gel morphology, and mechanical properties were studied. Generally, colonialization of the gels with macrophages resulted in high cell viability and proliferation. Furthermore, direct contact of the NPs to the cells reduced the level of nitric oxide (NO). The formation of multinucleated cells on the gels was low and further decreased by the NPs. For the HGs that produced the highest reduction in NO, extended ELISA studies showed reduced levels of the pro-inflammatory markers PGE2, IL-12 p40, TNF-α, and IL-6. Thus, HA/collagen-based gels containing KT NPs may represent a novel therapeutic approach for treating chronic wounds. Whether effects observed in vitro translate into a favorable profile on skin regeneration in vivo will require rigorous testing.

9.
J Neuroinflammation ; 9: 48, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22397398

RESUMO

BACKGROUND: An increase in intracellular calcium concentration [Ca2+]i is one of the first events to take place after brain ischemia. A key [Ca2+]i-regulated signaling molecule is the phosphatase calcineurin (CN), which plays important roles in the modulation of inflammatory cascades. Here, we have analyzed the role of endogenous regulator of CN 1 (Rcan1) in response to experimental ischemic stroke induced by middle cerebral artery occlusion. METHODS: Animals were subjected to focal cerebral ischemia with reperfusion. To assess the role of Rcan1 after stroke, we measured infarct volume after 48 h of reperfusion in Rcan1 knockout (KO) and wild-type (WT) mice. In vitro studies were performed in astrocyte-enriched cortical primary cultures subjected to 3% oxygen (hypoxia) and glucose deprivation (HGD). Adenoviral vectors were used to analyze the effect of overexpression of Rcan1-4 protein. Protein expression was examined by immunohistochemistry and immunoblotting and expression of mRNA by quantitative real-time Reverse-Transcription Polymerase Chain Reaction (real time qRT-PCR). RESULTS: Brain ischemia/reperfusion (I/R) injury in vivo increased mRNA and protein expression of the calcium-inducible Rcan1 isoform (Rcan1-4). I/R-inducible expression of Rcan1 protein occurred mainly in astroglial cells, and in an in vitro model of ischemia, HGD treatment of primary murine astrocyte cultures induced Rcan1-4 mRNA and protein expression. Exogenous Rcan1-4 overexpression inhibited production of the inflammatory marker cyclo-oxygenase 2. Mice lacking Rcan1 had higher expression of inflammation associated genes, resulting in larger infarct volumes. CONCLUSIONS: Our results support a protective role for Rcan1 during the inflammatory response to stroke, and underline the importance of the glial compartment in the inflammatory reaction that takes place after ischemia. Improved understanding of non-neuronal mechanisms in ischemic injury promises novel approaches to the treatment of acute ischemic stroke.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/genética , Infarto da Artéria Cerebral Média/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Traumatismo por Reperfusão/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Encéfalo/patologia , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Proteínas de Ligação ao Cálcio , Hipóxia Celular/fisiologia , Células Cultivadas , Córtex Cerebral/citologia , Ciclo-Oxigenase 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Glucose/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Musculares/deficiência , Monoéster Fosfórico Hidrolases/metabolismo , RNA Mensageiro/genética , Ratos , Transfecção
10.
ACS Appl Mater Interfaces ; 14(8): 10068-10080, 2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35179869

RESUMO

Supramolecular peptide-based hydrogels attract great attention in several fields, i.e., biomedicine, catalysis, energy, and materials chemistry, due to the noncovalent nature of the self-assembly and functional tunable properties defined by the amino acid sequence. In this work, we developed an injectable hybrid supramolecular hydrogel whose formation was triggered by electrostatic interactions between a phosphorylated tripeptide, Fmoc-FFpY (F: phenylalanine, pY: phosphorylated tyrosine), and cationic polymer nanoparticles made of vinylimidazole and ketoprofen (poly(HKT-co-VI) NPs). Hydrogel formation was assessed through inverted tube tests, and its fibrillary structure, around polymer NPs, was observed by transmission electron microscopy. Interestingly, peptide self-assembly yields the formation of nontwisted and twisted fibers, which could be attributed to ß-sheets and α-helix structures, respectively, as characterized by circular dichroism and infrared spectroscopies. An increase of the elastic modulus of the Fmoc-FFpY/polymer NPs hybrid hydrogels was observed with peptide concentration as well as its injectability property, due to its shear thinning behavior and self-healing ability. After checking their stability under physiological conditions, the cytotoxicity properties of these hybrid hydrogels were evaluated in contact with human dermal fibroblasts (FBH) and murine macrophages (RAW 264.7). Finally, the Fmoc-FFpY/polymer NPs hybrid hydrogels exhibited a great nitric oxide reduction (∼67%) up to basal values of pro-inflammatory RAW 264.7 cells, thus confirming their excellent anti-inflammatory properties for the treatment of localized inflammatory pathologies.


Assuntos
Hidrogéis , Nanopartículas , Animais , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Camundongos , Peptídeos/química , Peptídeos/farmacologia , Fenilalanina , Polímeros
11.
Glia ; 59(1): 94-107, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20967884

RESUMO

Astrocytes react to brain injury triggering neuroinflammatory processes that determine the degree of neuronal damage. However, the signaling events associated to astrocyte activation remain largely undefined. The nuclear factor of activated T-cells (NFAT) is a transcription factor family implicated in activation of immune cells. We previously characterized the expression of NFAT isoforms in cultured astrocytes, and NFAT activation in response to mechanical lesion. Here we analyze NFATc3 in two mouse models of inflammatory brain damage: hippocampal excitotoxicity induced by intracerebral kainic acid (KA) injection and cortical mechanical lesion. Immunofluorescence results demonstrated that NFATc3 is specifically induced in a subset of reactive astrocytes, and not in microglia or neurons. In KA-treated brains, NFATc3 expression is transient and NFATc3-positive astrocytes concentrate around damaged neurons in areas CA3 and CA1. Complementary Western blot and RT-PCR analysis revealed an NFAT-dependent induction of RCAN1-4 and COX-2 in hippocampus as soon as 6 h after KA exposure, indicating that NFAT activation precedes NFATc3 over-expression. Moreover, activation of NFAT by ATP increased NFATc3 mRNA levels in astrocyte cultures, suggesting that NFATc3 expression is controlled through an auto-regulatory loop. Meanwhile, stab wound enhanced NFATc3 expression specifically in a subclass of reactive astrocytes confined within the proximal layer of the glial scar, and GFAP immunoreactivity was attenuated in NFATc3-expressing astrocytes. In conclusion, our work establishes NFATc3 as a marker of activation for a specific population of astrocytes in response to brain damage, which may have consequences for neuronal survival.


Assuntos
Astrócitos/metabolismo , Lesões Encefálicas/metabolismo , Fatores de Transcrição NFATC/metabolismo , Análise de Variância , Animais , Astrócitos/patologia , Western Blotting , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Morte Celular , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Ácido Caínico/farmacologia , Camundongos , Microscopia Confocal , Fatores de Transcrição NFATC/genética , Neurônios/metabolismo , Neurônios/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Aten Primaria ; 43(4): 176-82, 2011 Apr.
Artigo em Espanhol | MEDLINE | ID: mdl-20537430

RESUMO

OBJECTIVE: To investigate the habits and risk factors of adolescents from two Health Centres in two semi-urban populations using a structured clinical interview with open questions. DESIGN: Cross-sectional descriptive study. SETTING: Two semi-urban populations from the Malaga area. PARTICIPANTS: Adolescents aged between 16 and 18 years old. We selected 5 medical clinics out of the 19 clinics in the Health Centres, using stratified random sampling. A total of 204 adolescents were included, with 62 (30.39%) of them not attending. OUTCOMES: 42.3% were overweight or obese. The BMI and MBP ratio was R=0.4 They ate fruit, vegetables or dairy products at least once a day 54.2%, 57.8% and 24.5%, respectively. 32.3% of the male teenagers and 63.5% of females did not exercise regularly. 21.8% were smokers, and this was related to a low socio-economical level (OR: 3.38 P=0.001 95% CI: 1.27 to 9) and with abandoning education (OR: 2.88 P=0.015 CI 95%. 1.20 to 6,86). 56.3% usually drink and this habit was also related to abandoning education. (OR: 3.5 95% CI: 1.43 to 8.94). 10.6% of the teenagers consumed illegal substances and their group of friends in 36.6% of the cases. 12.1% had unprotected sex. 12.4% and 13.4% did not use a crash helmet or seat belt, respectively. 24.2% have driven in a drunken state at some point. 20.4% have felt depressed at least once. CONCLUSIONS: Risk factors and life style habits prevalent in reference to weight, fruit, vegetables and dairy products consumption, sport, smoking, alcohol and depression problems have been similar to the ones found in other studies that have used anonymous surveys. Prevalence of substance abuse has been lower.


Assuntos
Hábitos , Adolescente , Estudos Transversais , Feminino , Humanos , Masculino , Fatores de Risco , Espanha , População Suburbana , Saúde da População Urbana
13.
Carbohydr Polym ; 273: 118619, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-34561015

RESUMO

Rosmarinic acid is an attractive candidate for skin applications because of its antioxidant, anti-inflammatory, and photoprotective functions, however, its poor bioavailability hampers its therapeutic outcome. In this context, synthesis of polymer conjugates is an alternative to enlarge its applications. This work describes the synthesis of novel water-soluble chitosan - rosmarinic acid conjugates (CSRA) that have great potential for skin applications. Chitosan was functionalized with different contents of rosmarinic acid as confirmed by ATR-FTIR, 1H NMR and UV spectroscopies. CSRA conjugates presented three-fold radical scavenger capacity compared to the free phenolic compound. Films were prepared by solvent-casting procedure and the biological activity of the lixiviates was studied in vitro. Results revealed that lixiviates reduced activation of inflamed macrophages, improved antibacterial capacity against E. coli with respect to native chitosan and free rosmarinic acid, and also attenuated UVB-induced cellular damage and reactive oxygen species production in fibroblasts and keratinocytes.


Assuntos
Anti-Inflamatórios/farmacologia , Quitosana/farmacologia , Cinamatos/farmacologia , Depsídeos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Protetores contra Radiação/farmacologia , Animais , Anti-Inflamatórios/síntese química , Anti-Inflamatórios/toxicidade , Quitosana/análogos & derivados , Quitosana/toxicidade , Cinamatos/síntese química , Cinamatos/toxicidade , Depsídeos/síntese química , Depsídeos/toxicidade , Escherichia coli/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Sequestradores de Radicais Livres/síntese química , Sequestradores de Radicais Livres/toxicidade , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Óxido Nítrico/metabolismo , Células RAW 264.7 , Protetores contra Radiação/síntese química , Protetores contra Radiação/toxicidade , Staphylococcus epidermidis/efeitos dos fármacos , Ácido Rosmarínico
14.
Mater Sci Eng C Mater Biol Appl ; 124: 112024, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33947532

RESUMO

Cytotoxic chemotherapy continues to be the main therapeutic option for patients with metastatic breast cancer. Several studies have reported a significant association between chronic inflammation, carcinogenesis and the presence of cancer stem cells (CSC). We hypothesized that the use of non-steroidal anti-inflammatory drugs targeted to the CSC population could help reducing tumor progression and dissemination in otherwise hard to treat metastatic breast cancer. Within this study cationic naproxen (NAP)-bearing polymeric nanoparticles (NPs) were obtained by self-assembly and they were coated with hyaluronic acid (HA) via electrostatic interaction. HA-coated and uncoated NAP-bearing NPs with different sizes were produced by changing the ionic strength of the aqueous preparation solutions (i.e. 300 and 350 nm or 100 and 130 nm in diameter, respectively). HA-NPs were fully characterized in terms of physicochemical parameters and biological response in cancer cells, macrophages and endothelial cells. Our results revealed that HA-coating of NPs provided a better control in NAP release and improved their hemocompatibility, while ensuring a strong CSC-targeting in MCF-7 breast cancer cells. Furthermore, the best polymeric NPs formulation significantly (p < 0.001) reduced MCF-7 cells viability when compared to free drug (i.e. 45 ± 6% for S-HA-NPs and 87 ± 10% for free NAP) by p53-dependent induction of apoptosis; and the migration of these cell line was also significantly (p < 0.01) reduced by the nano-formulated NAP (i.e. 76.4% of open wound for S-HA-NPs and 61.6% of open wound for NAP). This increased anti-cancer activity of HA-NAP-NPs might be related to the induction of apoptosis through alterations of the GSK-3ß-related COX-independent pathway. Overall, these findings suggest that the HA-NAP-NPs have the potential to improve the treatment of advanced breast cancer by increasing the anti-proliferative effect of NAP within the CSC subpopulation.


Assuntos
Antineoplásicos , Neoplasias da Mama , Nanopartículas , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Células Endoteliais , Glicogênio Sintase Quinase 3 beta , Humanos , Receptores de Hialuronatos , Ácido Hialurônico , Naproxeno/farmacologia , Células-Tronco Neoplásicas
15.
Macromol Biosci ; 20(7): e2000002, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32421256

RESUMO

Recent studies have demonstrated in vivo synergistic immunosuppressive and anti-inflammatory capacity of dexamethasone (Dx) and naproxen (NAP) in collagen-induced arthritis (CIA) rats. However, the molecular basis of this synergistic effect is barely understood. The low solubility of these drugs and their adverse effects hamper their efficacy on the treatment of inflammatory processes making nanoparticulated systems promising candidates to overcome these drawbacks. The aim of this work is the preparation of polymeric nanoparticles (NPs) that combine NAP and Dx in different concentrations, and the evaluation of the expression of key genes related to autoimmune diseases like CIA. To do so, self-assembled polymeric NPs that incorporate covalently-linked NAP and physically entrapped Dx are designed to have hydrodynamic properties that, according to bibliography, may improve retention and colocalization of both drugs at inflammation sites. The rapid uptake of NPs by macrophages is demonstrated using coumarine-6-loaded NPs. Dx is efficiently encapsulated and in vitro biological studies demonstrate that the Dx-loaded NAP-bearing NPs are noncytotoxic and reduce lipopolysaccharide-induced NO released levels at any of the tested concentrations. Moreover, at the molecular level, a significant synergistic reduction of Il12b transcript gene expression when combining Dx and NAP is demonstrated.


Assuntos
Dexametasona/farmacologia , Macrófagos/metabolismo , Nanopartículas/química , Naproxeno/farmacologia , Polímeros/química , Inibidores da Angiogênese/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Morte Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Hidrodinâmica , Interações Hidrofóbicas e Hidrofílicas , Subunidade p40 da Interleucina-12 , Macrófagos/efeitos dos fármacos , Camundongos , Peso Molecular , Nanopartículas/toxicidade , Nanopartículas/ultraestrutura , Naproxeno/síntese química , Naproxeno/química , Óxido Nítrico/metabolismo , Espectroscopia de Prótons por Ressonância Magnética , Células RAW 264.7
16.
Pharmaceutics ; 12(8)2020 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-32751993

RESUMO

Polymeric nanoparticles that combine dexamethasone and naproxen reduce inflammation and synergistically inhibit Interleukin-12b (Il12b) transcription in macrophages. This effect can be the result of a cyclooxygenase-dependent or a cyclooxygenase-independent mechanism. The aim of this work is to obtain potent anti-inflammatory polymeric nanoparticles by the combination of dexamethasone and ketoprofen, one of the most efficient cyclooxygenase-inhibitors among non-steroidal anti-inflammatory drugs, with appropriate hydrodynamic properties to facilitate accumulation and co-release of drugs in inflamed tissue. Nanoparticles are spherical with hydrodynamic diameter (117 ± 1 nm), polydispersity (0.139 ± 0.004), and surface charge (+30 ± 1 mV), which confer them with high stability and facilitate both macrophage uptake and internalization pathways to favor their retention at the inflamed areas and lysosomal degradation and drug release, respectively. In vitro biological studies concluded that the dexamethasone-loaded ketoprofen-bearing system is non-cytotoxic and efficiently reduces lipopolysaccharide-induced nitric oxide release. The RT-qPCR analysis shows that the ketoprofen nanoparticles were able to reduce to almost basal levels the expression of tested pro-inflammatory markers and increase the gene expression of anti-inflammatory cytokines under inflammatory conditions. However, the synergistic inhibition of Il12b observed in nanoparticles that combine dexamethasone and naproxen was not observed in nanoparticles that combine dexamethasone and ketoprofen, suggesting that the synergistic trans-repression of Il12b observed in the first case was not mediated by cyclooxygenase-dependent pathways.

17.
Front Immunol ; 11: 2120, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042124

RESUMO

Streptococcus pneumoniae is the main cause of bacterial pneumonia, a condition that currently produces significant global morbidity and mortality. The initial immune response to this bacterium occurs when the innate system recognizes common motifs expressed by many pathogens, events driven by pattern recognition receptors like the Toll-like family receptors (TLRs). In this study, lung myeloid-cell populations responsible for the innate immune response (IIR) against S. pneumoniae, and their dependence on the TLR4-signaling axis, were analyzed in TLR4-/- and Myeloid-Differentiation factor-88 deficient (MyD88-/-) mice. Neutrophils and monocyte-derived cells were recruited in infected mice 3-days post-infection. Compared to wild-type mice, there was an increased bacterial load in both these deficient mouse strains and an altered IIR, although TLR4-/- mice were more susceptible to bacterial infection. These mice also developed fewer alveolar macrophages, weaker neutrophil infiltration, less Ly6Chigh monocyte differentiation and a disrupted classical and non-classical monocyte profile. The pro-inflammatory cytokine profile (CXCL1, TNF-α, IL-6, and IL-1ß) was also severely affected by the lack of TLR4 and no induction of Th1 was observed in these mice. The respiratory burst (ROS production) after infection was profoundly dampened in TLR4-/- and MyD88-/- mice. These data demonstrate the complex dynamics of myeloid populations and a key role of the TLR4-signaling axis in the IIR to S. pneumoniae, which involves both the MyD88 and TRIF (Toll/IL-1R domain-containing adaptor-inducing IFN-ß) dependent pathways.


Assuntos
Pulmão/imunologia , Monócitos/imunologia , Fator 88 de Diferenciação Mieloide/fisiologia , Mielopoese/fisiologia , Pneumonia Pneumocócica/imunologia , Pneumonia Pneumocócica/patologia , Transdução de Sinais/fisiologia , Streptococcus pneumoniae/imunologia , Receptor 4 Toll-Like/fisiologia , Administração Intranasal , Animais , Carga Bacteriana , Citocinas/biossíntese , Imunidade Inata , Pulmão/patologia , Macrófagos Alveolares/imunologia , Camundongos , Monócitos/patologia , Fator 88 de Diferenciação Mieloide/deficiência , Infiltração de Neutrófilos , Espécies Reativas de Oxigênio/metabolismo , Células Th1/imunologia , Receptor 4 Toll-Like/deficiência
18.
Cell Rep ; 33(3): 108291, 2020 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33086061

RESUMO

Brain CD11c+ cells share features with microglia and dendritic cells (DCs). Sterile inflammation increases brain CD11c+ cells, but their phenotype, origin, and functions remain largely unknown. We report that, after cerebral ischemia, microglia attract DCs to the inflamed brain, and astroglia produce Flt3 ligand, supporting development and expansion of CD11c+ cells. CD11c+ cells in the inflamed brain are a complex population derived from proliferating microglia and infiltrating DCs, including a major subset of OX40L+ conventional cDC2, and also cDC1, plasmacytoid, and monocyte-derived DCs. Despite sharing certain morphological features and markers, CD11c+ microglia and DCs display differential expression of pattern recognition receptors and chemokine receptors. DCs excel CD11c- and CD11c+ microglia in the capacity to present antigen through MHCI and MHCII. Of note, cDC1s protect from brain injury after ischemia. We thus reveal aspects of the dynamics and functions of brain DCs in the regulation of inflammation and immunity.


Assuntos
Antígenos CD11/metabolismo , Células Dendríticas/metabolismo , Microglia/metabolismo , Animais , Antígenos/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Antígenos CD11/genética , Antígeno CD11c/genética , Antígeno CD11c/metabolismo , Citocinas/metabolismo , Células Dendríticas/fisiologia , Encefalite/imunologia , Encefalite/metabolismo , Citometria de Fluxo , Inflamação/imunologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Monócitos/metabolismo , Receptores de Quimiocinas/metabolismo
19.
Mol Neurobiol ; 56(2): 1248-1261, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29881946

RESUMO

Amyloid precursor protein (APP) is implicated in neural development as well as in the pathology of Alzheimer's disease (AD); however, its biological function still remains unclear. It has been reported that APP stimulates the proliferation and neuronal differentiation of neural stem cells (NSCs), while other studies suggest an important effect enhancing gliogenesis in NSCs. As expected, APP protein/mRNA is detected in hNS1 cells, a model cell line of human NSCs, both under proliferation and throughout the differentiation period. To investigate the potential function that APP plays in cell fate specification and differentiation of hNS1 cells, we transiently increased human APP levels in these cells and analyzed its cell intrinsic effects. Our data indicate that increased levels of APP induce early cell cycle exit and instructively direct hNS1 cell fate towards a glial phenotype, while decreasing neuronal differentiation. Since elevated APP levels also enhanced APP intracellular domain (AICD)-immunoreactivity, these effects could be, in part, mediated by the APP/AICD system. The AICD domain can play a potential role in signal transduction by its molecular interaction with different target genes such as GSK3B, whose expression was also increased in APP-overexpressing cells that, in turn, may contribute to promoting gliogenesis and inhibiting neurogenesis in NSCs. These data suggest an important action of APP in modulating hNSCs differentiation (probably in an AICD-GSK-3ß-dependent manner) and may thus be important for the future development of stem cell therapy strategies for the diseased mammalian brain.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Neuroglia/metabolismo , Neurônios/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem Celular , Humanos , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neurônios/citologia
20.
Sci Rep ; 9(1): 9361, 2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31249342

RESUMO

Calcium/Calcineurin/Nuclear Factor of Activated T cells (Ca/CN/NFAT) signalling pathway is the main calcium (Ca2+) dependent signalling pathway involved in the homeostasis of brain tissue. Here, we study the presence of NFATc members in human glioma by using U251 cells and a collection of primary human glioblastoma (hGB) cell lines. We show that NFATc3 member is the predominant member. Furthermore, by using constitutive active NFATc3 mutant and shRNA lentiviral vectors to achieve specific silencing of this NFATc member, we describe cytokines and molecules regulated by this pathway which are required for the normal biology of cancer cells. Implanting U251 in an orthotopic intracranial assay, we show that specific NFATc3 silencing has a role in tumour growth. In addition NFATc3 knock-down affects both the proliferation and migration capacities of glioma cells in vitro. Our data open the possibility of NFATc3 as a target for the treatment of glioma.


Assuntos
Astrocitoma/genética , Fatores de Transcrição NFATC/genética , Animais , Astrocitoma/metabolismo , Astrocitoma/patologia , Biomarcadores , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular/genética , Proliferação de Células/genética , Modelos Animais de Doenças , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Xenoenxertos , Humanos , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Fatores de Transcrição NFATC/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA