Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(7)2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-37047304

RESUMO

Sickle cell disease (SCD) is an inherited blood disorder caused by a ß-globin gene point mutation that results in the production of sickle hemoglobin that polymerizes upon deoxygenation, causing the sickling of red blood cells (RBCs). RBC deformation initiates a sequence of events leading to multiple complications, such as hemolytic anemia, vaso-occlusion, chronic inflammation, and tissue damage. Macrophages participate in extravascular hemolysis by removing damaged RBCs, hence preventing the release of free hemoglobin and heme, and triggering inflammation. Upon erythrophagocytosis, macrophages metabolize RBC-derived hemoglobin, activating mechanisms responsible for recycling iron, which is then used for the generation of new RBCs to try to compensate for anemia. In the bone marrow, macrophages can create specialized niches, known as erythroblastic islands (EBIs), which regulate erythropoiesis. Anemia and inflammation present in SCD may trigger mechanisms of stress erythropoiesis, intensifying RBC generation by expanding the number of EBIs in the bone marrow and creating new ones in extramedullary sites. In the current review, we discuss the distinct mechanisms that could induce stress erythropoiesis in SCD, potentially shifting the macrophage phenotype to an inflammatory profile, and changing their supporting role necessary for the proliferation and differentiation of erythroid cells in the disease. The knowledge of the soluble factors, cell surface and intracellular molecules expressed by EBI macrophages that contribute to begin and end the RBC's lifespan, as well as the understanding of their signaling pathways in SCD, may reveal potential targets to control the pathophysiology of the disease.


Assuntos
Anemia Falciforme , Linfo-Histiocitose Hemofagocítica , Humanos , Eritropoese , Eritrócitos , Macrófagos/metabolismo , Inflamação/metabolismo
2.
Photodiagnosis Photodyn Ther ; 39: 103015, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35843562

RESUMO

Ultraviolet (UV) light can inactivate SARS-CoV-2. However, the practicality of UV light is limited by the carcinogenic potential of mercury vapor-based UV lamps. Recent advances in the development of krypton chlorine (KrCl) excimer lamps hold promise, as these emit a shorter peak wavelength (222 nm), which is highly absorbed by the skin's stratum corneum and can filter out higher wavelengths. In this sense, UV 222 nm irradiation for the inactivation of virus particles in the air and surfaces is a potentially safer option as a germicidal technology. However, these same physical properties make it harder to reach microbes present in complex solutions, such as saliva, a critical source of SARS-CoV-2 transmission. We provide the first evaluation for using a commercial filtered KrCl excimer light source to inactivate SARS-CoV-2 in saliva spread on a surface. A conventional germicidal lamp (UV 254 nm) was also evaluated under the same condition. Using plaque-forming units (PFU) and Median Tissue Culture Infectious Dose (TCID50) per milliliter we found that 99.99% viral clearance (LD99.99) was obtained with 106.3 mJ/cm2 of UV 222 nm for virus in DMEM and 2417 mJ/cm2 for virus in saliva. Additionally, our results showed that the UV 254 nm had a greater capacity to inactivate the virus in both vehicles. Effective (after discounting light absorption) LD99.99 of UV 222 nm on the virus in saliva was ∼30 times higher than the value obtained with virus in saline solution (PBS), we speculated that saliva might be protecting the virus from surface irradiation in ways other than just by intensity attenuation of UV 222 nm. Due to differences between UV 222/254 nm capacities to interact and be absorbed by molecules in complex solutions, a higher dose of 222 nm will be necessary to reduce viral load in surfaces with contaminated saliva.


Assuntos
COVID-19 , Fotoquimioterapia , Desinfecção/métodos , Humanos , Fotoquimioterapia/métodos , SARS-CoV-2 , Saliva , Raios Ultravioleta
3.
Viruses ; 13(11)2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34834934

RESUMO

A SARS-CoV-2 B.1.1.7 variant of concern (VOC) has been associated with increased transmissibility, hospitalization, and mortality. This study aimed to explore the factors associated with B.1.1.7 VOC infection in the context of vaccination. On March 2021, we detected SARS-CoV-2 RNA in nasopharyngeal samples from 14 of 22 individuals vaccinated with a single-dose of ChAdOx1 (outbreak A, n = 26), and 22 of 42 of individuals with two doses of the CoronaVac vaccine (outbreak B, n = 52) for breakthrough infection rates for ChAdOx1 of 63.6% and 52.4% for CoronaVac. The outbreaks were caused by two independent clusters of the B.1.1.7 VOC. The serum of PCR-positive symptomatic SARS-CoV-2-infected individuals had ~1.8-3.4-fold more neutralizing capacity against B.1.1.7 compared to the serum of asymptomatic individuals. These data based on exploratory analysis suggest that the B.1.1.7 variant can infect individuals partially immunized with a single dose of an adenovirus-vectored vaccine or fully immunized with two doses of an inactivated vaccine, although the vaccines were able to reduce the risk of severe disease and death caused by this VOC, even in the elderly.


Assuntos
Vacinas contra COVID-19 , COVID-19/imunologia , COVID-19/virologia , SARS-CoV-2/classificação , SARS-CoV-2/genética , Vacinação , Adenoviridae , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Neutralizantes/imunologia , Brasil/epidemiologia , COVID-19/prevenção & controle , Teste Sorológico para COVID-19 , Estudos de Coortes , Surtos de Doenças/estatística & dados numéricos , Feminino , Vetores Genéticos , Humanos , Imunoglobulina G/sangue , Masculino , Pessoa de Meia-Idade , RNA Viral , Vacinas de Produtos Inativados , Sequenciamento Completo do Genoma , Adulto Jovem
4.
J Immunol ; 204(8): 2257-2268, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32169845

RESUMO

Plasmacytoid dendritic cells (pDCs) produce abundant type I IFNs (IFN-I) in response to viral nucleic acids. Generation of pDCs from bone marrow dendritic cell (DC) progenitors and their maintenance is driven by the transcription factor E2-2 and inhibited by its repressor Id2. In this study, we find that mouse pDCs selectively express the receptor for LIF that signals through STAT3. Stimulation of pDCs with LIF inhibited IFN-I, TNF, and IL-6 responses to CpG and induced expression of the STAT3 targets SOCS3 and Bcl3, which inhibit IFN-I and NF-κB signaling. Moreover, although STAT3 has been also reported to induce E2-2, LIF paradoxically induced its repressor Id2. A late-stage bone marrow DC progenitor expressed low amounts of LIFR and developed into pDCs less efficiently after being exposed to LIF, consistent with the induction of Id2. Conversely, pDC development and serum IFN-I responses to lymphocytic choriomeningitis virus infection were augmented in newly generated mice lacking LIFR in either CD11c+ or hematopoietic cells. Thus, an LIF-driven STAT3 pathway induces SOCS3, Bcl3, and Id2, which render pDCs and late DC progenitors refractory to physiological stimuli controlling pDC functions and development. This pathway can be potentially exploited to prevent inappropriate secretion of IFN-I in autoimmune diseases or promote IFN-I secretion during viral infections.


Assuntos
Células Dendríticas/citologia , Células Dendríticas/imunologia , Fator Inibidor de Leucemia/metabolismo , Animais , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Transcrição STAT3/imunologia , Transdução de Sinais/imunologia
5.
Nat Commun ; 9(1): 1513, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29666415

RESUMO

Chagas disease is caused by infection with the protozoan Trypanosoma cruzi (T. cruzi) and is an important cause of severe inflammatory heart disease. However, the mechanisms driving Chagas disease cardiomyopathy have not been completely elucidated. Here, we show that the canonical PI3Kγ pathway is upregulated in both human chagasic hearts and hearts of acutely infected mice. PI3Kγ-deficient mice and mutant mice carrying catalytically inactive PI3Kγ are more susceptible to T. cruzi infection. The canonical PI3Kγ signaling in myeloid cells is essential to restrict T. cruzi heart parasitism and ultimately to avoid myocarditis, heart damage, and death of mice. Furthermore, high PIK3CG expression correlates with low parasitism in human Chagas' hearts. In conclusion, these results indicate an essential role of the canonical PI3Kγ signaling pathway in the control of T. cruzi infection, providing further insight into the molecular mechanisms involved in the pathophysiology of chagasic heart disease.


Assuntos
Cardiomiopatia Chagásica/imunologia , Classe Ib de Fosfatidilinositol 3-Quinase/metabolismo , Transdução de Sinais/imunologia , Trypanosoma cruzi/imunologia , Adulto , Animais , Biópsia , Linhagem Celular , Cardiomiopatia Chagásica/parasitologia , Cardiomiopatia Chagásica/patologia , Classe Ib de Fosfatidilinositol 3-Quinase/genética , Modelos Animais de Doenças , Feminino , Coração/parasitologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Células Mieloides/imunologia , Células Mieloides/metabolismo , Miocárdio/imunologia , Miocárdio/patologia , Inibidores de Fosfoinositídeo-3 Quinase , Quinoxalinas/farmacologia , Tiazolidinedionas/farmacologia , Trypanosoma cruzi/patogenicidade , Regulação para Cima
6.
J Virol ; 89(9): 4720-37, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25717109

RESUMO

UNLABELLED: Oropouche virus (OROV) is a member of the Orthobunyavirus genus in the Bunyaviridae family and a prominent cause of insect-transmitted viral disease in Central and South America. Despite its clinical relevance, little is known about OROV pathogenesis. To define the host defense pathways that control OROV infection and disease, we evaluated OROV pathogenesis and immune responses in primary cells and mice that were deficient in the RIG-I-like receptor signaling pathway (MDA5, RIG-I, or MAVS), downstream regulatory transcription factors (IRF-3 or IRF-7), beta interferon (IFN-ß), or the receptor for type I IFN signaling (IFNAR). OROV replicated to higher levels in primary fibroblasts and dendritic cells lacking MAVS signaling, the transcription factors IRF-3 and IRF-7, or IFNAR than in wild-type (WT) cells. In mice, deletion of IFNAR, MAVS, or IRF-3 and IRF-7 resulted in uncontrolled OROV replication, hypercytokinemia, extensive liver damage, and death, whereas WT congenic animals failed to develop disease. Unexpectedly, mice with a selective deletion of IFNAR on myeloid cells (CD11c Cre(+) Ifnar(f/f) or LysM Cre(+) Ifnar(f/f)) did not sustain enhanced disease with OROV or a selective (flox/flox) deletion La Crosse virus, a closely related encephalitic orthobunyavirus. In bone marrow chimera studies, recipient irradiated Ifnar(-/-) mice reconstituted with WT hematopoietic cells sustained high levels of OROV replication and liver damage, whereas WT mice reconstituted with Ifnar(-/-) bone marrow were resistant to disease. Collectively, these results establish a dominant protective role for MAVS, IRF-3 and IRF-7, and IFNAR in restricting OROV infection and tissue injury and suggest that IFN signaling in nonmyeloid cells contributes to the host defense against orthobunyaviruses. IMPORTANCE: Oropouche virus (OROV) is an emerging arthropod-transmitted orthobunyavirus that causes episodic outbreaks of a debilitating febrile illness in humans in countries of South and Central America. The continued expansion of the range and number of its arthropod vectors increases the likelihood that OROV will spread into new regions. At present, the pathogenesis of OROV in humans or other vertebrate animals remains poorly understood. To define cellular mechanisms of control of OROV infection, we performed infection studies in a series of primary cells and mice that were deficient in key innate immune genes involved in pathogen recognition and control. Our results establish that a MAVS-dependent type I IFN signaling pathway has a dominant role in restricting OROV infection and pathogenesis in vivo.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Orthobunyavirus/imunologia , Orthobunyavirus/fisiologia , Transdução de Sinais , Animais , Infecções por Bunyaviridae/patologia , Infecções por Bunyaviridae/virologia , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos/imunologia , Fibroblastos/virologia , Camundongos Endogâmicos C57BL , Análise de Sobrevida
7.
PLoS Negl Trop Dis ; 7(8): e2370, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23991231

RESUMO

A multi-step cascade strategy using integrated ligand- and target-based virtual screening methods was developed to select a small number of compounds from the ZINC database to be evaluated for trypanocidal activity. Winnowing the database to 23 selected compounds, 12 non-covalent binding cruzain inhibitors with affinity values (K i) in the low micromolar range (3-60 µM) acting through a competitive inhibition mechanism were identified. This mechanism has been confirmed by determining the binding mode of the cruzain inhibitor Nequimed176 through X-ray crystallographic studies. Cruzain, a validated therapeutic target for new chemotherapy for Chagas disease, also shares high similarity with the mammalian homolog cathepsin L. Because increased activity of cathepsin L is related to invasive properties and has been linked to metastatic cancer cells, cruzain inhibitors from the same library were assayed against it. Affinity values were in a similar range (4-80 µM), yielding poor selectivity towards cruzain but raising the possibility of investigating such inhibitors for their effect on cell proliferation. In order to select the most promising enzyme inhibitors retaining trypanocidal activity for structure-activity relationship (SAR) studies, the most potent cruzain inhibitors were assayed against T. cruzi-infected cells. Two compounds were found to have trypanocidal activity. Using compound Nequimed42 as precursor, an SAR was established in which the 2-acetamidothiophene-3-carboxamide group was identified as essential for enzyme and parasite inhibition activities. The IC50 value for compound Nequimed42 acting against the trypomastigote form of the Tulahuen lacZ strain was found to be 10.6±0.1 µM, tenfold lower than that obtained for benznidazole, which was taken as positive control. In addition, by employing the strategy of molecular simplification, a smaller compound derived from Nequimed42 with a ligand efficiency (LE) of 0.33 kcal mol(-1) atom(-1) (compound Nequimed176) is highlighted as a novel non-peptidic, non-covalent cruzain inhibitor as a trypanocidal agent candidate for optimization.


Assuntos
Antiprotozoários/isolamento & purificação , Avaliação Pré-Clínica de Medicamentos/métodos , Proteínas de Protozoários/antagonistas & inibidores , Antiprotozoários/farmacologia , Cristalografia por Raios X , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Concentração Inibidora 50 , Cinética , Testes de Sensibilidade Parasitária/métodos , Ligação Proteica , Conformação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Relação Estrutura-Atividade , Trypanosoma cruzi/efeitos dos fármacos , Trypanosoma cruzi/enzimologia
8.
Biomed Pharmacother ; 65(5): 334-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21775091

RESUMO

Synthesis, characterization and biological studies of silver and gold complexes with 6-mercaptopurine (H2MP) are described. The Ag(I) and Au(I) complexes with HMP-, AgHMP and AuIHMP, were obtained by mixing an acidified H2MP aqueous solution with an equimolar aqueous solution of AgNO3 or Au(CN)2. The Au(III) complex with HMP-, AuIIIHMP, was obtained by adding an aqueous solution of K(AuCl4) to an acidified H2MP aqueous solution (1:1 molar ratio) and the final solution was acidified with HCl to pH=1.0. The Au(III)MP complex, KAu(MP)2, was obtained by adding an aqueous solution of K(AuCl4) to a basic H2MP solution (M:L - 1:2 molar ratio). Formulas for the complexes are: (Ag[C5H3N4S])*½H2O for AgHMP, (Au[C5H3N4S]) for AuIHMP, (Au[C5H3N4S][Cl]2)*2H2O for AuIIIHMP and K(Au[C5H2N4S]2)·2H2O for KAu(MP)2. The AuIHMP and KAu(MP)2 complexes decreased cell viability of HeLa cancer cells in vitro. The IC50 values for AuIHMP and KAu(MP)2 are 3.0 and 30.0 µM, respectively. Anti-M.tuberculosis assays showed a MIC value of 2.24 µM for AuIHMP and 5.12 µM for free MP while AgHMP is active at the concentration 93.2 µM.


Assuntos
Antineoplásicos/farmacologia , Antituberculosos/farmacologia , Complexos de Coordenação/síntese química , Complexos de Coordenação/farmacologia , Ouro/química , Mercaptopurina/análogos & derivados , Prata/química , Antineoplásicos/síntese química , Antineoplásicos/química , Antituberculosos/síntese química , Antituberculosos/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Complexos de Coordenação/química , Células HeLa , Humanos , Concentração Inibidora 50 , Iodetos/química , Mercaptopurina/síntese química , Mercaptopurina/química , Mercaptopurina/farmacologia , Mycobacterium tuberculosis/efeitos dos fármacos , Compostos Organoáuricos/síntese química , Compostos Organoáuricos/química , Compostos Organoáuricos/farmacologia , Compostos de Prata/química , Soluções/química , Espectroscopia de Luz Próxima ao Infravermelho/métodos
9.
Neuroimmunomodulation ; 17(6): 379-85, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20516719

RESUMO

OBJECTIVE: We subjected mice to acute cold stress and studied the effect on phagocytosis by peritoneal macrophages mediated by 3 types of phagocytic receptors: Fcgamma, complement receptors 3 (CR3) and mannose and beta-glucan receptors. METHODS: Mice were subjected to a cold stress condition (4 degrees C for 4 h), and then peritoneal macrophages were harvested and phagocytosis assays performed in vitro. RESULTS: We found a striking difference between resting and lipopolysaccharide (LPS)-activated macrophages (by intraperitoneal injection of LPS 4 days before the stress experiment): for resting macrophages cold stress caused a decrease in phagocytosis mediated by Fcgamma or mannose receptors, while for activated macrophages we observed an increase in phagocytosis by the 3 types of receptors. These effects were associated with an increase in plasma concentrations of corticosterone and catecholamines following the cold stress. In order to verify whether these hormone changes could account for the observed effects on phagocytosis, we performed in vitro assays by incubating macrophages harvested from nonstressed animals with these hormones for 4 h at 37 degrees C and measuring their phagocytic capacity. The following experiments were done: (a) with resting (nonactivated) macrophages; (b) with macrophages previously activated in vitro by incubation with LPS; (c) with macrophages previously activated in vivo by intraperitoneal injection of mice with LPS, 4 days before harvesting the cells. We found that for resting macrophages, corticosterone decreased phagocytosis mediated by Fcgamma and mannose and beta-glucan receptors, but catecholamines had no effect. For macrophages activated either in vivo or in vitro, catecholamines caused an increase in phagocytosis (excluding mannose receptors) while corticosterone had no effect. CONCLUSION: The above findings suggest that stress can regulate phagocytosis in different ways, depending on the kind of phagocytic receptor involved, the level of stress hormones and the physiological state of the macrophages.


Assuntos
Catecolaminas/farmacologia , Temperatura Baixa , Corticosterona/farmacologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Fase de Repouso do Ciclo Celular/imunologia , Estresse Fisiológico/imunologia , Animais , Catecolaminas/sangue , Catecolaminas/fisiologia , Células Cultivadas , Temperatura Baixa/efeitos adversos , Corticosterona/sangue , Corticosterona/fisiologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neuroimunomodulação/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/efeitos dos fármacos
10.
Neuroimmunomodulation ; 17(2): 79-87, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19923852

RESUMO

BACKGROUND AND AIMS: Stress can alter many aspects of the immune response, and many studies have been conducted on the effects of stress on inflammatory processes, but little is known about its influence on the resolution of inflammation in tissue homeostasis, which includes the clearance of apoptotic cells by macrophages in a non-phlogistic way. In the present study, we investigated the effect of acute cold stress on the phagocytosis of apoptotic cells by macrophages. METHODS: Mice were submitted to acute cold stress (4 degrees C for 4 h) and the capacity of peritoneal macrophages to phagocyte apoptotic thymocytes and to secrete anti-inflammatory cytokines was evaluated. Plasma corticosterone and catecholamine levels were investigated to assess their effect on the phagocytic capacity of macrophages in vitro. RESULTS: We showed that acute cold stress decreases phagocytosis of apoptotic cells at the inflammatory site by lipopolysaccharide-activated macrophages but did not affect resting macrophages. The inhibitory effect on phagocytosis is accompanied by a reduced level of TGF-beta and higher IL-10 secretion. After stress, plasma concentrations of corticosterone increased 6-fold, epinephrine 2-fold and norepinephrine 1.7-fold compared to control mice. In vitro experiments showed that the decrease in phagocytosis after stress could be attributed, at least in part, to the effects of corticosterone; epinephrine and norepinephrine had no effect. CONCLUSIONS: The current study shows that acute cold stress decreases phagocytosis of apoptotic cells from an inflammatory environment by macrophages, and this inhibition is mediated by the intracellular glucocorticoid receptor.


Assuntos
Apoptose/imunologia , Corticosterona/metabolismo , Macrófagos/imunologia , Fagocitose/imunologia , Estresse Fisiológico/imunologia , Linfócitos T/imunologia , Doença Aguda , Animais , Apoptose/efeitos dos fármacos , Catecolaminas/metabolismo , Temperatura Baixa/efeitos adversos , Modelos Animais de Doenças , Epinefrina/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/fisiopatologia , Mediadores da Inflamação/farmacologia , Interleucina-10/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Norepinefrina/metabolismo , Receptores de Glucocorticoides/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA