Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 212(2): 245-257, 2024 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-38047900

RESUMO

CD8 T cells are emerging as important mediators in atherosclerosis and cardiovascular disease (CVD). Immune activation may play a particular role in people with HIV (PWH) who are at an increased risk of CVD, even after controlling for known CVD risk factors. Latent CMV infection is associated with increased CVD risk for both PWH and people without HIV, and human CMV-specific CD4 and CD8 T cells are enriched for an immunosenescent phenotype. We previously showed that CMV coinfection in PWH promotes vascular homing and activation of inflammatory CD4 T cells through the CD2-LFA-3 axis. However, the role of CD2/LFA3 costimulation of CD8 T cells in PWH with CMV has yet to be described. In the present study, we demonstrate that CD2 expression on CX3CR1+CD57+CD28- inflammescent CD8 T cells is increased on cells from CMV-seropositive PWH. In vitro CD2/LFA-3 costimulation enhances TCR-mediated activation of these inflammatory CD8 memory T cells. Finally, we show that LFA-3 is highly expressed in aortas of SIV-infected rhesus macaques and in atherosclerotic plaques of people without HIV. Our findings are consistent with a model in which CMV infection enhances CD2 expression on highly proinflammatory CD8 T cells that can then be stimulated by LFA-3 expressed in the vasculature, even in the absence of CD28 costimulation. This model, in which CMV infection exacerbates toxic cytokine and granzyme production by CD8 T cells within the vasculature, highlights a potential therapeutic target in atherosclerosis development and progression, especially for PWH.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Infecções por Citomegalovirus , Infecções por HIV , Animais , Humanos , Antígenos CD28/metabolismo , Infecções por HIV/tratamento farmacológico , Citomegalovirus , Antígenos CD58/metabolismo , Macaca mulatta , Linfócitos T CD8-Positivos , Linfócitos T CD4-Positivos , Aterosclerose/metabolismo
2.
PLoS Pathog ; 16(9): e1008885, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32976527

RESUMO

Atherosclerotic cardiovascular disease (ASCVD) remains an important cause of morbidity in the general population and risk for ASCVD is increased approximately 2-fold in persons living with HIV infection (PLWH). This risk is linked to elevated CD8 T cell counts that are abundant in atherosclerotic plaques and have been implicated in disease pathogenesis yet the mechanisms driving T cell recruitment to and activation within plaques are poorly defined. Here we investigated the role of CD8 T cells in atherosclerosis in a non-human primate model of HIV infection and in the HIV-uninfected elderly; we sought to identify factors that promote the activation, function, and recruitment to endothelium of CX3CR1+ CD8 T cells. We measured elevated expression of CX3CL1 and IL-15, and increased CD8 T cell numbers in the aortas of rhesus macaques infected with SIV or SHIV, and demonstrated similar findings in atherosclerotic vessels of HIV-uninfected humans. We found that recombinant TNF enhanced the production and release of CX3CL1 and bioactive IL-15 from aortic endothelial cells, but not from aortic smooth muscle cells. IL-15 in turn promoted CX3CR1 surface expression on and TNF synthesis by CD8 T cells, and IL-15-treated CD8 T cells exhibited enhanced CX3CL1-dependent chemoattraction toward endothelial cells in vitro. Finally, we show that CD8 T cells in human atherosclerotic plaques have an activated, resident phenotype consistent with in vivo IL-15 and CX3CL1 exposure. In this report, we define a novel model of CD8 T cell involvement in atherosclerosis whereby CX3CL1 and IL-15 operate in tandem within the vascular endothelium to promote infiltration by activated CX3CR1+ memory CD8 T cells that drive further endothelial activation via TNF. We propose that these interactions are prevalent in aging and in PLWH, populations where circulating activated CX3CR1+ CD8 T cell numbers are often expanded.


Assuntos
Aterosclerose/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Quimiocina CX3CL1/metabolismo , Infecções por HIV/metabolismo , Interleucina-15/metabolismo , Idoso , Animais , Células Endoteliais/metabolismo , Humanos , Macaca mulatta/metabolismo , Receptores de Quimiocinas/metabolismo
3.
J Immunol ; 204(10): 2722-2733, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32229536

RESUMO

Cytotoxic CD4 T cells are linked to cardiovascular morbidities and accumulate in both HIV and CMV infections, both of which are associated with increased risk of cardiovascular disease (CVD). In this study, we identify CMV coinfection as a major driver of the cytotoxic phenotype, characterized by elevated CD57 expression and reduced CD28 expression, in circulating CD4 T cells from people living with HIV infection, and investigate potential mechanisms linking this cell population to CVD. We find that human CD57+ CD4 T cells express high levels of the costimulatory receptor CD2 and that CD2/LFA-3 costimulation results in a more robust and polyfunctional effector response to TCR signals, compared with CD28-mediated costimulation. CD57+ CD4 T cells also express the vascular endothelium-homing receptor CX3CR1 and migrate toward CX3CL1-expressing endothelial cells in vitro. IL-15 promotes the cytotoxic phenotype, elevates CX3CR1 expression, and enhances the trafficking of CD57+ CD4 T cells to endothelium and may therefore be important in linking these cells to cardiovascular complications. Finally, we demonstrate the presence of activated CD57+ CD4 T cells and expression of CX3CL1 and LFA-3 in atherosclerotic plaque tissues from HIV-uninfected donors. Our findings are consistent with a model in which cytotoxic CD4 T cells contribute to CVD in HIV/CMV coinfection and in atherosclerosis via CX3CR1-mediated trafficking and CD2/LFA-3-mediated costimulation. This study identifies several targets for therapeutic interventions and may help bridge the gap in understanding how CMV infection and immunity are linked to increased cardiovascular risk in people living with HIV infection.


Assuntos
Vasos Sanguíneos/fisiologia , Linfócitos T CD4-Positivos/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/fisiologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Placa Aterosclerótica/imunologia , Antígenos CD28/metabolismo , Antígenos CD57/metabolismo , Antígenos CD58/metabolismo , Receptor 1 de Quimiocina CX3C/metabolismo , Movimento Celular , Quimiocina CX3CL1/metabolismo , Coinfecção , Citotoxicidade Imunológica , Humanos , Receptores CXCR3/metabolismo , Risco
4.
Eur J Immunol ; 50(12): 2055-2066, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32548862

RESUMO

Recent studies have implicated a role for adenosine-dependent immunosuppression in head and neck tumor microenvironments. We describe expression of CD73, an enzyme critical to the generation of adenosine from extracellular AMP, in T cells and other cell types within human head and neck tumors. Flow cytometric analyses of tumor-infiltrating cells indicate that CD3+ cells are the predominant source of CD73 among immune infiltrating cells and that CD73 expression, especially among CD8+ T cells, is inversely related to indices of T cell infiltration and T cell activation in the microenvironment of head and neck tumors. We provide evidence that CD73 expression on peripheral T cells and levels of soluble CD73 in circulation are correlated with CD73 expression on CD8+ T cells in tumors. Moreover, fluorescent microscopy studies reveal that CD8+ CD73+ cells are observed in close proximity to tumor cells as well as in surrounding tissue. In vitro studies with peripheral blood T cells indicate that anti-CD3-stimulation causes loss of CD73 expression, especially among cells that undergo proliferation and that exogenous AMP can impair T cell proliferation, while sustaining CD73 expression. These data suggest that CD8+ CD73+ T cells may be especially important mediators of immunosuppression in human head and neck cancer.


Assuntos
5'-Nucleotidase/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias de Cabeça e Pescoço/imunologia , Linfócitos do Interstício Tumoral/imunologia , Microambiente Tumoral/imunologia , Idoso , Idoso de 80 Anos ou mais , Proliferação de Células/fisiologia , Feminino , Proteínas Ligadas por GPI/imunologia , Humanos , Tolerância Imunológica/imunologia , Ativação Linfocitária/imunologia , Masculino , Pessoa de Meia-Idade
5.
Curr HIV/AIDS Rep ; 18(3): 198-210, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33709322

RESUMO

PURPOSE OF REVIEW: Systemic inflammation increases as a consequence of aging (inflammaging) and contributes to age-related morbidities. Inflammation in people living with HIV is elevated compared with the general population even after prolonged suppression of viremia with anti-retroviral therapy. Mechanisms that contribute to inflammation during aging and in treated HIV disease are potentially interactive, leading to an exaggerated inflammatory phenotype in people with HIV. RECENT FINDINGS: Recent studies highlight roles for anti-retroviral therapy, co-infections, immune system alterations, and microbiome perturbations as important contributors to HIV-associated inflammation. These factors likely contribute to increased risk of age-related morbidities in people living with HIV. Understanding mechanisms that exaggerate the inflammaging process in people with HIV may lead to improved intervention strategies, ultimately, extending both lifespan and healthspan.


Assuntos
Infecções por HIV , Envelhecimento , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Humanos , Inflamação , Fenótipo
6.
J Infect Dis ; 220(1): 73-77, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-30698729

RESUMO

Circulating CD8+ T cells and monocytes are activated during human immunodeficiency virus (HIV) infection and colocalize in the aortas of simian immunodeficiency virus-infected nonhuman primates. We hypothesized that CD8+ T cells could exert a proatherosclerotic effect via paracrine actions on monocytes. We found that T-cell receptor-stimulated CD8+ T cells induce monocytes to express tissue factor, a potent activator of coagulation. Tumor necrosis factor was both necessary and sufficient for this effect.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Monócitos/imunologia , Tromboplastina/imunologia , Fator de Necrose Tumoral alfa/imunologia , Coagulação Sanguínea/imunologia , Células Cultivadas , Células Endoteliais/imunologia , Infecções por HIV/imunologia , Humanos , Receptores de Antígenos de Linfócitos T/imunologia
7.
Blood ; 127(21): 2618-29, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-27015965

RESUMO

A prothrombotic state and increased platelet reactivity are common in dyslipidemia and oxidative stress. Lipid peroxidation, a major consequence of oxidative stress, generates highly reactive products, including hydroxy-ω-oxoalkenoic acids that modify autologous proteins generating biologically active derivatives. Phosphatidylethanolamine, the second most abundant eukaryotic phospholipid, can also be modified by hydroxy-ω-oxoalkenoic acids. However, the conditions leading to accumulation of such derivatives in circulation and their biological activities remain poorly understood. We now show that carboxyalkylpyrrole-phosphatidylethanolamine derivatives (CAP-PEs) are present in the plasma of hyperlipidemic ApoE(-/-) mice. CAP-PEs directly bind to TLR2 and induces platelet integrin αIIbß3 activation and P-selectin expression in a Toll-like receptor 2 (TLR2)-dependent manner. Platelet activation by CAP-PEs includes assembly of TLR2/TLR1 receptor complex, induction of downstream signaling via MyD88/TIRAP, phosphorylation of IRAK4, and subsequent activation of tumor necrosis factor receptor-associated factor 6. This in turn activates the Src family kinases, spleen tyrosine kinase and PLCγ2, and platelet integrins. Murine intravital thrombosis studies demonstrated that CAP-PEs accelerate thrombosis in TLR2-dependent manner and that TLR2 contributes to accelerate thrombosis in mice in the settings of hyperlipidemia. Our study identified the novel end-products of lipid peroxidation, accumulating in circulation in hyperlipidemia and inducing platelet activation by promoting cross-talk between innate immunity and integrin activation signaling pathways.


Assuntos
Apolipoproteínas E/deficiência , Plaquetas/metabolismo , Hiperlipidemias/metabolismo , Fosfatidiletanolaminas/metabolismo , Ativação Plaquetária , Trombose/metabolismo , Receptor 2 Toll-Like/metabolismo , Animais , Hiperlipidemias/genética , Hiperlipidemias/patologia , Quinases Associadas a Receptores de Interleucina-1/genética , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fosfatidiletanolaminas/genética , Fosforilação/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Trombose/genética , Trombose/patologia , Receptor 1 Toll-Like/genética , Receptor 1 Toll-Like/metabolismo , Receptor 2 Toll-Like/genética
8.
J Immunol ; 194(10): 4698-704, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25870237

RESUMO

IL-7 is a key homeostatic cytokine that provides signals for T cell survival and proliferation in vivo. In this article, we provide evidence that IL-7 utilization is enhanced by a novel mechanism of cytokine "recycling" during which T cells treated with rIL-7 are rapidly induced to express p-STAT5 and are subsequently able to recycle biologically active cytokine for release to neighboring cells in soluble form. Our observations indicate that the ability of cells to recycle IL-7 is dependent on IL-7R α-chain (CD127) and endocytosis, consistent with a model whereby IL-7 is internalized via receptor interactions before recycling. These observations provide evidence of a novel mechanism that enables cells to optimally use IL-7.


Assuntos
Interleucina-7/imunologia , Interleucina-7/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Ativação Linfocitária/imunologia
9.
J Infect Dis ; 214 Suppl 2: S51-7, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27625431

RESUMO

Untreated human immunodeficiency virus (HIV) infection is characterized by progressive CD4(+) T-cell depletion and CD8(+) T-cell expansion, and CD4(+) T-cell depletion is linked directly to the risk for opportunistic infections and infection-associated mortality. With suppression of HIV replication by antiretroviral therapy, circulating CD4(+) Tcell numbers typically improve while CD8(+) T-cell expansion persists, and both CD4(+) T-cell cytopenia and CD8(+) T-cell expansion are associated with morbidity and mortality. In this brief review, we report on the role that selected homeostatic and inflammatory cytokines may play both in the failure of CD4(+) T-cell restoration and the CD8(+) T-cell expansion that characterize HIV infection.


Assuntos
Citocinas/imunologia , Infecções por HIV/imunologia , Linfócitos T/imunologia , Animais , Homeostase , Humanos
10.
J Infect Dis ; 213(9): 1419-27, 2016 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-26671887

RESUMO

BACKGROUND: Human immunodeficiency virus (HIV) infection is associated with increased risk of thromboembolic and cardiovascular comorbid conditions. Although systemic inflammation is linked to cardiovascular risk, direct evidence of vascular inflammation and endothelial dysfunction is lacking. METHODS: We examined by immunofluorescence microscopy thoracic aortas from 16 simian immunodeficiency virus (SIV)- or simian-human immunodeficiency virus (SHIV)-infected and 16 uninfected rhesus macaques. RESULTS: Focal endothelial proliferation and subendothelial inflammatory cells were found in sections of all infected animals, compared with minimal changes in sections from the 16 uninfected controls. In the infected animals, we detected increased endothelial levels of bacterial 16s ribosomal DNA as well as increased subendothelial accumulation of CD68(+) monocytes/macrophages (P< .001) and CD8(+) T lymphocytes (P< .001). Endothelial dysfunction was manifested by decreased levels of endothelial nitric oxide synthase (P< .005) and Krüppel-like factor 2 (KLF2) (P< .005). KLF2 expression was decreased in primary human aortic endothelial cells exposed to bacterial lipopolysaccharide or to oxidized low-density lipoprotein in vitro, and this could be prevented by simvastatin. CONCLUSIONS: SIV and SHIV infection lead to endothelial inflammation, dysfunction, and decreased KLF2 expression reflecting early atherosclerotic changes. Translocated bacterial components and lipid oxidation products may induce endothelial dysfunction in HIV infection that could be prevented by statin treatment.


Assuntos
Endotélio Vascular/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Fatores de Transcrição Kruppel-Like/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Vasculite/imunologia , Animais , Aorta Torácica/citologia , Aorta Torácica/imunologia , Aorta Torácica/microbiologia , Células Cultivadas , DNA Bacteriano/análise , Endotélio Vascular/microbiologia , Infecções por HIV/fisiopatologia , Humanos , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/fisiopatologia
11.
J Infect Dis ; 214(12): 1808-1816, 2016 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-27703039

RESUMO

Increases in inflammation, coagulation, and CD8+ T-cell numbers are associated with an elevated cardiovascular disease (CVD) risk in human immunodeficiency virus (HIV)-infected antiretroviral therapy (ART) recipients. Circulating memory CD8+ T cells that express the vascular endothelium-homing receptor CX3CR1 (fractalkine receptor) are enriched in HIV-infected ART recipients. Thrombin-activated receptor (PAR-1) expression is increased in HIV-infected ART recipients and is particularly elevated on CX3CR1+ CD8+ T cells, suggesting that these cells could interact with coagulation elements. Indeed, thrombin directly enhanced T-cell receptor-mediated interferon γ production by purified CD8+ T cells but was attenuated by thrombin-induced release of transforming growth factor ß by platelets. We have therefore identified a population of circulating memory CD8+ T cells in HIV infection that may home to endothelium, can be activated by clot-forming elements, and are susceptible to platelet-mediated regulation. Complex interactions between inflammatory elements and coagulation at endothelial surfaces may play an important role in CVD risk in HIV-infected ART recipients.


Assuntos
Plaquetas/metabolismo , Linfócitos T CD8-Positivos/imunologia , Infecções por HIV/patologia , Receptores de Quimiocinas/análise , Subpopulações de Linfócitos T/imunologia , Linfócitos T CD8-Positivos/química , Linfócitos T CD8-Positivos/efeitos dos fármacos , Receptor 1 de Quimiocina CX3C , Infecções por HIV/imunologia , Humanos , Subpopulações de Linfócitos T/química , Subpopulações de Linfócitos T/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
12.
Biochim Biophys Acta ; 1853(3): 604-10, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25541282

RESUMO

Monitoring of autophagy is challenging because of its multiple steps and lack of single befitting technique for a complete mechanistic understanding, which makes the task complicated. Here, we evaluate the functionality of autophagy triggered by salinomycin (anti-cancer stem cell agent) using flow cytometry and advanced microscopy. We show that salinomycin does induce functional autophagy at lower concentrations and such a dose is cell type-dependent. For example, PC3 cells show active autophagic flux at 10 µM concentration of salinomycin while murine embryonic fibroblasts already show an inhibition of flux at such doses. A higher concentration of salinomycin (i.e. 30 µM) inhibits autophagic flux in both cell types. The data confirms our previous findings that salinomycin is an inducer of autophagy, whereas autophagic flux inhibition is a secondary response.


Assuntos
Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Citometria de Fluxo/métodos , Piranos/farmacologia , Animais , Rastreamento de Células/métodos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Camundongos
13.
Clin Infect Dis ; 62(3): 392-6, 2016 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-26400999

RESUMO

BACKGROUND: Persistent CD8 T-cell expansion, low CD4/CD8 T-cell ratios, and heightened inflammation persist in antiretroviral therapy (ART)-treated human immunodeficiency virus (HIV) infection and are associated with increased risk of morbid outcomes. We explored the role of cytomegalovirus (CMV) infection in CD8 lymphocytosis and inflammation in ART-treated HIV infection. METHODS: Absolute CD4 and CD8 T-cell counts were abstracted from clinical records and compared among 32 HIV-infected CMV-seronegative subjects, 126 age, CD4 and gender-matched HIV-infected CMV-seropositive subjects, and among 21 HIV-uninfected controls (9 CMV-negative, 12 CMV-positive). Plasma inflammatory indices were measured in a subset by ELISA. RESULTS: Median CD8 counts/µL were higher in HIV-positive/CMV-positive patients (795) than in HIV-positive/CMV-negative subjects (522, P = .006) or in healthy controls (451, P = .0007), whereas CD8 T-cell counts were similar to controls' levels in HIV-positive/CMV-negative subjects. Higher plasma levels of IP-10 (P = .0011), TNF-RII (P = .0002), and D-dimer (P = .0444) were also found in coinfected patients than in HIV-positive/CMV-negative subjects. CONCLUSIONS: CMV infection is associated with higher CD8 T-cell counts, resultant lower CD4/CD8 ratios, and increased systemic inflammation in ART-treated HIV infection. CMV infection may contribute to risk for morbid outcomes in treated HIV infection.


Assuntos
Antirretrovirais/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Coinfecção/patologia , Infecções por Citomegalovirus/patologia , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Adulto , Relação CD4-CD8 , Linfócitos T CD4-Positivos/imunologia , Estudos de Coortes , Citocinas/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Infecções por HIV/patologia , Humanos , Contagem de Linfócitos , Linfocitose/patologia , Masculino , Pessoa de Meia-Idade , Plasma/química
14.
Circ Res ; 112(1): 103-12, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23071157

RESUMO

RATIONALE: A prothrombotic state and increased platelet reactivity are common in pathophysiological conditions associated with oxidative stress and infections. Such conditions are associated with an appearance of altered-self ligands in circulation that can be recognized by Toll-like receptors (TLRs). Platelets express a number of TLRs, including TLR9; however, the role of TLR in platelet function and thrombosis is poorly understood. OBJECTIVE: To investigate the biological activities of carboxy(alkylpyrrole) protein adducts, an altered-self ligand generated in oxidative stress, on platelet function and thrombosis. METHODS AND RESULTS: In this study we show that carboxy(alkylpyrrole) protein adducts represent novel unconventional ligands for TLR9. Furthermore, using human and murine platelets, we demonstrate that carboxy(alkylpyrrole) protein adducts promote platelet activation, granule secretion, and aggregation in vitro and thrombosis in vivo via the TLR9/MyD88 pathway. Platelet activation by TLR9 ligands induces IRAK1 and AKT phosphorylation, and it is Src kinase-dependent. Physiological platelet agonists act synergistically with TLR9 ligands by inducing TLR9 expression on the platelet surface. CONCLUSIONS: Our study demonstrates that platelet TLR9 is a functional platelet receptor that links oxidative stress, innate immunity, and thrombosis.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária , Albumina Sérica/metabolismo , Trombose/sangue , Receptor Toll-Like 9/sangue , Animais , Plaquetas/imunologia , Antígenos CD36/deficiência , Antígenos CD36/genética , Linhagem Celular , Modelos Animais de Doenças , Genes Reporter , Humanos , Imunidade Inata , Quinases Associadas a Receptores de Interleucina-1/sangue , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Estresse Oxidativo , Fosfatidilinositol 3-Quinase/sangue , Fosforilação , Agregação Plaquetária , Proteínas Proto-Oncogênicas c-akt/sangue , Receptores Depuradores Classe B/deficiência , Receptores Depuradores Classe B/genética , Transdução de Sinais , Trombose/genética , Trombose/imunologia , Fatores de Tempo , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/genética , Receptor 6 Toll-Like/deficiência , Receptor 6 Toll-Like/genética , Receptor Toll-Like 9/deficiência , Receptor Toll-Like 9/genética , Transfecção , Quinases da Família src/sangue
15.
Biochim Biophys Acta ; 1833(9): 2057-69, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23639289

RESUMO

The molecular mechanism of Salinomycin's toxicity is not fully understood. Various studies reported that Ca(2+), cytochrome c, and caspase activation play a role in Salinomycin-induced cytotoxicity. Furthermore, Salinomycin may target Wnt/ß-catenin signaling pathway to promote differentiation and thus elimination of cancer stem cells. In this study, we show a massive autophagic response to Salinomycin (substantially stronger than to commonly used autophagic inducer Rapamycin) in prostrate-, breast cancer cells, and to lesser degree in human normal dermal fibroblasts. Interestingly, autophagy induced by Salinomycin is a cell protective mechanism in all tested cancer cell lines. Furthermore, Salinomycin induces mitophagy, mitoptosis and increased mitochondrial membrane potential (∆Ψ) in a subpopulation of cells. Salinomycin strongly, and in time-dependent manner decreases cellular ATP level. Contrastingly, human normal dermal fibroblasts treated with Salinomycin show some initial decrease in mitochondrial mass, however they are largely resistant to Salinomycin-triggered ATP-depletion. Our data provide new insight into the molecular mechanism of preferential toxicity of Salinomycin towards cancer cells, and suggest possible clinical application of Salinomycin in combination with autophagy inhibitors (i.e. clinically-used Chloroquine). Furthermore, we discuss preferential Salinomycins toxicity in the context of Warburg effect.


Assuntos
Antibacterianos/farmacologia , Autofagia/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Citotoxinas/farmacologia , Derme/metabolismo , Fibroblastos/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitofagia/efeitos dos fármacos , Neoplasias da Próstata/metabolismo , Piranos/farmacologia , Trifosfato de Adenosina/genética , Trifosfato de Adenosina/metabolismo , Animais , Autofagia/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Derme/patologia , Feminino , Fibroblastos/patologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Mitofagia/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Via de Sinalização Wnt/efeitos dos fármacos
16.
iScience ; 26(9): 107706, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37694142

RESUMO

Direct recognition of Mycobacterium tuberculosis (Mtb)-infected cells is required for protection by CD4+ T cells. While impaired T cell recognition of Mtb-infected macrophages was demonstrated in mice, data are lacking for humans. Using T cells and monocyte-derived macrophages (MDMs) from individuals with latent Mtb infection (LTBI), we quantified the frequency of memory CD4+ T cell activation in response to autologous MDMs infected with virulent Mtb. We observed robust T cell activation in response to Mtb infection of M1-like macrophages differentiated using GM-CSF, while M2-like macrophages differentiated using M-CSF were poorly recognized. However, non-infected GM-CSF and M-CSF MDMs loaded with exogenous antigens elicited similar CD4+ T cell activation. IL-10 was preferentially secreted by infected M-CSF MDMs, and neutralization improved T cell activation. These results suggest that preferential infection of macrophages with an M2-like phenotype limits T cell-mediated protection against Mtb. Vaccine development should focus on T cell recognition of Mtb-infected macrophages.

17.
Front Mol Biosci ; 9: 824954, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35355507

RESUMO

While platelets are the essential mediators of hemostasis, they are being increasingly recognized for their potential of contributing to host defenses. Here, using immunofluorescent microscopy, western blot, and ELISA, we found that human ß-defensin 3 (hBD-3), an important antimicrobial peptide produced by epithelial cells, can be detected in human platelets and megakaryocytes. Flow cytometry and immuno-electron microscopy revealed hBD-3 on the surface of thrombin activated platelets. Moreover, hBD-3 was also found in platelet derived extracellular vesicles (p-EVs), isolated from platelet poor plasma and from platelet supernatants following thrombin stimulation. Incubation of platelets with hBD-3 peptide resulted in modest platelet activation and pre-incubation of platelets with synthetic hBD-3 prior to exposure to thrombin appeared to increase hBD-3 content in platelet lysates as well as in p-EVs, suggesting that hBD-3 can be initially taken up by platelets, perhaps via their open canalicular system. Interestingly, in vitro exposure of primary human endothelial cells to either hBD-3 peptide or purified p-EVs, caused significant endothelial dysfunction as documented by diminished levels of phosphorylated endothelial nitric oxide synthase (eNOS), Krüppel like factor-2 (KLF-2), and elevated relative expression of von Willebrand Factor (vWF). Pre-incubation of platelets with hBD-3 appeared to augment endothelial dysfunction caused by p-EVs. Overall, the current study provides evidence that hBD-3 enriched EVs can be released by activated platelets and may play a role in positive feedback of platelet activation as well as in endothelial dysfunction. Theoretically, these effects could contribute to both cellular recruitment to the endothelium creating a pro-thrombotic vascular microenvironment which serve as a bridge between innate immunity and hemostasis.

19.
Microbiol Spectr ; 9(3): e0073521, 2021 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-34935423

RESUMO

SARS-CoV-2 infection can cause compromised respiratory function and thrombotic events. SARS-CoV-2 binds to and mediates downregulation of angiotensin converting enzyme 2 (ACE2) on cells that it infects. Theoretically, diminished enzymatic activity of ACE2 may result in increased concentrations of pro-inflammatory molecules, angiotensin II, and Bradykinin, contributing to SARS-CoV-2 pathology. Using immunofluorescence microscopy of lung tissues from uninfected, and SARS-CoV-2 infected individuals, we find evidence that ACE2 is highly expressed in human pulmonary alveolar epithelial cells and significantly reduced along the alveolar lining of SARS-CoV-2 infected lungs. Ex vivo analyses of primary human cells, indicated that ACE2 is readily detected in pulmonary alveolar epithelial and aortic endothelial cells. Exposure of these cells to spike protein of SARS-CoV-2 was sufficient to reduce ACE2 expression. Moreover, exposure of endothelial cells to spike protein-induced dysfunction, caspase activation, and apoptosis. Exposure of endothelial cells to bradykinin caused calcium signaling and endothelial dysfunction (increased expression of von Willibrand Factor and decreased expression of Krüppel-like Factor 2) but did not adversely affect viability in primary human aortic endothelial cells. Computer-assisted analyses of molecules with potential to bind bradykinin receptor B2 (BKRB2), suggested a potential role for aspirin as a BK antagonist. When tested in our in vitro model, we found evidence that aspirin can blunt cell signaling and endothelial dysfunction caused by bradykinin in these cells. Interference with interactions of spike protein or bradykinin with endothelial cells may serve as an important strategy to stabilize microvascular homeostasis in COVID-19 disease. IMPORTANCE SARS-CoV-2 causes complex effects on microvascular homeostasis that potentially contribute to organ dysfunction and coagulopathies. SARS-CoV-2 binds to, and causes downregulation of angiotensin converting enzyme 2 (ACE2) on cells that it infects. It is thought that reduced ACE2 enzymatic activity can contribute to inflammation and pathology in the lung. Our studies add to this understanding by providing evidence that spike protein alone can mediate adverse effects on vascular cells. Understanding these mechanisms of pathogenesis may provide rationale for interventions that could limit microvascular events associated with SARS-CoV-2 infection.


Assuntos
COVID-19/fisiopatologia , Células Endoteliais/virologia , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Epiteliais Alveolares/citologia , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/virologia , Enzima de Conversão de Angiotensina 2/química , Enzima de Conversão de Angiotensina 2/genética , Enzima de Conversão de Angiotensina 2/metabolismo , Aorta/citologia , Aorta/metabolismo , Aorta/virologia , Apoptose , Bradicinina/química , Bradicinina/metabolismo , COVID-19/genética , COVID-19/metabolismo , COVID-19/virologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Homeostase , Humanos , Pulmão/irrigação sanguínea , Pulmão/metabolismo , Pulmão/virologia , Microcirculação , Receptores da Bradicinina/química , Receptores da Bradicinina/genética , Receptores da Bradicinina/metabolismo , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética
20.
Biochim Biophys Acta ; 1793(8): 1335-42, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19374922

RESUMO

Apoptin, a small protein from chicken anemia virus, has attracted great attention, because it specifically kills tumor cells while leaving normal cells unharmed. The subcellular localization of apoptin appears to be crucial for this tumor-selective activity. In normal cells, apoptin resides in the cytoplasm, whereas in cancerous cells it translocates into the nucleus. The nuclear translocation of apoptin is largely controlled by its phosphorylation. In tumor cells, apoptin causes the nuclear accumulation of survival kinases including Akt and is phosphorylated by CDK2. Thereby, apoptin redirects survival signals into cell death responses. Apoptin also binds as a multimeric complex to DNA and interacts with several nuclear targets, such as the anaphase-promoting complex, resulting in a G2/M phase arrest. The proapoptotic signal of apoptin is then transduced from the nucleus to cytoplasm by Nur77, which triggers a p53-independent mitochondrial death pathway. In this review, we summarize recent discoveries of apoptin's mechanism of action that might provide intriguing insights for the development of novel tumor-selective anticancer drugs.


Assuntos
Proteínas do Capsídeo/fisiologia , Vírus da Anemia da Galinha/fisiologia , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Animais , Apoptose/fisiologia , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Vírus da Anemia da Galinha/química , Vírus da Anemia da Galinha/genética , Vírus da Anemia da Galinha/patogenicidade , Galinhas , Infecções por Circoviridae/patologia , Infecções por Circoviridae/veterinária , Infecções por Circoviridae/virologia , Genes Virais , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , Sarcoma Aviário/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA