Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Mol Cell ; 82(13): 2385-2400.e9, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35594856

RESUMO

Inflammation observed in SARS-CoV-2-infected patients suggests that inflammasomes, proinflammatory intracellular complexes, regulate various steps of infection. Lung epithelial cells express inflammasome-forming sensors and constitute the primary entry door of SARS-CoV-2. Here, we describe that the NLRP1 inflammasome detects SARS-CoV-2 infection in human lung epithelial cells. Specifically, human NLRP1 is cleaved at the Q333 site by multiple coronavirus 3CL proteases, which triggers inflammasome assembly and cell death and limits the production of infectious viral particles. Analysis of NLRP1-associated pathways unveils that 3CL proteases also inactivate the pyroptosis executioner Gasdermin D (GSDMD). Subsequently, caspase-3 and GSDME promote alternative cell pyroptosis. Finally, analysis of pyroptosis markers in plasma from COVID-19 patients with characterized severe pneumonia due to autoantibodies against, or inborn errors of, type I interferons (IFNs) highlights GSDME/caspase-3 as potential markers of disease severity. Overall, our findings identify NLRP1 as a sensor of SARS-CoV-2 infection in lung epithelia.


Assuntos
COVID-19 , Proteases 3C de Coronavírus , Células Epiteliais , Inflamassomos , Proteínas NLR , SARS-CoV-2 , COVID-19/genética , COVID-19/metabolismo , COVID-19/virologia , Caspase 3/metabolismo , Proteases 3C de Coronavírus/genética , Proteases 3C de Coronavírus/metabolismo , Células Epiteliais/metabolismo , Humanos , Inflamassomos/genética , Inflamassomos/metabolismo , Pulmão/metabolismo , Pulmão/virologia , Proteínas NLR/genética , Proteínas NLR/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Proteínas de Ligação a Fosfato/genética , Proteínas de Ligação a Fosfato/metabolismo , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Piroptose , SARS-CoV-2/enzimologia , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade
2.
Nature ; 617(7960): 386-394, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37100912

RESUMO

Inflammation is a complex physiological process triggered in response to harmful stimuli1. It involves cells of the immune system capable of clearing sources of injury and damaged tissues. Excessive inflammation can occur as a result of infection and is a hallmark of several diseases2-4. The molecular bases underlying inflammatory responses are not fully understood. Here we show that the cell surface glycoprotein CD44, which marks the acquisition of distinct cell phenotypes in the context of development, immunity and cancer progression, mediates the uptake of metals including copper. We identify a pool of chemically reactive copper(II) in mitochondria of inflammatory macrophages that catalyses NAD(H) redox cycling by activating hydrogen peroxide. Maintenance of NAD+ enables metabolic and epigenetic programming towards the inflammatory state. Targeting mitochondrial copper(II) with supformin (LCC-12), a rationally designed dimer of metformin, induces a reduction of the NAD(H) pool, leading to metabolic and epigenetic states that oppose macrophage activation. LCC-12 interferes with cell plasticity in other settings and reduces inflammation in mouse models of bacterial and viral infections. Our work highlights the central role of copper as a regulator of cell plasticity and unveils a therapeutic strategy based on metabolic reprogramming and the control of epigenetic cell states.


Assuntos
Plasticidade Celular , Cobre , Inflamação , Transdução de Sinais , Animais , Camundongos , Cobre/metabolismo , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , NAD/metabolismo , Transdução de Sinais/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Peróxido de Hidrogênio/metabolismo , Epigênese Genética/efeitos dos fármacos , Metformina/análogos & derivados , Oxirredução , Plasticidade Celular/efeitos dos fármacos , Plasticidade Celular/genética , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética
3.
PLoS Pathog ; 19(8): e1011559, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37619220

RESUMO

Mycobacterium abscessus (Mabs) drives life-shortening mortality in cystic fibrosis (CF) patients, primarily because of its resistance to chemotherapeutic agents. To date, our knowledge on the host and bacterial determinants driving Mabs pathology in CF patient lung remains rudimentary. Here, we used human airway organoids (AOs) microinjected with smooth (S) or rough (R-)Mabs to evaluate bacteria fitness, host responses to infection, and new treatment efficacy. We show that S Mabs formed biofilm, and R Mabs formed cord serpentines and displayed a higher virulence. While Mabs infection triggers enhanced oxidative stress, pharmacological activation of antioxidant pathways resulted in better control of Mabs growth and reduced virulence. Genetic and pharmacological inhibition of the CFTR is associated with better growth and higher virulence of S and R Mabs. Finally, pharmacological activation of antioxidant pathways inhibited Mabs growth, at least in part through the quinone oxidoreductase NQO1, and improved efficacy in combination with cefoxitin, a first line antibiotic. In conclusion, we have established AOs as a suitable human system to decipher mechanisms of CF-driven respiratory infection by Mabs and propose boosting of the NRF2-NQO1 axis as a potential host-directed strategy to improve Mabs infection control.


Assuntos
Fibrose Cística , Mycobacterium abscessus , Humanos , Fibrose Cística/tratamento farmacológico , Antioxidantes , Oxirredução , Estresse Oxidativo
4.
PLoS Pathog ; 18(7): e1010305, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35849616

RESUMO

Multiple regulated neutrophil cell death programs contribute to host defense against infections. However, despite expressing all necessary inflammasome components, neutrophils are thought to be generally defective in Caspase-1-dependent pyroptosis. By screening different bacterial species, we found that several Pseudomonas aeruginosa (P. aeruginosa) strains trigger Caspase-1-dependent pyroptosis in human and murine neutrophils. Notably, deletion of Exotoxins U or S in P. aeruginosa enhanced neutrophil death to Caspase-1-dependent pyroptosis, suggesting that these exotoxins interfere with this pathway. Mechanistically, P. aeruginosa Flagellin activates the NLRC4 inflammasome, which supports Caspase-1-driven interleukin (IL)-1ß secretion and Gasdermin D (GSDMD)-dependent neutrophil pyroptosis. Furthermore, P. aeruginosa-induced GSDMD activation triggers Calcium-dependent and Peptidyl Arginine Deaminase-4-driven histone citrullination and translocation of neutrophil DNA into the cell cytosol without inducing extracellular Neutrophil Extracellular Traps. Finally, we show that neutrophil Caspase-1 contributes to IL-1ß production and susceptibility to pyroptosis-inducing P. aeruginosa strains in vivo. Overall, we demonstrate that neutrophils are not universally resistant for Caspase-1-dependent pyroptosis.


Assuntos
Inflamassomos , Piroptose , Animais , Proteínas Reguladoras de Apoptose/genética , Caspase 1/metabolismo , Exotoxinas/metabolismo , Humanos , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/microbiologia , Pseudomonas aeruginosa/metabolismo
5.
Vet Res ; 55(1): 81, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38926765

RESUMO

The escalation of antibiotic resistance, pandemics, and nosocomial infections underscores the importance of research in both animal and human infectious diseases. Recent advancements in three-dimensional tissue cultures, or "organoids", have revolutionized the development of in vitro models for infectious diseases. Our study conducts a bibliometric analysis on the use of organoids in modeling infectious diseases, offering an in-depth overview of this field's current landscape. We examined scientific contributions from 2009 onward that focused on organoids in host‒pathogen interactions using the Web of Science Core Collection and OpenAlex database. Our analysis included temporal trends, reference aging, author, and institutional productivity, collaborative networks, citation metrics, keyword cluster dynamics, and disruptiveness of organoid models. VOSviewer, CiteSpace, and Python facilitated this analytical assessment. The findings reveal significant growth and advancements in organoid-based infectious disease research. Analysis of keywords and impactful publications identified three distinct developmental phases in this area that were significantly influenced by outbreaks of Zika and SARS-CoV-2 viruses. The research also highlights the synergistic efforts between academia and publishers in tackling global pandemic challenges. Through mostly consolidating research efforts, organoids are proving to be a promising tool in infectious disease research for both human and animal infectious disease. Their integration into the field necessitates methodological refinements for better physiological emulation and the establishment of extensive organoid biobanks. These improvements are crucial for fully harnessing the potential of organoids in understanding infectious diseases and advancing the development of targeted treatments and vaccines.


Assuntos
Bibliometria , Organoides , Organoides/virologia , Animais , Humanos , Doenças Transmissíveis/veterinária , Doenças Transmissíveis/epidemiologia , Modelos Animais de Doenças , COVID-19/epidemiologia , COVID-19/virologia
6.
Mol Microbiol ; 117(3): 682-692, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34605588

RESUMO

Respiratory infections remain a major global health concern. Tuberculosis is one of the top 10 causes of death worldwide, while infections with Non-Tuberculous Mycobacteria are rising globally. Recent advances in human tissue modeling offer a unique opportunity to grow different human "organs" in vitro, including the human airway, that faithfully recapitulates lung architecture and function. Here, we have explored the potential of human airway organoids (AOs) as a novel system in which to assess the very early steps of mycobacterial infection. We reveal that Mycobacterium tuberculosis (Mtb) and Mycobacterium abscessus (Mabs) mainly reside as extracellular bacteria and infect epithelial cells with very low efficiency. While the AO microenvironment was able to control, but not eliminate Mtb, Mabs thrives. We demonstrate that AOs responded to infection by modulating cytokine, antimicrobial peptide, and mucin gene expression. Given the importance of myeloid cells in mycobacterial infection, we co-cultured infected AOs with human monocyte-derived macrophages and found that these cells interact with the organoid epithelium. We conclude that adult stem cell (ASC)-derived AOs can be used to decipher very early events of mycobacteria infection in human settings thus offering new avenues for fundamental and therapeutic research.


Assuntos
Mycobacterium abscessus , Mycobacterium tuberculosis , Tuberculose , Humanos , Macrófagos/microbiologia , Micobactérias não Tuberculosas , Organoides , Tuberculose/microbiologia
7.
PLoS Pathog ; 17(9): e1009927, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34516571

RESUMO

Regulated cell necrosis supports immune and anti-infectious strategies of the body; however, dysregulation of these processes drives pathological organ damage. Pseudomonas aeruginosa expresses a phospholipase, ExoU that triggers pathological host cell necrosis through a poorly characterized pathway. Here, we investigated the molecular and cellular mechanisms of ExoU-mediated necrosis. We show that cellular peroxidised phospholipids enhance ExoU phospholipase activity, which drives necrosis of immune and non-immune cells. Conversely, both the endogenous lipid peroxidation regulator GPX4 and the pharmacological inhibition of lipid peroxidation delay ExoU-dependent cell necrosis and improve bacterial elimination in vitro and in vivo. Our findings also pertain to the ExoU-related phospholipase from the bacterial pathogen Burkholderia thailandensis, suggesting that exploitation of peroxidised phospholipids might be a conserved virulence mechanism among various microbial phospholipases. Overall, our results identify an original lipid peroxidation-based virulence mechanism as a strong contributor of microbial phospholipase-driven pathology.


Assuntos
Proteínas de Bactérias/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Peroxidação de Lipídeos/fisiologia , Infecções por Pseudomonas/metabolismo , Pseudomonas aeruginosa/patogenicidade , Animais , Humanos , Camundongos , Camundongos Knockout , Necrose/metabolismo , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/metabolismo , Virulência/fisiologia
8.
J Immunol ; 207(7): 1857-1870, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34479945

RESUMO

The lungs harbor multiple resident microbial communities, otherwise known as the microbiota. There is an emerging interest in deciphering whether the pulmonary microbiota modulate local immunity, and whether this knowledge could shed light on mechanisms operating in the response to respiratory pathogens. In this study, we investigate the capacity of a pulmonary Lactobacillus strain to modulate the lung T cell compartment and assess its prophylactic potential upon infection with Mycobacterium tuberculosis, the etiological agent of tuberculosis. In naive mice, we report that a Lactobacillus murinus (Lagilactobacillus murinus) strain (CNCM I-5314) increases the presence of lung Th17 cells and of a regulatory T cell (Treg) subset known as RORγt+ Tregs. In particular, intranasal but not intragastric administration of CNCM I-5314 increases the expansion of these lung leukocytes, suggesting a local rather than systemic effect. Resident Th17 and RORγt+ Tregs display an immunosuppressive phenotype that is accentuated by CNCM I-5314. Despite the well-known ability of M. tuberculosis to modulate lung immunity, the immunomodulatory effect by CNCM I-5314 is dominant, as Th17 and RORγt+ Tregs are still highly increased in the lung at 42-d postinfection. Importantly, CNCM I-5314 administration in M. tuberculosis-infected mice results in reduction of pulmonary inflammation, without increasing M. tuberculosis burden. Collectively, our findings provide evidence for an immunomodulatory capacity of CNCM I-5314 at steady state and in a model of chronic inflammation in which it can display a protective role, suggesting that L. murinus strains found in the lung may shape local T cells in mice and, perhaps, in humans.


Assuntos
Lactobacillus/fisiologia , Pulmão/imunologia , Mycobacterium tuberculosis/fisiologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Tuberculose Pulmonar/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Pulmão/microbiologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia
9.
PLoS Pathog ; 16(10): e1008929, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33002063

RESUMO

The ability of Mycobacterium tuberculosis (Mtb) to persist inside host cells relies on metabolic adaptation, like the accumulation of lipid bodies (LBs) in the so-called foamy macrophages (FM), which are favorable to Mtb. The activation state of macrophages is tightly associated to different metabolic pathways, such as lipid metabolism, but whether differentiation towards FM differs between the macrophage activation profiles remains unclear. Here, we aimed to elucidate whether distinct macrophage activation states exposed to a tuberculosis-associated microenvironment or directly infected with Mtb can form FM. We showed that the triggering of signal transducer and activator of transcription 6 (STAT6) in interleukin (IL)-4-activated human macrophages (M(IL-4)) prevents FM formation induced by pleural effusion from patients with tuberculosis. In these cells, LBs are disrupted by lipolysis, and the released fatty acids enter the ß-oxidation (FAO) pathway fueling the generation of ATP in mitochondria. Accordingly, murine alveolar macrophages, which exhibit a predominant FAO metabolism, are less prone to become FM than bone marrow derived-macrophages. Interestingly, direct infection of M(IL-4) macrophages with Mtb results in the establishment of aerobic glycolytic pathway and FM formation, which could be prevented by FAO activation or inhibition of the hypoxia-inducible factor 1-alpha (HIF-1α)-induced glycolytic pathway. In conclusion, our results demonstrate that Mtb has a remarkable capacity to induce FM formation through the rewiring of metabolic pathways in human macrophages, including the STAT6-driven alternatively activated program. This study provides key insights into macrophage metabolism and pathogen subversion strategies.


Assuntos
Células Espumosas/microbiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Metabolismo dos Lipídeos , Macrófagos/microbiologia , Mycobacterium tuberculosis/patogenicidade , Animais , Gotículas Lipídicas/metabolismo , Ativação de Macrófagos/fisiologia , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Mycobacterium tuberculosis/fisiologia , Tuberculose/microbiologia
10.
Int J Mol Sci ; 23(21)2022 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-36362409

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic continues to cause significant morbidity and mortality worldwide. Since a large portion of the world's population is currently unvaccinated or incompletely vaccinated and has limited access to approved treatments against COVID-19, there is an urgent need to continue research on treatment options, especially those at low cost and which are immediately available to patients, particularly in low- and middle-income countries. Prior in vitro and observational studies have shown that fluoxetine, possibly through its inhibitory effect on the acid sphingomyelinase/ceramide system, could be a promising antiviral and anti-inflammatory treatment against COVID-19. In this report, we evaluated the potential antiviral and anti-inflammatory activities of fluoxetine in a K18-hACE2 mouse model of SARS-CoV-2 infection, and against variants of concern in vitro, i.e., SARS-CoV-2 ancestral strain, Alpha B.1.1.7, Gamma P1, Delta B1.617 and Omicron BA.5. Fluoxetine, administrated after SARS-CoV-2 infection, significantly reduced lung tissue viral titres and expression of several inflammatory markers (i.e., IL-6, TNFα, CCL2 and CXCL10). It also inhibited the replication of all variants of concern in vitro. A modulation of the ceramide system in the lung tissues, as reflected by the increase in the ratio HexCer 16:0/Cer 16:0 in fluoxetine-treated mice, may contribute to explain these effects. Our findings demonstrate the antiviral and anti-inflammatory properties of fluoxetine in a K18-hACE2 mouse model of SARS-CoV-2 infection, and its in vitro antiviral activity against variants of concern, establishing fluoxetine as a very promising candidate for the prevention and treatment of SARS-CoV-2 infection and disease pathogenesis.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2 , Animais , Camundongos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Ceramidas , Modelos Animais de Doenças , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico
11.
Proc Natl Acad Sci U S A ; 114(4): E540-E549, 2017 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-28069953

RESUMO

Immune response against pathogens is a tightly regulated process that must ensure microbial control while preserving integrity of the infected organs. Tuberculosis (TB) is a paramount example of a chronic infection in which antimicrobial immunity is protective in the vast majority of infected individuals but can become detrimental if not finely tuned. Here, we report that C-type lectin dendritic cell (DC) immunoreceptor (DCIR), a key component in DC homeostasis, is required to modulate lung inflammation and bacterial burden in TB. DCIR is abundantly expressed in pulmonary lesions in Mycobacterium tuberculosis-infected nonhuman primates during both latent and active disease. In mice, we found that DCIR deficiency impairs STAT1-mediated type I IFN signaling in DCs, leading to increased production of IL-12 and increased differentiation of T lymphocytes toward Th1 during infection. As a consequence, DCIR-deficient mice control M. tuberculosis better than WT animals but also develop more inflammation characterized by an increased production of TNF and inducible NOS (iNOS) in the lungs. Altogether, our results reveal a pathway by which a C-type lectin modulates the equilibrium between infection-driven inflammation and pathogen's control through sustaining type I IFN signaling in DCs.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/imunologia , Lectinas Tipo C/imunologia , Tuberculose/imunologia , Animais , Feminino , Lectinas Tipo C/genética , Macaca mulatta , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Fator de Transcrição STAT1/imunologia , Transdução de Sinais
12.
Mem Inst Oswaldo Cruz ; 114: e190102, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31411311

RESUMO

BACKGROUND: Once in the pulmonary alveoli, Mycobacterium tuberculosis (Mtb) enters into contact with alveolar macrophages and dendritic cells (DCs). DCs represent the link between the innate and adaptive immune system owing to their capacity to be both a sentinel and an orchestrator of the antigen-specific immune responses against Mtb. The effect that the virulence of Mtb has on the interaction between the bacilli and human DCs has not been fully explored. OBJECTIVE: To evaluate the effect of Mtb virulence on human monocyte-derived DCs. METHODS: We exposed human monocyte-derived DCs to Mtb clinical strains (isolated from an epidemiological Mtb diversity study in Mexico) bearing different degrees of virulence and evaluated the capacity of DCs to internalise the bacilli, control intracellular growth, engage cell death pathways, express markers for activation and antigen presentation, and expand to stimulate autologous CD4+ T cells proliferation. FINDINGS: In the case of the hypervirulent Mtb strain (Phenotype 1, strain 9005186, lineage 3), we report that DCs internalise and neutralise intracellular growth of the bacilli, undergo low rates of apoptosis, and contribute poorly to T-cell expansion, as compared to the H37Rv reference strain. In the case of the hypovirulent Mtb strain (Phenotype 4, strain 9985449, lineage 4), although DCs internalise and preclude proliferation of the bacilli, the DCs also display a high level of apoptosis, massive levels of apoptosis that prevent them from maintaining autologous CD4+ T cells in a co-culture system, as compared to H37Rv. MAIN CONCLUSIONS: Our findings suggest that variability in virulence among Mtb clinical strains affects the capacity of DCs to respond to pathogenic challenge and mount an immune response against it, highlighting important parallels to studies previously done in mouse models.


Assuntos
Células Dendríticas/virologia , Ativação Linfocitária , Mycobacterium tuberculosis/patogenicidade , Linfócitos T Reguladores/parasitologia , Animais , Humanos , Camundongos , Transdução de Sinais , Virulência
14.
Mem Inst Oswaldo Cruz ; 113(4): e170326, 2018 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-29513874

RESUMO

BACKGROUND: Type 2 diabetes (T2D) is a risk factor for the development of tuberculosis (TB), although the associated mechanisms are not known. OBJECTIVES: To study the association between T2D and the basal phenotype of macrophages, and their immune response to Mycobacterium tuberculosis (Mtb) infection. METHODS: We evaluated the influence of T2D on the response of monocyte-derived macrophages (MDM) to Mtb in patients with T2D (n = 10) compared to healthy subjects (n = 9), before and after infection with Mtb clinical isolates bearing different degrees of virulence. The levels of cell surface markers for activation secreted cytokines and chemokines, bacterial association, and intracellular bacterial growth were evaluated. FINDINGS: The expression levels of HLA-DR, CD80, and CD86 were low while those of of PD-L1 were high in uninfected MDMs derived from patients with diabetes; as a result of Mtb infection, changes were only observed in the expression levels of PD-L1. The levels of cytokines (e.g., IL-6, IL-1ß, IL-10, and IL-12) and chemokines (e.g., MCP-1, MIG, and RANTES) are perturbed in MDMs derived from patients with diabetes, both before infection and in response to Mtb infection. In response to the more virulent Mtb strains, the levels of association and bacterial clearance were diminished in MDMs derived from patients with diabetes. CONCLUSIONS: T2D affects the basal activation state of the macrophages and its capacity to respond and control Mtb infection.


Assuntos
Diabetes Mellitus Tipo 2/imunologia , Macrófagos/imunologia , Mycobacterium tuberculosis/crescimento & desenvolvimento , Mycobacterium tuberculosis/patogenicidade , Fenótipo , Tuberculose/imunologia , Adulto , Idoso , Análise de Variância , Glicemia/análise , Estudos de Casos e Controles , Quimiocinas/análise , Contagem de Colônia Microbiana , Citocinas/análise , Diabetes Mellitus Tipo 2/complicações , Feminino , Humanos , Macrófagos/metabolismo , Masculino , Pessoa de Meia-Idade , Valores de Referência , Fatores de Risco , Estatísticas não Paramétricas , Virulência
16.
Circulation ; 132(6): 490-501, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26068045

RESUMO

BACKGROUND: Leukocyte migration is critical for the infiltration of monocytes and accumulation of monocyte-derived macrophages in inflammation. Considering that Hck and Fgr are instrumental in this process, their impact on atherosclerosis and on lesion inflammation and stability was evaluated. METHODS AND RESULTS: Hematopoietic Hck/Fgr-deficient, LDLr(-/-) chimeras, obtained by bone marrow transplantation, had smaller but, paradoxically, less stable lesions with reduced macrophage content, overt cap thinning, and necrotic core expansion as the most prominent features. Despite a Ly6C(high)-skewed proinflammatory monocyte phenotype, Hck/Fgr deficiency led to disrupted adhesion of myeloid cells to and transmigration across endothelial monolayers in vitro and atherosclerotic plaques in vivo, as assessed by intravital microscopy, flow cytometry, and histological examination of atherosclerotic arteries. Moreover, Hck/Fgr-deficient macrophages showed blunted podosome formation and mesenchymal migration capacity. In consequence, transmigrated double-knockout macrophages were seen to accumulate in the fibrous cap, potentially promoting its focal erosion, as observed for double-knockout chimeras. CONCLUSIONS: The hematopoietic deficiency of Hck and Fgr led to attenuated atherosclerotic plaque formation by abrogating endothelial adhesion and transmigration; paradoxically, it also promoted plaque instability by causing monocyte subset imbalance and subendothelial accumulation, raising a note of caution regarding src kinase-targeted intervention in plaque inflammation.


Assuntos
Quimiotaxia de Leucócito/fisiologia , Macrófagos Peritoneais/patologia , Monócitos/patologia , Placa Aterosclerótica/patologia , Proteínas Proto-Oncogênicas c-hck/deficiência , Proteínas Proto-Oncogênicas/deficiência , Quinases da Família src/deficiência , Animais , Apoptose , Adesão Celular , Extensões da Superfície Celular/ultraestrutura , Células Cultivadas , Células Endoteliais , Proteínas da Matriz Extracelular/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Migração e Rolagem de Leucócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Placa Aterosclerótica/enzimologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-hck/genética , Proteínas Proto-Oncogênicas c-hck/fisiologia , Quimera por Radiação , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores de LDL/fisiologia , Migração Transendotelial e Transepitelial , Quinases da Família src/genética , Quinases da Família src/fisiologia
17.
FASEB J ; 29(5): 1914-29, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25609430

RESUMO

Models of microbe-elicited peritonitis have been invaluable to identify mechanisms underlying inflammation resolution, but whether resolution mechanisms differ from an inflammatory agent to another has not been determined. Thus, we analyzed the cellular and molecular components of the resolution phase of non-microbe-induced inflammation. In thioglycollate (TG)-induced peritonitis, resolution started at 12 h (Tmax) and displayed a 22 h resolution interval (Ri). During resolution, lipoxin A4, resolvin (Rv) D1 and RvD2, protectin D1 (PD1), and maresin 1 (MaR1) were transiently produced while RvD5 was continually generated. In addition, docosahexaenoic acid (DHA)-derived mediators were produced to a higher extent than in microbial peritonitis. We also investigated leukocyte infiltration and clearance in peritoneal tissues surrounding the inflammatory site. In the omentum, resolution parameters, neutrophil apoptosis, and efferocytosis were similar to those of the peritoneal cavity. However, we noticed long-term persistence of M2-polarized macrophages and B-lymphocytes in the omentum after TG administration, whereas zymosan injection caused M1/M2-macrophage and T-lymphocyte persistence regardless of the magnitude of the inflammatory response. Our study indicates that some aspects of resolution are shaped in a stimulus-specific manner, and it ultimately argues that the tissues surrounding the inflammatory site must also be considered to address the inflammatory response globally.


Assuntos
Linfócitos B/imunologia , Inflamação/imunologia , Leucócitos/imunologia , Macrófagos/imunologia , Peritonite/imunologia , Peritonite/metabolismo , Tioglicolatos/toxicidade , Animais , Apoptose/imunologia , Linfócitos B/metabolismo , Linfócitos B/patologia , Western Blotting , Células Cultivadas , Ácidos Docosa-Hexaenoicos/genética , Ácidos Docosa-Hexaenoicos/metabolismo , Feminino , Citometria de Fluxo , Técnicas Imunoenzimáticas , Inflamação/metabolismo , Inflamação/patologia , Leucócitos/metabolismo , Leucócitos/patologia , Lipídeos/análise , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Omento/imunologia , Omento/metabolismo , Omento/patologia , Peritonite/induzido quimicamente , Fagocitose/fisiologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Zimosan/toxicidade
18.
J Biol Chem ; 289(11): 7897-906, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24482227

RESUMO

We have shown previously that tyrosine phosphorylation of Wiskott-Aldrich syndrome protein (WASP) is important for diverse macrophage functions including phagocytosis, chemotaxis, podosome dynamics, and matrix degradation. However, the specific tyrosine kinase mediating WASP phosphorylation is still unclear. Here, we provide evidence that Hck, which is predominantly expressed in leukocytes, can tyrosine phosphorylate WASP and regulates WASP-mediated macrophage functions. We demonstrate that tyrosine phosphorylation of WASP in response to stimulation with CX3CL1 or via Fcγ receptor ligation were severely reduced in Hck(-/-) bone marrow-derived macrophages (BMMs) or in RAW/LR5 macrophages in which Hck expression was silenced using RNA-mediated interference (Hck shRNA). Consistent with reduced WASP tyrosine phosphorylation, phagocytosis, chemotaxis, and matrix degradation are reduced in Hck(-/-) BMMs or Hck shRNA cells. In particular, WASP phosphorylation was primarily mediated by the p61 isoform of Hck. Our studies also show that Hck and WASP are required for passage through a dense three-dimensional matrix and transendothelial migration, suggesting that tyrosine phosphorylation of WASP by Hck may play a role in tissue infiltration of macrophages. Consistent with a role for this pathway in invasion, WASP(-/-) BMMs do not invade into tumor spheroids with the same efficiency as WT BMMs and cells expressing phospho-deficient WASP have reduced ability to promote carcinoma cell invasion. Altogether, our results indicate that tyrosine phosphorylation of WASP by Hck is required for proper macrophage functions.


Assuntos
Macrófagos/citologia , Proteínas Proto-Oncogênicas c-hck/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/química , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Animais , Células da Medula Óssea/citologia , Linhagem Celular , Movimento Celular , Quimiotaxia , Quimiotaxia de Leucócito , Colágeno/química , Cruzamentos Genéticos , Células Endoteliais/citologia , Macrófagos/metabolismo , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação , Isoformas de Proteínas/química , Interferência de RNA , Migração Transendotelial e Transepitelial , Tirosina/química
19.
Immunol Cell Biol ; 92(8): 699-708, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24890643

RESUMO

Mononuclear phagocytes (MP) comprise monocytes, macrophages (MΦ) and dendritic cells (DC), including their lineage-committed progenitors, which together have an eminent role in health and disease. Lipid-based siRNA-mediated gene inactivation is an established approach to investigate gene function in MP cells. However, although there are few protocols dedicated for siRNA-mediated gene inactivation in primary human DC and MΦ, there are none available for primary human monocytes. Moreover, there is no available method to perform comparative studies of a siRNA-mediated gene silencing in primary monocytes and other MP cells. Here, we describe a protocol optimized for the lipid-based delivery of siRNA to perform gene silencing in primary human blood monocytes, which is applicable to DCs, and differs from the classical route of siRNA delivery into MΦs. Along with this protocol, we provide a comparative analysis of how monocytes, DC and MΦ are efficiently transfected with the target siRNA without affecting cell viability, resulting in strong gene knockdown efficiency, including the simultaneous inactivation of two genes. Moreover, siRNA delivery does not affect classical functions in MP such as differentiation, phagocytosis and migration, demonstrating that this protocol does not induce non-specific major alterations in these cells. As a proof-of-principle, a functional analysis of hematopoietic cell kinase (Hck) shows for the first time that this kinase regulates the protease-dependent migration mode in human monocytes. Collectively, this protocol enables efficient gene inactivation in primary MP, suggesting a wide spectrum of applications such as siRNA-based high-throughput screening, which could ultimately improve our knowledge about MP biology.


Assuntos
Células Dendríticas/metabolismo , Inativação Gênica , Macrófagos/metabolismo , Monócitos/metabolismo , RNA Interferente Pequeno/genética , Animais , Diferenciação Celular/genética , Movimento Celular/genética , Movimento Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Macrófagos/citologia , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Monócitos/citologia , Monócitos/imunologia , Fagocitose/imunologia , Proteínas Proto-Oncogênicas c-hck/genética , Proteínas Proto-Oncogênicas c-hck/metabolismo , Interferência de RNA , RNA Mensageiro/genética , Transfecção
20.
FASEB J ; 27(9): 3608-18, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23742809

RESUMO

In osteoclasts, Src controls podosome organization and bone degradation, which leads to an osteopetrotic phenotype in src(-/-) mice. Since this phenotype was even more severe in src(-/-)hck(-/-) mice, we examined the individual contribution of Hck in bone homeostasis. Compared to wt mice, hck(-/-) mice exhibited an osteopetrotic phenotype characterized by an increased density of trabecular bone and decreased bone degradation, although osteoclastogenesis was not impaired. Podosome organization and matrix degradation were found to be defective in hck(-/-) osteoclast precursors (preosteoclast) but were normal in mature hck(-/-) osteoclasts, probably through compensation by Src, which was specifically overexpressed in mature osteoclasts. As a consequence of podosome defects, the 3-dimensional migration of hck(-/-) preosteoclasts was strongly affected in vitro. In vivo, this translated by altered bone homing of preosteoclasts in hck(-/-) mice: in metatarsals of 1-wk-old mice, when bone formation strongly depends on the recruitment of these cells, reduced numbers of osteoclasts and abnormal developing trabecular bone were observed. This phenotype was still detectable in adults. In summmary, Hck is one of the very few effectors of preosteoclast recruitment described to date and thereby plays a critical role in bone remodeling.


Assuntos
Osso e Ossos/citologia , Osso e Ossos/metabolismo , Movimento Celular/fisiologia , Osteoclastos/citologia , Osteopetrose/metabolismo , Proteínas Proto-Oncogênicas c-hck/metabolismo , Animais , Movimento Celular/genética , Células Cultivadas , Feminino , Homeostase/genética , Homeostase/fisiologia , Masculino , Camundongos , Camundongos Knockout , Osteoclastos/metabolismo , Osteopetrose/genética , Proteínas Proto-Oncogênicas c-hck/genética , Quinases da Família src/genética , Quinases da Família src/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA