Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell ; 183(1): 94-109.e23, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32937105

RESUMO

Cardiomyocytes are subjected to the intense mechanical stress and metabolic demands of the beating heart. It is unclear whether these cells, which are long-lived and rarely renew, manage to preserve homeostasis on their own. While analyzing macrophages lodged within the healthy myocardium, we discovered that they actively took up material, including mitochondria, derived from cardiomyocytes. Cardiomyocytes ejected dysfunctional mitochondria and other cargo in dedicated membranous particles reminiscent of neural exophers, through a process driven by the cardiomyocyte's autophagy machinery that was enhanced during cardiac stress. Depletion of cardiac macrophages or deficiency in the phagocytic receptor Mertk resulted in defective elimination of mitochondria from the myocardial tissue, activation of the inflammasome, impaired autophagy, accumulation of anomalous mitochondria in cardiomyocytes, metabolic alterations, and ventricular dysfunction. Thus, we identify an immune-parenchymal pair in the murine heart that enables transfer of unfit material to preserve metabolic stability and organ function. VIDEO ABSTRACT.


Assuntos
Macrófagos/metabolismo , Mitocôndrias/metabolismo , Miócitos Cardíacos/metabolismo , Idoso , Animais , Apoptose , Autofagia , Feminino , Coração/fisiologia , Homeostase , Humanos , Macrófagos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Mitocôndrias/fisiologia , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/fisiologia , Fagocitose/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , c-Mer Tirosina Quinase/metabolismo
2.
Cell ; 153(5): 1025-35, 2013 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-23706740

RESUMO

Unique among leukocytes, neutrophils follow daily cycles of release from and migration back into the bone marrow, where they are eliminated. Because removal of dying cells generates homeostatic signals, we explored whether neutrophil elimination triggers circadian events in the steady state. Here, we report that the homeostatic clearance of neutrophils provides cues that modulate the physiology of the bone marrow. We identify a population of CD62L(LO) CXCR4(HI) neutrophils that have "aged" in the circulation and are eliminated at the end of the resting period in mice. Aged neutrophils infiltrate the bone marrow and promote reductions in the size and function of the hematopoietic niche. Modulation of the niche depends on macrophages and activation of cholesterol-sensing nuclear receptors and is essential for the rhythmic egress of hematopoietic progenitors into the circulation. Our results unveil a process that synchronizes immune and hematopoietic rhythms and expand the ascribed functions of neutrophils beyond inflammation. PAPERFLICK:


Assuntos
Medula Óssea/fisiologia , Ritmo Circadiano , Neutrófilos/citologia , Neutrófilos/fisiologia , Animais , Movimento Celular , Senescência Celular , Feminino , Células-Tronco Hematopoéticas/metabolismo , Homeostase , Receptores X do Fígado , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/imunologia , Receptores Nucleares Órfãos/metabolismo
3.
Nat Immunol ; 14(8): 831-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23770640

RESUMO

Macrophages are professional phagocytic cells that orchestrate innate immune responses and have considerable phenotypic diversity at different anatomical locations. However, the mechanisms that control the heterogeneity of tissue macrophages are not well characterized. Here we found that the nuclear receptor LXRα was essential for the differentiation of macrophages in the marginal zone (MZ) of the spleen. LXR-deficient mice were defective in the generation of MZ and metallophilic macrophages, which resulted in abnormal responses to blood-borne antigens. Myeloid-specific expression of LXRα or adoptive transfer of wild-type monocytes restored the MZ microenvironment in LXRα-deficient mice. Our results demonstrate that signaling via LXRα in myeloid cells is crucial for the generation of splenic MZ macrophages and identify an unprecedented role for a nuclear receptor in the generation of specialized macrophage subsets.


Assuntos
Hematopoese/imunologia , Macrófagos/imunologia , Receptores Nucleares Órfãos/imunologia , Baço/imunologia , Animais , Benzoatos/farmacologia , Benzilaminas/farmacologia , Diferenciação Celular/imunologia , Citometria de Fluxo , Imunidade Celular/imunologia , Imuno-Histoquímica , Receptores X do Fígado , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Receptores Nucleares Órfãos/agonistas , Transdução de Sinais/imunologia , Organismos Livres de Patógenos Específicos , Baço/citologia
4.
EMBO J ; 38(19): e101233, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31414712

RESUMO

Tissues in multicellular organisms are populated by resident macrophages, which perform both generic and tissue-specific functions. The latter are induced by signals from the microenvironment and rely on unique tissue-specific molecular programs requiring the combinatorial action of tissue-specific and broadly expressed transcriptional regulators. Here, we identify the transcription factors Bhlhe40 and Bhlhe41 as novel regulators of alveolar macrophages (AMs)-a population that provides the first line of immune defense and executes homeostatic functions in lung alveoli. In the absence of these factors, AMs exhibited decreased proliferation that resulted in a severe disadvantage of knockout AMs in a competitive setting. Gene expression analyses revealed a broad cell-intrinsic footprint of Bhlhe40/Bhlhe41 deficiency manifested by a downregulation of AM signature genes and induction of signature genes of other macrophage lineages. Genome-wide characterization of Bhlhe40 DNA binding suggested that these transcription factors directly repress the expression of lineage-inappropriate genes in AMs. Taken together, these results identify Bhlhe40 and Bhlhe41 as key regulators of AM self-renewal and guardians of their identity.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Perfilação da Expressão Gênica/métodos , Proteínas de Homeodomínio/genética , Macrófagos Alveolares/citologia , Acetilação , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Proliferação de Células , Autorrenovação Celular , Sobrevivência Celular , Regulação para Baixo , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Macrófagos Alveolares/metabolismo , Camundongos , Especificidade de Órgãos , Fenótipo , Análise de Sequência de RNA
5.
Arterioscler Thromb Vasc Biol ; 42(2): 145-155, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34911360

RESUMO

BACKGROUND: Monocytes are thought to be involved in venous thrombosis but the role of individual monocyte subpopulations on thrombus formation, clot inflammation, and degradation is an important unresolved issue. We investigate the role of inflammatory Ly6Chi monocytes in deep vein thrombosis and their potential therapeutic impact. METHODS: Frequencies and compositions of blood monocytes were analyzed by flow cytometry in CCR2-/- (C-C chemokine receptor type 2) and wild-type mice of different ages and after treatment with the NR4A1 (nuclear receptor group 4 family A member 1, Nur77) agonist CnsB (cytosporone B). TF (tissue factor) sufficient and deficient Ly6Chi monocytes were adoptively transferred into aged CCR2-/- mice. Thrombus formation and size were followed by ultrasound over a 3-week period after surgical reduction of blood flow (stenosis) in the inferior vena cava. RESULTS: Reduced numbers of peripheral monocytes in aged (>30 w) CCR2-/- mice are accompanied by reduced thrombus formation after inferior vena cava ligation. Reducing the number of inflammatory Ly6Chi monocytes in wild-type mice by CsnB treatment before ligation, similarly suspends clotting, while later treatment (d1 or d4) reduces thrombus growth and accelerates resolution. We describe how changes in inflammatory monocyte numbers affect the gradual differentiation of monocytes in thrombi and show that only tissue factor-competent Ly6Chi monocytes restore thrombosis in aged CCR2-/- mice. CONCLUSIONS: We conclude that the number of inflammatory Ly6Chi monocytes controls deep vein thrombosis formation, growth, and resolution and can be therapeutically manipulated with a NR4A1 agonist at all disease stages.


Assuntos
Inflamação/patologia , Monócitos/patologia , Trombose/patologia , Animais , Células Cultivadas , Humanos , Inflamação/sangue , Contagem de Leucócitos , Camundongos Endogâmicos C57BL , Monócitos/citologia , Trombose/sangue , Veia Cava Inferior/patologia
6.
Pharmacol Res ; 163: 105292, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33171305

RESUMO

Resolution failure of exacerbated inflammation triggered by Influenza A virus (IAV) prevents return of pulmonary homeostasis and survival, especially when associated with secondary pneumococcal infection. Therapeutic strategies based on pro-resolving molecules have great potential against acute inflammatory diseases. Angiotensin-(1-7) [Ang-(1-7)] is a pro-resolving mediator that acts on its Mas receptor (MasR) to promote resolution of inflammation. We investigated the effects of Ang-(1-7) and the role of MasR in the context of primary IAV infection and secondary pneumococcal infection and evaluated pulmonary inflammation, virus titers and bacteria counts, and pulmonary damage. Therapeutic treatment with Ang-(1-7) decreased neutrophil recruitment, lung injury, viral load and morbidity after a primary IAV infection. Ang-(1-7) induced apoptosis of neutrophils and efferocytosis of these cells by alveolar macrophages, but had no direct effect on IAV replication in vitro. MasR-deficient (MasR-/-) mice were highly susceptible to IAV infection, displaying uncontrolled inflammation, increased viral load and greater lethality rate, as compared to WT animals. Ang-(1-7) was not protective in MasR-/- mice. Interestingly, Ang-(1-7) given during a sublethal dose of IAV infection greatly reduced morbidity associated with a subsequent S. pneumoniae infection, as seen by decrease in the magnitude of neutrophil influx, number of bacteria in the blood leading to a lower lethality. Altogether, these results show that Ang-(1-7) is highly protective against severe primary IAV infection and protects against secondary bacterial infection of the lung. These effects are MasR-dependent. Mediators of resolution of inflammation, such as Ang-(1-7), should be considered for the treatment of pulmonary viral infections.


Assuntos
Angiotensina I/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Fragmentos de Peptídeos/uso terapêutico , Infecções Pneumocócicas/tratamento farmacológico , Pneumonia Viral/tratamento farmacológico , Proteínas Proto-Oncogênicas/imunologia , Receptores Acoplados a Proteínas G/imunologia , Células A549 , Angiotensina I/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Líquido da Lavagem Broncoalveolar/citologia , Líquido da Lavagem Broncoalveolar/imunologia , Citocinas/imunologia , Cães , Humanos , Vírus da Influenza A , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Células Madin Darby de Rim Canino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Fragmentos de Peptídeos/farmacologia , Peroxidase/imunologia , Fagocitose/efeitos dos fármacos , Infecções Pneumocócicas/imunologia , Infecções Pneumocócicas/patologia , Pneumonia Viral/imunologia , Pneumonia Viral/patologia , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/genética , Receptores Acoplados a Proteínas G/genética , Streptococcus pneumoniae
7.
Cell Mol Life Sci ; 77(19): 3823-3830, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32248248

RESUMO

The infiltration of naïve tissues by myeloid cells has been long related to their clearance and the physiological cell turnover, however, increasing evidence shows that they can additionally fulfill specific, non-immune functions in different tissues. There is also growing evidence to support that infiltrated granulocytes and monocytes respond to different environments by modulating gene expression and cytokine production, which in turn contribute to the normal function of the host tissue. This review will address the roles of immigrated myeloid cells in different tissues and their crosstalk with the host tissue environments.


Assuntos
Granulócitos/imunologia , Monócitos/imunologia , Comunicação Celular , Citocinas/metabolismo , Expressão Gênica , Granulócitos/metabolismo , Humanos , Imunomodulação , Pulmão/imunologia , Pulmão/metabolismo , Monócitos/metabolismo , Infiltração de Neutrófilos , Neutrófilos/imunologia , Neutrófilos/metabolismo
8.
Cell Immunol ; 330: 151-158, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29779612

RESUMO

Macrophage heterogeneity in the spleen has been long documented, with four subsets populating the different splenic compartments. The diverse environments on the splenic compartments determine their varied phenotype and functions. In the white pulp, highly phagocytic macrophages contribute to the generation of the immune response. The marginal zone contains two populations of macrophages, which also contribute to the immune response. Their strategic position in the bloodstream and their unique phenotype confer them a crucial role in the defense against blood borne pathogens, placing them at the crossroad between innate and adaptive immune responses. Macrophages in the red pulp are classically linked to homeostatic and metabolic functions in erythrocyte phagocytosis and iron recycling. We review here recent advances demonstrating the importance of macrophage ontogeny and organ development in determining the phenotype, transcriptional profile and, ultimately, the functions of the populations of splenic macrophages.


Assuntos
Metabolismo Energético/imunologia , Homeostase/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Baço/imunologia , Imunidade Adaptativa/imunologia , Animais , Eritrócitos/imunologia , Humanos , Imunidade Inata/imunologia , Baço/citologia , Baço/metabolismo
9.
Immunity ; 31(2): 245-58, 2009 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-19646905

RESUMO

Effective clearance of apoptotic cells by macrophages is essential for immune homeostasis. The transcriptional pathways that allow macrophages to sense and respond to apoptotic cells are poorly defined. We found that liver X receptor (LXR) signaling was important for both apoptotic cell clearance and the maintenance of immune tolerance. Apoptotic cell engulfment activated LXR and thereby induced the expression of Mer, a receptor tyrosine kinase critical for phagocytosis. LXR-deficient macrophages exhibited a selective defect in phagocytosis of apoptotic cells and an aberrant proinflammatory response to them. As a consequence of these defects, mice lacking LXRs manifested a breakdown in self-tolerance and developed autoantibodies and autoimmune glomerulonephritis. Treatment with an LXR agonist ameliorated disease progression in a mouse model of lupus-like autoimmunity. Thus, activation of LXR by apoptotic cells engages a virtuous cycle that promotes their own clearance and couples engulfment to the suppression of inflammatory pathways.


Assuntos
Apoptose/imunologia , Doenças Autoimunes/imunologia , Proteínas de Ligação a DNA/agonistas , Macrófagos/imunologia , Receptores Citoplasmáticos e Nucleares/agonistas , Baço/imunologia , Animais , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Autoimunidade/imunologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Tolerância Imunológica/imunologia , Receptores X do Fígado , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nucleares Órfãos , Fagocitose/imunologia , Proteínas Proto-Oncogênicas/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/imunologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/imunologia , Transdução de Sinais/imunologia , Baço/citologia , Baço/metabolismo , c-Mer Tirosina Quinase
10.
Science ; 384(6691): eabo7027, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38574142

RESUMO

Macrophages are functionally heterogeneous cells essential for apoptotic cell clearance. Apoptotic cells are defined by homogeneous characteristics, ignoring their original cell lineage identity. We found that in an interleukin-4 (IL-4)-enriched environment, the sensing of apoptotic neutrophils by macrophages triggered their tissue remodeling signature. Engulfment of apoptotic hepatocytes promoted a tolerogenic phenotype, whereas phagocytosis of T cells had little effect on IL-4-induced gene expression. In a mouse model of parasite-induced pathology, the transfer of macrophages conditioned with IL-4 and apoptotic neutrophils promoted parasitic egg clearance. Knockout of phagocytic receptors required for the uptake of apoptotic neutrophils and partially T cells, but not hepatocytes, exacerbated helminth infection. These findings suggest that the identity of apoptotic cells may contribute to the development of distinct IL-4-driven immune programs in macrophages.


Assuntos
Apoptose , Interleucina-4 , Macrófagos , Fagocitose , Esquistossomose mansoni , Animais , Camundongos , Apoptose/imunologia , Hepatócitos/imunologia , Interleucina-4/genética , Interleucina-4/metabolismo , Macrófagos/imunologia , Camundongos Knockout , Neutrófilos/imunologia , Fagocitose/imunologia , Esquistossomose mansoni/genética , Esquistossomose mansoni/imunologia , Modelos Animais de Doenças
11.
Biochim Biophys Acta ; 1812(8): 982-94, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21193033

RESUMO

The liver X receptors (LXRα and LXRß) are members of the nuclear receptor family of transcription factors that play essential roles in the transcriptional control of lipid metabolism. LXRs are endogenously activated by modified forms of cholesterol known as oxysterols and control the expression of genes important for cholesterol uptake, efflux, transport, and excretion in multiple tissues. In addition to their role as cholesterol sensors, a number of studies have implicated LXRs in the modulation of innate and adaptive immune responses. Both through activation and repression mechanisms, LXRs regulate diverse aspects of inflammatory gene expression in macrophages. The ability of LXRs to coordinate metabolic and immune responses constitutes an attractive therapeutic target for the treatment of chronic inflammatory disorders. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.


Assuntos
Inflamação/fisiopatologia , Macrófagos/fisiologia , Receptores Nucleares Órfãos/fisiologia , Adaptação Fisiológica/imunologia , Animais , Apoptose , Humanos , Imunidade Inata/fisiologia , Inflamação/genética , Mediadores da Inflamação/metabolismo , Receptores X do Fígado , Macrófagos/metabolismo , Fatores de Transcrição/fisiologia
12.
Biomater Adv ; 134: 112551, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35513950

RESUMO

The coating composition of nanomedicines is one of the main features in determining the medicines' fate, clearance, and immunoresponse in the body. To highlight the coatings' impact in pulmonary administration, two micellar superparamagnetic iron oxide nanoparticles (SPION) were compared. These nanoparticles are similar in size and charge but have different coatings: either phosphatidylcholine (PC-SPION) or bovine serum albumin (BSA-SPION). The aim of the study was to increase the understanding of the nano-bio interaction with the cellular and non-cellular components of the lung and underline valuable coatings either for local lung-targeted drug delivery in theranostic application or patient-friendly route systemic administration. PC-SPION and BSA-SPION were deposited in the alveoli by in vivo instillation and, despite the complexity of imaging the lung, SPION were macroscopically visualized by MRI. Impressively, PC-SPION were retained within the lungs for at least a week, while BSA-SPION were cleared more rapidly. The different lung residence times were confirmed by histological analysis and supported by a flow cytometry analysis of the SPION interactions with different myeloid cell populations. To further comprehend the way in which these nanoformulations interact with lung components at the molecular level, we used fluorescence spectroscopy, turbidity measurements, and dynamic light scattering to evaluate the interactions of the two SPION with surfactant protein A (SP-A), a key protein in setting up the nanoparticle behavior in the alveolar fluid. We found that SP-A induced aggregation of PC-SPION, but not BSA-SPION, which likely caused PC-SPION retention in the lung without inducing inflammation. In conclusion, the two SPION show different outcomes from interaction with SP-A leading to distinctive fate in the lung. PC-SPION hold great promise as imaging and theranostic agents when prolonged pulmonary drug delivery is required.


Assuntos
Nanopartículas , Proteína A Associada a Surfactante Pulmonar , Humanos , Pulmão/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Nanopartículas/química , Soroalbumina Bovina
13.
Nat Commun ; 13(1): 945, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35177618

RESUMO

Inflammation triggers secondary brain damage after stroke. The meninges and other CNS border compartments serve as invasion sites for leukocyte influx into the brain thus promoting tissue damage after stroke. However, the post-ischemic immune response of border compartments compared to brain parenchyma remains poorly characterized. Here, we deeply characterize tissue-resident leukocytes in meninges and brain parenchyma and discover that leukocytes respond differently to stroke depending on their site of residence. We thereby discover a unique phenotype of myeloid cells exclusive to the brain after stroke. These stroke-associated myeloid cells partially resemble neurodegenerative disease-associated microglia. They are mainly of resident microglial origin, partially conserved in humans and exhibit a lipid-phagocytosing phenotype. Blocking markers specific for these cells partially ameliorates stroke outcome thus providing a potential therapeutic target. The injury-response of myeloid cells in the CNS is thus compartmentalized, adjusted to the type of injury and may represent a therapeutic target.


Assuntos
Infarto da Artéria Cerebral Média/complicações , Células Mieloides/imunologia , Doenças Neuroinflamatórias/imunologia , Idoso , Idoso de 80 Anos ou mais , Animais , Encéfalo/citologia , Encéfalo/imunologia , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Humanos , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Microglia/citologia , Microglia/imunologia , Pessoa de Meia-Idade , Doenças Neuroinflamatórias/patologia , Pia-Máter/citologia , Pia-Máter/imunologia , Pia-Máter/patologia
14.
Curr Opin Pharmacol ; 53: 18-26, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32361182

RESUMO

Macrophages are immune myeloid cells with an extreme ability to modulate their phenotype in response to insults and/or pathogens. The immunomodulatory capacity of macrophages is also patent during development as they adapt their phenotype to the host tissue environment establishing the heterogeneous populations of tissue-resident macrophages. An important mechanism of immunomodulation in macrophages occurs through the regulation of transcriptional activity. Numerous transcription factors are associated with macrophage plasticity, among them, several nuclear receptors. The nuclear receptors Liver X Receptors (LXRα and LXRß) have also revealed as active players during macrophage adaptations in diverse scenarios. This review will address the different mechanisms by which LXRs contribute to immunomodulation in macrophages by connecting lipid metabolism and immunity through transcriptional regulation.


Assuntos
Imunomodulação , Metabolismo dos Lipídeos , Receptores X do Fígado/imunologia , Macrófagos/imunologia , Animais , Diferenciação Celular , Humanos
15.
Elife ; 92020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33287957

RESUMO

Liver metabolism follows diurnal fluctuations through the modulation of molecular clock genes. Disruption of this molecular clock can result in metabolic disease but its potential regulation by immune cells remains unexplored. Here, we demonstrated that in steady state, neutrophils infiltrated the mouse liver following a circadian pattern and regulated hepatocyte clock-genes by neutrophil elastase (NE) secretion. NE signals through c-Jun NH2-terminal kinase (JNK) inhibiting fibroblast growth factor 21 (FGF21) and activating Bmal1 expression in the hepatocyte. Interestingly, mice with neutropenia, defective neutrophil infiltration or lacking elastase were protected against steatosis correlating with lower JNK activation, reduced Bmal1 and increased FGF21 expression, together with decreased lipogenesis in the liver. Lastly, using a cohort of human samples we found a direct correlation between JNK activation, NE levels and Bmal1 expression in the liver. This study demonstrates that neutrophils contribute to the maintenance of daily hepatic homeostasis through the regulation of the NE/JNK/Bmal1 axis.


Every day, the body's biological processes work to an internal clock known as the circadian rhythm. This rhythm is controlled by 'clock genes' that are switched on or off by daily physical and environmental cues, such as changes in light levels. These daily rhythms are very finely tuned, and disturbances can lead to serious health problems, such as diabetes or high blood pressure. The ability of the body to cycle through the circadian rhythm each day is heavily influenced by the clock of one key organ: the liver. This organ plays a critical role in converting food and drink into energy. There is evidence that neutrophils ­ white blood cells that protect the body by being the first response to inflammation ­ can influence how the liver performs its role in obese people, by for example, releasing a protein called elastase. Additionally, the levels of neutrophils circulating in the blood change following a daily pattern. Crespo, González-Terán et al. wondered whether neutrophils enter the liver at specific times of the day to control liver's daily rhythm. Crespo, González-Terán et al. revealed that neutrophils visit the liver in a pattern that peaks when it gets light and dips when it gets dark by counting the number of neutrophils in the livers of mice at different times of the day. During these visits, neutrophils secreted elastase, which activated a protein called JNK in the cells of the mice's liver. This subsequently blocked the activity of another protein, FGF21, which led to the activation of the genes that allow cells to make fat molecules for storage. JNK activation also switched on the clock gene, Bmal1, ultimately causing fat to build up in the mice's liver. Crespo, González-Terán et al. also found that, in samples from human livers, the levels of elastase, the activity of JNK, and whether the Bmal1 gene was switched on were tightly linked. This suggests that neutrophils may be controlling the liver's rhythm in humans the same way they do in mice. Overall, this research shows that neutrophils can control and reset the liver's daily rhythm using a precisely co-ordinated series of molecular changes. These insights into the liver's molecular clock suggest that elastase, JNK and BmaI1 may represent new therapeutic targets for drugs or smart medicines to treat metabolic diseases such as diabetes or high blood pressure.


Assuntos
Proteínas CLOCK/metabolismo , Regulação da Expressão Gênica/fisiologia , Hepatócitos/metabolismo , Neutrófilos/fisiologia , Animais , Proteínas CLOCK/genética , Células Cultivadas , Ritmo Circadiano , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Inflamação/metabolismo , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Camundongos Transgênicos , Neutropenia
16.
Mol Cell Biol ; 38(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29507185

RESUMO

The liver X receptors (LXRs) are ligand-activated nuclear receptors with established roles in the maintenance of lipid homeostasis in multiple tissues. LXRs exert additional biological functions as negative regulators of inflammation, particularly in macrophages. However, the transcriptional responses controlled by LXRs in other myeloid cells, such as dendritic cells (DCs), are still poorly understood. Here we used gain- and loss-of-function models to characterize the impact of LXR deficiency on DC activation programs. Our results identified an LXR-dependent pathway that is important for DC chemotaxis. LXR-deficient mature DCs are defective in stimulus-induced migration in vitro and in vivo Mechanistically, we show that LXRs facilitate DC chemotactic signaling by regulating the expression of CD38, an ectoenzyme important for leukocyte trafficking. Pharmacological or genetic inactivation of CD38 activity abolished the LXR-dependent induction of DC chemotaxis. Using the low-density lipoprotein receptor-deficient (LDLR-/-) LDLR-/- mouse model of atherosclerosis, we also demonstrated that hematopoietic CD38 expression is important for the accumulation of lipid-laden myeloid cells in lesions, suggesting that CD38 is a key factor in leukocyte migration during atherogenesis. Collectively, our results demonstrate that LXRs are required for the efficient emigration of DCs in response to chemotactic signals during inflammation.


Assuntos
Quimiotaxia/fisiologia , Células Dendríticas/fisiologia , Receptores X do Fígado/fisiologia , ADP-Ribosil Ciclase 1/metabolismo , Animais , Células Cultivadas , Células Dendríticas/citologia , Inflamação , Metabolismo dos Lipídeos , Receptores X do Fígado/genética , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Nucleares Órfãos , Receptores Citoplasmáticos e Nucleares , Transdução de Sinais
17.
J Exp Med ; 215(11): 2778-2795, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30282719

RESUMO

Immune protection relies on the capacity of neutrophils to infiltrate challenged tissues. Naive tissues, in contrast, are believed to remain free of these cells and protected from their toxic cargo. Here, we show that neutrophils are endowed with the capacity to infiltrate multiple tissues in the steady-state, a process that follows tissue-specific dynamics. By focusing in two particular tissues, the intestine and the lungs, we find that neutrophils infiltrating the intestine are engulfed by resident macrophages, resulting in repression of Il23 transcription, reduced G-CSF in plasma, and reinforced activity of distant bone marrow niches. In contrast, diurnal accumulation of neutrophils within the pulmonary vasculature influenced circadian transcription in the lungs. Neutrophil-influenced transcripts in this organ were associated with carcinogenesis and migration. Consistently, we found that neutrophils dictated the diurnal patterns of lung invasion by melanoma cells. Homeostatic infiltration of tissues unveils a facet of neutrophil biology that supports organ function, but can also instigate pathological states.


Assuntos
Neoplasias Pulmonares/imunologia , Pulmão/imunologia , Melanoma/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Animais , Feminino , Fator Estimulador de Colônias de Granulócitos/genética , Fator Estimulador de Colônias de Granulócitos/imunologia , Interleucina-23/genética , Interleucina-23/imunologia , Pulmão/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Knockout , Invasividade Neoplásica/genética , Invasividade Neoplásica/imunologia , Neutrófilos/patologia , Transcrição Gênica/imunologia
18.
J Exp Med ; 214(5): 1281-1296, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28432199

RESUMO

Tissue-resident macrophages display varying phenotypic and functional properties that are largely specified by their local environment. One of these functions, phagocytosis, mediates the natural disposal of billions of cells, but its mechanisms and consequences within living tissues are poorly defined. Using a parabiosis-based strategy, we identified and isolated macrophages from multiple tissues as they phagocytosed blood-borne cellular material. Phagocytosis was circadianally regulated and mediated by distinct repertoires of receptors, opsonins, and transcription factors in macrophages from each tissue. Although the tissue of residence defined the core signature of macrophages, phagocytosis imprinted a distinct antiinflammatory profile. Phagocytic macrophages expressed CD206, displayed blunted expression of Il1b, and supported tissue homeostasis. Thus, phagocytosis is a source of macrophage heterogeneity that acts together with tissue-derived factors to preserve homeostasis.


Assuntos
Macrófagos/fisiologia , Fagocitose/fisiologia , Animais , Feminino , Interleucina-1beta/metabolismo , Lectinas Tipo C/metabolismo , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Opsonizantes/fisiologia , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição/fisiologia
19.
Front Immunol ; 5: 211, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24860573

RESUMO

Clearance of apoptotic cells by macrophages occurs as a coordinated process to ensure tissue homeostasis. Macrophages play a dual role in this process; first, a rapid and efficient phagocytosis of the dying cells is needed to eliminate uncleared corpses that can promote inflammation. Second, after engulfment, macrophages exhibit an anti-inflammatory phenotype, to avoid unwanted immune reactions against cell components. Several nuclear receptors, including liver X receptor and proliferator-activated receptor, have been linked to these two important features of macrophages during apoptotic cell clearance. This review outlines the emerging implications of nuclear receptors in the response of macrophages to cell clearance. These include activation of genes implicated in metabolism, to process the additional cellular content provided by the engulfed cells, as well as inflammatory genes, to maintain apoptotic cell clearance as an "immunologically silent" process. Remarkably, genes encoding receptors for the so-called "eat-me" signals are also regulated by activated nuclear receptors after phagocytosis of apoptotic cells, thus enhancing the efficiency of macrophages to clear dead cells.

20.
Int J Hematol ; 99(6): 685-94, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24634109

RESUMO

Two cellular systems of paramount importance for mammalian physiology, the myeloid and the hematopoietic, have received a great deal of attention in the past decade. Myeloid leukocytes, classically involved in mediating innate immune responses, are now known to regulate other important aspects of the organism's physiology, from development to regulation of metabolic functions. In parallel, many diverse cellular and molecular components have been identified in the bone marrow (BM) that are required for the regulation and lifelong preservation of hematopoietic stem and progenitor cells (HSPC). Since the production of blood and immune elements by these multipotent cells responds to environmental signals, it is not entirely surprising that the hematopoietic niches in which HSPC are located can in turn be regulated by the immune system. We review here recent evidence demonstrating that two components of the innate immune system, macrophages and neutrophils, regulate the function of the hematopoietic niche in ways that may favor both the retention and the release of HSPC from the BM. We propose that the highly migratory nature of neutrophils, the presence of a network of tissue-resident macrophages in the BM and possibly in other tissues, and the superb capacity of these innate immune cells to respond to stress endow them with regulatory functions that are ultimately relayed to the hematopoietic niche.


Assuntos
Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Homeostase , Imunidade Inata , Nicho de Células-Tronco , Animais , Comunicação Celular , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa