Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 745
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Annu Rev Immunol ; 33: 643-75, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25861979

RESUMO

Macrophages are myeloid immune cells that are strategically positioned throughout the body tissues, where they ingest and degrade dead cells, debris, and foreign material and orchestrate inflammatory processes. Here we review two major recent paradigm shifts in our understanding of tissue macrophage biology. The first is the realization that most tissue-resident macrophages are established prenatally and maintained through adulthood by longevity and self-renewal. Their generation and maintenance are thus independent from ongoing hematopoiesis, although the cells can be complemented by adult monocyte-derived macrophages. Second, aside from being immune sentinels, tissue macrophages form integral components of their host tissue. This entails their specialization in response to local environmental cues to contribute to the development and specific function of their tissue of residence. Factors that govern tissue macrophage specialization are emerging. Moreover, tissue specialization is reflected in discrete gene expression profiles of macrophages, as well as epigenetic signatures reporting actual and potential enhancer usage.


Assuntos
Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Diferenciação Celular , Humanos , Imunidade , Macrófagos/classificação , Macrófagos/citologia , Especificidade de Órgãos/imunologia , Fenótipo
2.
Immunity ; 52(6): 957-970, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32553181

RESUMO

Resident tissue macrophages (RTMs) have a broad spectrum of immune- and non-immune-related tissue-supporting activities. The roots of this heterogeneity and versatility are only beginning to be understood. Here, we propose a conceptual framework for considering the RTM heterogeneity that organizes the factors shaping RTM identity within four cardinal points: (1) ontogeny and the view that adult RTM populations comprise a defined mixture of cells that arise from either embryonic precursors or adult monocytes; (2) local factors unique to the niche of residence, evolving during development and aging; (3) inflammation status; and (4) the cumulative effect of time spent in a specific tissue that contributes to the resilient adaptation of macrophages to their dynamic environment. We review recent findings within this context and discuss the technological advances that are revolutionizing the study of macrophage biology.


Assuntos
Biomarcadores , Plasticidade Celular , Macrófagos/imunologia , Macrófagos/metabolismo , Animais , Plasticidade Celular/genética , Plasticidade Celular/imunologia , Microambiente Celular , Suscetibilidade a Doenças , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Ativação de Macrófagos , Macrófagos/classificação , Monócitos/imunologia , Monócitos/metabolismo , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Fenótipo
3.
Immunity ; 50(2): 418-431.e6, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30770245

RESUMO

Sepsis is a bi-phasic inflammatory disease that threatens approximately 30 million lives and claims over 14 million annually, yet little is known regarding the molecular switches and pathways that regulate this disease. Here, we have described ABCF1, an ATP-Binding Cassette (ABC) family member protein, which possesses an E2 ubiquitin enzyme activity, through which it controls the Lipopolysaccharide (LPS)- Toll-like Receptor-4 (TLR4) mediated gram-negative insult by targeting key proteins for K63-polyubiquitination. Ubiquitination by ABCF1 shifts the inflammatory profile from an early phase MyD88-dependent to a late phase TRIF-dependent signaling pathway, thereby regulating TLR4 endocytosis and modulating macrophage polarization from M1 to M2 phase. Physiologically, ABCF1 regulates the shift from the inflammatory phase of sepsis to the endotoxin tolerance phase, and modulates cytokine storm and interferon-ß (IFN-ß)-dependent production by the immunotherapeutic mediator, SIRT1. Consequently, ABCF1 controls sepsis induced mortality by repressing hypotension-induced renal circulatory dysfunction.


Assuntos
Transportadores de Cassetes de Ligação de ATP/imunologia , Macrófagos/imunologia , Sepse/imunologia , Choque Séptico/imunologia , Enzimas de Conjugação de Ubiquitina/imunologia , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Interferon beta/imunologia , Interferon beta/metabolismo , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos/classificação , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Interferência de RNA , Sepse/genética , Sepse/metabolismo , Choque Séptico/genética , Choque Séptico/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Enzimas de Conjugação de Ubiquitina/genética , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação/imunologia
4.
Nature ; 572(7771): 670-675, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31391580

RESUMO

Macrophages are considered to contribute to chronic inflammatory diseases such as rheumatoid arthritis1. However, both the exact origin and the role of macrophages in inflammatory joint disease remain unclear. Here we use fate-mapping approaches in conjunction with three-dimensional light-sheet fluorescence microscopy and single-cell RNA sequencing to perform a comprehensive spatiotemporal analysis of the composition, origin and differentiation of subsets of macrophages within healthy and inflamed joints, and study the roles of these macrophages during arthritis. We find that dynamic membrane-like structures, consisting of a distinct population of CX3CR1+ tissue-resident macrophages, form an internal immunological barrier at the synovial lining and physically seclude the joint. These barrier-forming macrophages display features that are otherwise typical of epithelial cells, and maintain their numbers through a pool of locally proliferating CX3CR1- mononuclear cells that are embedded into the synovial tissue. Unlike recruited monocyte-derived macrophages, which actively contribute to joint inflammation, these epithelial-like CX3CR1+ lining macrophages restrict the inflammatory reaction by providing a tight-junction-mediated shield for intra-articular structures. Our data reveal an unexpected functional diversification among synovial macrophages and have important implications for the general role of macrophages in health and disease.


Assuntos
Articulações/citologia , Macrófagos/citologia , Macrófagos/fisiologia , Membrana Sinovial/citologia , Sinoviócitos/citologia , Sinoviócitos/fisiologia , Junções Íntimas/fisiologia , Animais , Artrite/imunologia , Artrite/patologia , Receptor 1 de Quimiocina CX3C/análise , Receptor 1 de Quimiocina CX3C/metabolismo , Rastreamento de Células , Feminino , Perfilação da Expressão Gênica , Humanos , Inflamação/imunologia , Inflamação/patologia , Articulações/patologia , Macrófagos/classificação , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Componente Principal , RNA-Seq , Análise de Célula Única , Sinoviócitos/classificação , Sinoviócitos/metabolismo , Transcriptoma/genética
5.
Immunity ; 42(3): 419-30, 2015 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-25786174

RESUMO

Macrophage polarization involves a coordinated metabolic and transcriptional rewiring that is only partially understood. By using an integrated high-throughput transcriptional-metabolic profiling and analysis pipeline, we characterized systemic changes during murine macrophage M1 and M2 polarization. M2 polarization was found to activate glutamine catabolism and UDP-GlcNAc-associated modules. Correspondingly, glutamine deprivation or inhibition of N-glycosylation decreased M2 polarization and production of chemokine CCL22. In M1 macrophages, we identified a metabolic break at Idh, the enzyme that converts isocitrate to alpha-ketoglutarate, providing mechanistic explanation for TCA cycle fragmentation. (13)C-tracer studies suggested the presence of an active variant of the aspartate-arginosuccinate shunt that compensated for this break. Consistently, inhibition of aspartate-aminotransferase, a key enzyme of the shunt, inhibited nitric oxide and interleukin-6 production in M1 macrophages, while promoting mitochondrial respiration. This systems approach provides a highly integrated picture of the physiological modules supporting macrophage polarization, identifying potential pharmacologic control points for both macrophage phenotypes.


Assuntos
Redes Reguladoras de Genes/imunologia , Imunidade Inata , Macrófagos/metabolismo , Mitocôndrias/metabolismo , Transcrição Gênica/imunologia , Animais , Ácido Argininossuccínico/imunologia , Ácido Argininossuccínico/metabolismo , Aspartato Aminotransferase Mitocondrial/genética , Aspartato Aminotransferase Mitocondrial/imunologia , Ácido Aspártico/imunologia , Ácido Aspártico/metabolismo , Quimiocina CCL22/genética , Quimiocina CCL22/imunologia , Ciclo do Ácido Cítrico , Regulação da Expressão Gênica , Glutamina/deficiência , Glicosilação , Interleucina-6/genética , Interleucina-6/imunologia , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/imunologia , Macrófagos/classificação , Macrófagos/citologia , Macrófagos/imunologia , Redes e Vias Metabólicas/genética , Redes e Vias Metabólicas/imunologia , Camundongos , Mitocôndrias/genética , Mitocôndrias/imunologia , Óxido Nítrico/imunologia , Óxido Nítrico/metabolismo , Transdução de Sinais , Uridina Difosfato N-Acetilglicosamina/imunologia , Uridina Difosfato N-Acetilglicosamina/metabolismo
6.
Biochem Biophys Res Commun ; 596: 63-70, 2022 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-35114586

RESUMO

Owing to lacking protective effect of estrogen, OVX mice have higher risk of non-alcoholic fatty liver disease compared with normal female mice, when fed with high fat diet. Our study was to explore how estrogen protect against nonalcoholic steatohepatitis in female mice. We found that, lacking estrogen, M1 macrphages was activated and promoted steatohepatitis in obese OVX mice. And, ERα was responsible for estrogen to inhibit M1 macrphages activation and steatohepatitis. ERα knockdown aggravated M1 macrophages infiltration by transcriptionally upregulated its CCR2 expression. CCR2 antagonist effectively improved nonalcoholic steatohepatitis, ER stress and insulin resistance in ERα knockdown obese female mice. These results demonstrated ERα mediated M1 macrophages activation played a key role in nonalcoholic steatohepatitis.


Assuntos
Receptor alfa de Estrogênio/genética , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Hepatopatia Gordurosa não Alcoólica/genética , Animais , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Dieta Hiperlipídica/efeitos adversos , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/classificação , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ovariectomia , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , Interferência de RNA , Receptores CCR2/genética , Receptores CCR2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Pharmacol Sci ; 148(1): 116-124, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34924115

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease with increased M1 macrophages. The classical activated M1 macrophages produce various cytokines to control inflammation. Wilforlide A is a natural product that displays anti-inflammatory activities. However, the effect of Wilforlide A on RA progression and the potential mechanisms are unclear. Herein, the collagen-induced arthritis (CIA) mouse was used as an experimental model of RA. The administration of Wilforlide A reduced clinical scores, joint swelling and histological damage in ankle joints of RA mice. The secreted pro-inflammatory factors (MCP1, GM-CSF and M-CSF) and M1 biomarker iNOS in synovium were inhibited by Wilforlide A. In vitro, macrophages deriving from THP-1 cells were stimulated with LPS/IFN-γ to mimic M1 polarization. Similarly, Wilforlide A blocked macrophages polarizing towards M1 subsets. The in vitro results demonstrated that Wilforlide A suppressed LPS/IFN-γ-induced TLR4 upregulation, IκBα degradation and NF-κB p65 activation. In addition, TAK242 (a TLR4 inhibitor) treatment caused a similar inhibitory effect on M1 polarization with Wilforlide A, whereas it was less than the combination of TAK242 and Wilforlide A. Therefore, this work supports that Wilforlide A ameliorates M1 macrophage polarization in RA, which is partially mediated by TLR4/NF-κB signaling pathway inactivation.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Polaridade Celular/efeitos dos fármacos , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Ácido Oleanólico/análogos & derivados , Fitoterapia , Animais , Anti-Inflamatórios , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Mediadores da Inflamação/metabolismo , Macrófagos/classificação , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos DBA , NF-kappa B/metabolismo , Ácido Oleanólico/farmacologia , Ácido Oleanólico/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Receptor 4 Toll-Like/metabolismo
8.
Proc Natl Acad Sci U S A ; 116(47): 23705-23713, 2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31685620

RESUMO

Inflammation plays an important role in pathological angiogenesis. Receptor-interacting protein 1 (RIP1) is highly expressed in inflammatory cells and is known to play an important role in the regulation of apoptosis, necroptosis, and inflammation; however, a comprehensive description of its role in angiogenesis remains elusive. Here, we show that RIP1 is abundantly expressed in infiltrating macrophages during angiogenesis, and genetic or pharmacological inhibition of RIP1 kinase activity using kinase-inactive RIP1K45A/K45A mice or necrostatin-1 attenuates angiogenesis in laser-induced choroidal neovascularization, Matrigel plug angiogenesis, and alkali injury-induced corneal neovascularization in mice. The inhibitory effect on angiogenesis is mediated by caspase activation through a kinase-independent function of RIP1 and RIP3. Mechanistically, infiltrating macrophages are the key target of RIP1 kinase inhibition to attenuate pathological angiogenesis. Inhibition of RIP1 kinase activity is associated with caspase activation in infiltrating macrophages and decreased expression of proangiogenic M2-like markers but not M1-like markers. Similarly, in vitro, catalytic inhibition of RIP1 down-regulates the expression of M2-like markers in interleukin-4-activated bone marrow-derived macrophages, and this effect is blocked by simultaneous caspase inhibition. Collectively, these results demonstrate a nonnecrotic function of RIP1 kinase activity and suggest that RIP1-mediated modulation of macrophage activation may be a therapeutic target of pathological angiogenesis.


Assuntos
Proteínas Ativadoras de GTPase/fisiologia , Macrófagos/fisiologia , Neovascularização Patológica/enzimologia , Animais , Biomarcadores , Caspases/metabolismo , Células Cultivadas , Colágeno , Lesões da Córnea/induzido quimicamente , Lesões da Córnea/etiologia , Neovascularização da Córnea/enzimologia , Neovascularização da Córnea/etiologia , Neovascularização da Córnea/patologia , Neovascularização da Córnea/prevenção & controle , Combinação de Medicamentos , Ativação Enzimática , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Células Endoteliais da Veia Umbilical Humana , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Marcação In Situ das Extremidades Cortadas , Indóis/farmacologia , Indóis/uso terapêutico , Laminina , Lasers/efeitos adversos , Macrófagos/classificação , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neovascularização Patológica/patologia , Oligopeptídeos/farmacologia , Proteoglicanas , RNA Mensageiro/biossíntese , Proteína Serina-Treonina Quinases de Interação com Receptores/deficiência , Proteína Serina-Treonina Quinases de Interação com Receptores/fisiologia , Receptores de Fatores de Crescimento do Endotélio Vascular/uso terapêutico , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico
9.
Circulation ; 142(5): 466-482, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32403949

RESUMO

BACKGROUND: The human heart requires a complex ensemble of specialized cell types to perform its essential function. A greater knowledge of the intricate cellular milieu of the heart is critical to increase our understanding of cardiac homeostasis and pathology. As recent advances in low-input RNA sequencing have allowed definitions of cellular transcriptomes at single-cell resolution at scale, we have applied these approaches to assess the cellular and transcriptional diversity of the nonfailing human heart. METHODS: Microfluidic encapsulation and barcoding was used to perform single nuclear RNA sequencing with samples from 7 human donors, selected for their absence of overt cardiac disease. Individual nuclear transcriptomes were then clustered based on transcriptional profiles of highly variable genes. These clusters were used as the basis for between-chamber and between-sex differential gene expression analyses and intersection with genetic and pharmacologic data. RESULTS: We sequenced the transcriptomes of 287 269 single cardiac nuclei, revealing 9 major cell types and 20 subclusters of cell types within the human heart. Cellular subclasses include 2 distinct groups of resident macrophages, 4 endothelial subtypes, and 2 fibroblast subsets. Comparisons of cellular transcriptomes by cardiac chamber or sex reveal diversity not only in cardiomyocyte transcriptional programs but also in subtypes involved in extracellular matrix remodeling and vascularization. Using genetic association data, we identified strong enrichment for the role of cell subtypes in cardiac traits and diseases. Intersection of our data set with genes on cardiac clinical testing panels and the druggable genome reveals striking patterns of cellular specificity. CONCLUSIONS: Using large-scale single nuclei RNA sequencing, we defined the transcriptional and cellular diversity in the normal human heart. Our identification of discrete cell subtypes and differentially expressed genes within the heart will ultimately facilitate the development of new therapeutics for cardiovascular diseases.


Assuntos
Miocárdio/citologia , Transcrição Gênica , Adipócitos/metabolismo , Adulto , Idoso , Fármacos Cardiovasculares/farmacologia , Fármacos Cardiovasculares/uso terapêutico , Células Endoteliais/classificação , Células Endoteliais/metabolismo , Fibroblastos/classificação , Fibroblastos/metabolismo , Ontologia Genética , Coração/inervação , Átrios do Coração/citologia , Cardiopatias/tratamento farmacológico , Ventrículos do Coração/citologia , Homeostase , Humanos , Subpopulações de Linfócitos/metabolismo , Macrófagos/classificação , Macrófagos/metabolismo , Técnicas Analíticas Microfluídicas , Pessoa de Meia-Idade , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/metabolismo , Pericitos/metabolismo , RNA-Seq , Caracteres Sexuais , Análise de Célula Única , Transcriptoma
10.
Lab Invest ; 101(10): 1382-1393, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34158590

RESUMO

Hepatorenal fibrocystic disease (HRFCD) is a genetically inherited disorder related to primary cilia dysfunction in which patients display varying levels of fibrosis, bile duct expansion, and inflammation. In mouse models of HRFCD, the phenotype is greatly impacted by the genetic background in which the mutation is placed. Macrophages are a common factor associated with progression of HRFCD and are also strongly influenced by the genetic background. These data led us to hypothesize that macrophage subtypes that change in relation to the genetic background are responsible for the variable phenotypic outcomes in HRFCD. To test this hypothesis, we utilized a mouse model of HRFCD (Ift88Orpk mice) on the C57BL/6 and BALB/c inbred backgrounds that have well-documented differences in macrophage subtypes. Our analyses of infiltrating macrophage subtypes confirm that genetic strain influences the subtype of infiltrating macrophage present during normal postnatal liver development and in Ift88Orpk livers (Ly6clo in C57BL/6 vs Ly6chi in BALB/c). Each infiltrating macrophage subtype was similarly associated with a unique phenotypic outcome as analysis of liver tissue shows that C57BL/6 Ift88Orpk mice have increased bile duct expansion, but reduced levels of fibrosis compared to BALB/c Ift88Orpk livers. RNA sequencing data suggest that the ability to infiltrate macrophage subtypes to influence the phenotypic outcome may be due to unique ligand-receptor signaling between infiltrating macrophages and cilia dysfunctional biliary epithelium. To evaluate whether specific macrophage subtypes cause the observed phenotypic divergence, we analyzed the liver phenotype in BALB/c Ift88Orpk mice on a CCR2-/- background. Unexpectedly, the loss of Ly6chi macrophages, which were strongly enriched in BALB/c Ift88Orpk mice, did not significantly alter liver fibrosis. These data indicate that macrophage subtypes may correlate with HRFCD phenotypic outcome, but do not directly cause the pathology.


Assuntos
Cirrose Hepática , Macrófagos , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Fígado/metabolismo , Macrófagos/classificação , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fenótipo
11.
Clin Immunol ; 229: 108764, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34089860

RESUMO

C57BL/6 mice with pristane-induced lupus develop macrophage-dependent diffuse alveolar hemorrhage (DAH), which is blocked by treatment with liver X receptor (LXR) agonists and is exacerbated by low IL-10 levels. Serp-1, a myxomavirus-encoded serpin that impairs macrophage activation and plasminogen activation, blocks DAH caused by MHV68 infection. We investigated whether Serp-1 also could block DAH in pristane-induced lupus. Pristane-induced DAH was prevented by treatment with recombinant Serp-1 and macrophages from Serp1-treated mice exhibited an anti-inflammatory M2-like phenotype. Therapy activated LXR, promoting M2 polarization and expression of Kruppel-like factor-4 (KLH4), which upregulates IL-10. In contrast, deficiency of tissue plasminogen activator or plasminogen activator inhibitor had little effect on DAH. We conclude that Serp-1 blocks pristane-induced lung hemorrhage by enhancing LXR-regulated M2 macrophage polarization and KLH4-regulated IL-10 production. In view of the similarities between DAH in pristane-treated mice and SLE patients, Serp-1 may represent a potential new therapy for this severe complication of SLE.


Assuntos
Lúpus Eritematoso Sistêmico/terapia , Macrófagos/efeitos dos fármacos , Serpinas/farmacologia , Proteínas Virais/farmacologia , Animais , Coagulação Sanguínea , Feminino , Hemorragia/sangue , Hemorragia/patologia , Hemorragia/prevenção & controle , Interleucina-10/biossíntese , Fator 4 Semelhante a Kruppel , Receptores X do Fígado/metabolismo , Pneumopatias/sangue , Pneumopatias/patologia , Pneumopatias/prevenção & controle , Lúpus Eritematoso Sistêmico/induzido quimicamente , Lúpus Eritematoso Sistêmico/imunologia , Macrófagos/classificação , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Myxoma virus/genética , Células RAW 264.7 , Serpinas/genética , Terpenos/toxicidade , Proteínas Virais/genética
12.
Cytokine ; 137: 155305, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33002742

RESUMO

A way to delay aging and the related low-grade chronic inflammatory state is to study the model of positive physiology such as the Long-Living Individuals (LLIs). Our recent studies have shown higher levels of the host defense BPI Fold-Containing Family B Member 4 (BPIFB4) protein in the LLIs' blood. Notably, BPIFB4 has been shown to influence monocytes typesetting and M2 anti-inflammatory phenotype (CD206+CD163++) macrophages skewing. According to the role of a complex cytokine milieu in guiding the macrophage polarization, here we found that circulating concentrations of thymus and activation regulated chemokine (TARC)/CCL17 and small-inducible cytokine B10 (IP-10)/CXCL10) cytokines, were additionally associated with the LLIs' state. In a differentiation process in vitro, the addition of LLIs' plasma to the cell culture medium, enhanced the ability of monocytes, either from LLIs or controls, to acquire a M2 phenotype. Interestingly, a neutralizing antibody against TARC blunted the M2 skewing effect of the LLIs' plasma. Collectively, these data indicate that exceptional longevity may associate with a peculiar anti-inflammatory myeloid profile responsible for improved reparative processes and reduced inflammatory status mediated in part by TARC and M2 generation.


Assuntos
Envelhecimento/sangue , Quimiocina CCL17/sangue , Longevidade , Macrófagos/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Quimiocina CXCL10/sangue , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Ativação de Macrófagos , Macrófagos/classificação , Macrófagos/imunologia , Masculino , Pessoa de Meia-Idade
13.
FASEB J ; 34(5): 6888-6906, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32232901

RESUMO

Previously, we identified a mechanism of inflammation control directed by ribosomal protein L13a and "GAIT" (Gamma Activated Inhibitor of Translation) elements in target mRNAs and showed that its elimination in myeloid cell-specific L13a knockout mice (L13a KO) increased atherosclerosis susceptibility and severity. Here, we investigated the mechanistic basis of this endogenous defense against atherosclerosis. We compared molecular and cellular aspects of atherosclerosis in high-fat diet (HFD)-fed L13a KO and intact (control) mice. HFD treatment of control mice induced release of L13a from 60S ribosome, formation of RNA-binding complex, and subsequent GAIT element-mediated translational silencing. Atherosclerotic plaques from HFD-treated KO mice showed increased infiltration of M1 type inflammatory macrophages. Macrophages from KO mice showed increased phagocytic activity and elevated expression of LDL receptor and pro-inflammatory mediators. NanoString analysis of the plaques from KO mice showed upregulation of a number of mRNAs encoding inflammatory proteins. Bioinformatics analysis suggests the presence of the potential GAIT elements in the 3'UTRs of several of these mRNAs. Macrophage induces L13a/GAIT-dependent translational silencing of inflammatory genes in response to HFD as an endogenous defense against atherosclerosis in ApoE-/- model.


Assuntos
Aterosclerose/prevenção & controle , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Proteínas Ribossômicas/deficiência , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Diferenciação Celular , Colesterol/metabolismo , Dieta Hiperlipídica/efeitos adversos , Feminino , Macrófagos/classificação , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Knockout para ApoE , Células Mieloides/metabolismo , Células Mieloides/patologia , Fagocitose , Placa Aterosclerótica/etiologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de LDL/metabolismo , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo
14.
FASEB J ; 34(5): 6984-6998, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32232913

RESUMO

Rictor is an essential component that directly activates the mammalian target of rapamycin (mTOR) activity, which contributes to the intrinsic axon growth capacity of adult sensory neurons after injury. However, whether its action also applies to regeneration after spinal cord injury (SCI) remains unknown. In this study, rats were given spinal cord contusion at the T9-10 level to establish the SCI model and were subsequently treated with intraspinal cord injection of a Rictor overexpression lentiviral vector to locally upregulate the Rictor expression in the injured spinal cord. Thereafter, we investigated the therapeutic effects of Rictor overexpression in the injured spinal cords of SCI rats. Rictor overexpression not only significantly attenuated the acute inflammatory response and cell death after SCI but also markedly increased the shift in macrophages around the lesion from the M1 to M2 phenotype compared to those of the control lentiviral vector injection-treated group. Furthermore, Rictor overexpression dramatically increased neurogenesis in the lesion epicenter, subsequently promoting the tissue repair and functional recovery in SCI rats. Interestingly, the mechanism underlying the beneficial effects of Rictor overexpression on SCI may be associated with the Rictor overexpression playing a role in the anti-inflammatory response and driving macrophage polarization toward the M2 phenotype, which benefits resident neuronal and oligodendrocyte survival. Our findings demonstrate that Rictor is an effective target that affects the generation of molecules that inhibit spinal cord regeneration. In conclusion, localized Rictor overexpression represents a promising potential strategy for the repair of SCI.


Assuntos
Proteína Companheira de mTOR Insensível à Rapamicina/fisiologia , Traumatismos da Medula Espinal/terapia , Animais , Apoptose , Sobrevivência Celular , Modelos Animais de Doenças , Feminino , Humanos , Macrófagos/classificação , Macrófagos/metabolismo , Macrófagos/patologia , Neurônios Motores/patologia , Neurônios Motores/fisiologia , Plasticidade Neuronal , Oligodendroglia/patologia , Oligodendroglia/fisiologia , Proteína Companheira de mTOR Insensível à Rapamicina/genética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recuperação de Função Fisiológica/genética , Recuperação de Função Fisiológica/fisiologia , Remielinização , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Regulação para Cima
15.
Circ Res ; 125(3): 265-281, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31318648

RESUMO

Advanced heart failure (HF) is a progressive disease characterized by recurrent hospitalizations and high risk of mortality. Indeed, outcomes in late stages of HF approximate those seen in patients with various aggressive malignancies. Clinical trials assessing beneficial outcomes of new treatments in patients with cancer have used innovative approaches to measure impact on total disease burden or surrogates to assess treatment efficacy. Although most cardiovascular outcomes trials continue to use time-to-first event analyses to assess the primary efficacy end point, such analyses do not adequately reflect the impact of new treatments on the totality of the chronic disease burden. Consequently, patient enrichment and other strategies for ongoing clinical trial design, as well as new statistical methodologies, are important considerations, particularly when studying a population with advanced chronic HF. The DREAM-HF trial (Double-Blind Randomized Assessment of Clinical Events With Allogeneic Mesenchymal Precursor Cells in Advanced Heart Failure) is an ongoing, randomized, sham-controlled phase 3 study of the efficacy and safety of mesenchymal precursor cells as immunotherapy in patients with advanced chronic HF with reduced ejection fraction. Mesenchymal precursor cells have a unique multimodal mechanism of action that is believed to result in polarization of proinflammatory type 1 macrophages in the heart to an anti-inflammatory type 2 macrophage state, inhibition of maladaptive adverse left ventricular remodeling, reversal of cardiac and peripheral endothelial dysfunction, and recovery of deranged vasculature. The objective of DREAM-HF is to confirm earlier phase 2 results and evaluate whether mesenchymal precursor cells will reduce the rate of nonfatal recurrent HF-related major adverse cardiac events while delaying or preventing progression of HF to terminal cardiac events. DREAM-HF is an example of an ongoing contemporary events-driven cardiovascular cell-based immunotherapy study that has utilized the concepts of baseline disease enrichment, prognostic enrichment, and predictive enrichment to improve its efficiency by using accumulating data from within as well as external to the trial. Adaptive enrichment designs and strategies are important components of a rational approach to achieve clinical research objectives in shorter clinical trial timelines and with increased cost-effectiveness without compromising ethical standards or the overall statistical integrity of the study. The DREAM-HF trial also presents an alternative approach to traditional composite time-to-first event primary efficacy end points. Statistical methodologies such as the joint frailty model provide opportunities to expand the scope of events-driven HF with reduced ejection fraction clinical trials to utilize time to recurrent nonfatal HF-related major adverse cardiac events as the primary efficacy end point without compromising the integrity of the statistical analyses for terminal cardiac events. In advanced chronic HF with reduced ejection fraction studies, the joint frailty model is utilized to reflect characteristics of the high-risk patient population with important unmet therapeutic needs. In some cases, use of the joint frailty model may substantially reduce sample size requirements. In addition, using an end point that is acceptable to the Food and Drug Administration and the European Medicines Agency, such as recurrent nonfatal HF-related major adverse cardiac events, enables generation of clinically relevant pharmacoeconomic data while providing comprehensive views of the patient's overall cardiovascular disease burden. The major goal of this review is to provide lessons learned from the ongoing DREAM-HF trial that relate to biologic plausibility and flexible clinical trial design and are potentially applicable to other development programs of innovative therapies for patients with advanced cardiovascular disease. Clinical Trial Registration: URL: https://www.clinicaltrials.gov. Unique identifier: NCT02032004.


Assuntos
Ensaios Clínicos Fase III como Assunto/métodos , Insuficiência Cardíaca/terapia , Imunoterapia/métodos , Transplante de Células-Tronco Mesenquimais , Estudos Multicêntricos como Assunto/métodos , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Diferenciação Celular , Método Duplo-Cego , Endotélio Vascular/fisiopatologia , Determinação de Ponto Final , Necessidades e Demandas de Serviços de Saúde , Insuficiência Cardíaca/economia , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/fisiopatologia , Humanos , Inflamação , Macrófagos/classificação , Macrófagos/imunologia , Neovascularização Patológica/etiologia , Projetos de Pesquisa , Volume Sistólico , Resultado do Tratamento , Remodelação Ventricular
16.
Cell Mol Life Sci ; 77(10): 1919-1932, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31720740

RESUMO

The presence of macrophages within the plaque is a defining hallmark of atherosclerosis. Macrophages are exposed to various microenvironments such as oxidized lipids and cytokines which effect their phenotypic differentiation and activation. Classically, macrophages have been divided into two groups: M1 and M2 macrophages induced by T-helper 1 and T-helper 2 cytokines, respectively. However, for a decade, greater phenotypic heterogeneity and plasticity of these cells have since been reported in various models. In addition to M1 and M2 macrophage phenotypes, the concept of additional macrophage phenotypes such as M (Hb), Mox, and M4 has emerged. Understanding the mechanisms and functions of distinct phenotype of macrophages can lead to determination of their potential role in atherosclerotic plaque pathogenesis. However, there are still many unresolved controversies regarding their phenotype and function with respect to atherosclerosis. Here, we summarize and focus on the differential subtypes of macrophages in atherosclerotic plaques and their differing functional roles based upon microenvironments such as lipid, intraplaque hemorrhage, and plaque regression.


Assuntos
Aterosclerose/metabolismo , Metabolismo dos Lipídeos/genética , Macrófagos/metabolismo , Placa Aterosclerótica/metabolismo , Aterosclerose/patologia , Diferenciação Celular/genética , Linhagem da Célula/genética , Microambiente Celular/genética , Citocinas/metabolismo , Humanos , Ativação de Macrófagos/genética , Macrófagos/classificação , Fenótipo , Placa Aterosclerótica/patologia
17.
Proc Natl Acad Sci U S A ; 115(22): E5096-E5105, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29760084

RESUMO

Obesity is characterized by an accumulation of macrophages in adipose, some of which form distinct crown-like structures (CLS) around fat cells. While multiple discrete adipose tissue macrophage (ATM) subsets are thought to exist, their respective effects on adipose tissue, and the transcriptional mechanisms that underlie the functional differences between ATM subsets, are not well understood. We report that obese fat tissue of mice and humans contain multiple distinct populations of ATMs with unique tissue distributions, transcriptomes, chromatin landscapes, and functions. Mouse Ly6c ATMs reside outside of CLS and are adipogenic, while CD9 ATMs reside within CLS, are lipid-laden, and are proinflammatory. Adoptive transfer of Ly6c ATMs into lean mice activates gene programs typical of normal adipocyte physiology. By contrast, adoptive transfer of CD9 ATMs drives gene expression that is characteristic of obesity. Importantly, human adipose tissue contains similar ATM populations, including lipid-laden CD9 ATMs that increase with body mass. These results provide a higher resolution of the cellular and functional heterogeneity within ATMs and provide a framework within which to develop new immune-directed therapies for the treatment of obesity and related sequela.


Assuntos
Tecido Adiposo/citologia , Inflamação/fisiopatologia , Macrófagos , Animais , Exossomos/química , Feminino , Humanos , Inflamação/genética , Macrófagos/química , Macrófagos/classificação , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Obesidade/fisiopatologia , Tetraspanina 29/análise , Tetraspanina 29/metabolismo , Transcriptoma/genética
18.
Biochem Biophys Res Commun ; 529(3): 562-568, 2020 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-32736674

RESUMO

Obesity has been recognized as a low-grade, chronic inflammatory disease that leads to an increase in obesity-associated disorders, including type 2 diabetes (T2D), fatty liver diseases and cancer. Glucagon-like peptide-1 (GLP-1) is an effective drug for T2D, and it not only has glucose-regulating effects but also has anti-inflammatory effects in obesity. In our previous study, we designed a novel GLP-1 analogue, (EX-4)2-Fc, which has been shown to reduce body weight and improve glucose tolerance in vivo. In this study, we observed that (EX-4)2-Fc also has anti-inflammatory functions in adipose tissue. After the treatment of diet-induced obesity (DIO) mice with (EX-4)2-Fc, we found that the inflammatory response in adipose tissue was significantly attenuated. (Ex-4)2-Fc can reduce obesity-associated proinflammatory cytokine levels and macrophage numbers in DIO mice. In addition, (EX-4)2-Fc treatment resulted in proinflammatory M1-type macrophages beginning to transform into anti-inflammatory M2-type macrophages. The inflammatory mitogen-activated protein kinase (MAPK) signalling pathway and nuclear factor kappa B (NF-κB) were altered in adipose tissue after (EX-4)2-Fc treatment. Leptin has been proven to be closely related to immunity, and we demonstrated that the effect of (EX-4)2-Fc on adipocyte inflammation was related to leptin. The data suggested that (EX-4)2-Fc could modulate the inflammatory response by inhibiting the expression of leptin in adipose tissue.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Inflamação/prevenção & controle , Leptina/antagonistas & inibidores , Obesidade/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Citocinas/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Peptídeo 1 Semelhante ao Glucagon/química , Inflamação/metabolismo , Leptina/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/classificação , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Obesos , NF-kappa B/metabolismo , Obesidade/etiologia , Transdução de Sinais/efeitos dos fármacos
19.
Cell Immunol ; 349: 104064, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32061375

RESUMO

The shift of emphasis from short-term to long-term graft outcomes has led to renewed interests in how the innate immune cells regulate transplant survival, an area that is traditionally dominated by T cells in the adaptive system. This shift is driven largely by the limited efficacy of current immunosuppression protocols which primarily target T cells in preventing chronic graft loss, as well as by the rapid advance of basic sciences in the realm of innate immunity. In fact, the innate immune cells have emerged as key players in the allograft response in various models, contributing to both graft rejection and graft acceptance. Here, we focus on the macrophages, highlighting their diversity, plasticity and emerging features in transplant models, as well as recent developments in our studies of diverse subsets of macrophages. We also discuss challenges, unsolved questions, and emerging approaches in therapeutically modulating macrophages in further improvement of transplant outcomes.


Assuntos
Aloenxertos/imunologia , Rejeição de Enxerto/imunologia , Sobrevivência de Enxerto/imunologia , Macrófagos/imunologia , Animais , Antígenos CD/análise , Carragenina/farmacologia , Diferenciação Celular , Linhagem da Célula , Citocinas/imunologia , Humanos , Imunidade Inata , Memória Imunológica , Macrófagos/classificação , Macrófagos/efeitos dos fármacos , Camundongos , Modelos Imunológicos , Células Supressoras Mieloides/imunologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores
20.
Curr Atheroscler Rep ; 22(8): 31, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32556603

RESUMO

PURPOSE: Macrophages play vital roles in the development of atherosclerosis in responding to lipid accumulation and inflammation. Macrophages were classified as inflammatory (M1) and alternatively activated (M2) macrophage types based on results of in vitro experiments. On the other hand, the composition of macrophages in vivo is more complex and remains unresolved. This review summarizes the transcriptional variations of macrophages in atherosclerosis plaques that were discovered by single-cell RNA sequencing (scRNA-seq) to better understand their contribution to atherosclerosis. RECENT FINDINGS: ScRNA-seq provides a more detailed transcriptional landscape of macrophages in atherosclerosis, which challenges the traditional view. By mining the data of GSE97310, we discovered the transcriptional variations of macrophages in LDLR-/- mice that were fed with high-fat diet (HFD) for 11 and 20 weeks. Cells were represented in a two-dimensional tSNE plane and clusters were identified and annotated via Seurat and SingleR respectively, which were R toolkits for single-cell genomics. The results showed that in healthy conditions, Trem2hi (high expression of triggering receptors expressed on myeloid cells 2)-positive, inflammatory, and resident-like macrophages make up 68%, 18%, and 6% of total macrophages respectively. When mice were fed with HFD for 11 weeks, Trem2hi, monocytes, and monocyte-derived dendritic cells take possession of 40%, 18%, and 17% of total macrophages respectively. After 20 weeks of HFD feeding, Trem2hi, inflammatory, and resident-like macrophages occupied 12%, 37%, and 35% of total macrophages respectively. The phenotypes of macrophages are very different from the previous studies. In general, Trem2hi macrophages are the most abundant population in healthy mice, while the proportion of monocytes increases after 11 weeks of HFD. Most importantly, inflammatory and resident-like macrophages make up 70% of the macrophage populations after 20 weeks of HFD. These strongly indicate that inflammatory and resident-like macrophages promote the progression of atherosclerosis plaques.


Assuntos
Aterosclerose/metabolismo , Macrófagos/classificação , Macrófagos/metabolismo , Placa Aterosclerótica/metabolismo , Animais , Células Dendríticas/metabolismo , Humanos , Inflamação/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Monócitos/metabolismo , Fenótipo , RNA-Seq/métodos , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Análise de Célula Única/métodos , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA