Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 458
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35163544

RESUMO

Understanding the interaction between nanoparticles and immune cells is essential for the evaluation of nanotoxicity and development of nanomedicines. However, to date, there is little data on the membrane microstructure and biochemical changes in nanoparticle-loaded immune cells. In this study, we observed the microstructure of nanoparticle-loaded macrophages and changes in lipid droplets using holotomography analysis. Quantitatively analyzing the refractive index distribution of nanoparticle-loaded macrophages, we identified the interactions between nanoparticles and macrophages. The results showed that, when nanoparticles were phagocytized by macrophages, the number of lipid droplets and cell volume increased. The volume and mass of the lipid droplets slightly increased, owing to the absorption of nanoparticles. Meanwhile, the number of lipid droplets increased more conspicuously than the other factors. Furthermore, alveolar macrophages are involved in the development and progression of asthma. Studies have shown that macrophages play an essential role in the maintenance of asthma-related inflammation and tissue damage, suggesting that macrophage cells may be applied to asthma target delivery strategies. Therefore, we investigated the target delivery efficiency of gold nanoparticle-loaded macrophages at the biodistribution level, using an ovalbumin-induced asthma mouse model. Normal and severe asthma models were selected to determine the difference in the level of inflammation in the lung. Consequently, macrophages had increased mobility in models of severe asthma, compared to those of normal asthma disease. In this regard, the detection of observable differences in nanoparticle-loaded macrophages may be of primary interest, as an essential endpoint analysis for investigating nanomedical applications and immunotheragnostic strategies.


Assuntos
Asma/diagnóstico por imagem , Ouro/farmacocinética , Lipopolissacarídeos/efeitos adversos , Pulmão/química , Macrófagos/transplante , Ovalbumina/efeitos adversos , Animais , Asma/induzido quimicamente , Asma/metabolismo , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Estudos de Viabilidade , Feminino , Pulmão/diagnóstico por imagem , Macrófagos/química , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Nanopartículas Metálicas , Camundongos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Células RAW 264.7 , Distribuição Tecidual , Tomografia
2.
Clin Sci (Lond) ; 136(4): 291-307, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35194640

RESUMO

Ulcerative colitis (UC) is majorly associated with dysregulation of the dynamic cross-talk among microbial metabolites, intestinal epithelial cells, and macrophages. Several studies have reported the significant role of butyrate in host-microbiota communication. However, whether butyrate provides anti-inflammatory profiles in macrophages, thus contributing to UC intestinal mucus barrier protection, has currently remained elusive. In the current study, we found that butyrate increased mucin production and the proportion of mucin-secreting goblet cells in the colon crypt in a macrophage-dependent manner by using clodronate liposomes. Furthermore, in vivo and in vitro studies were conducted, validating that butyrate facilitates M2 macrophage polarization with the elevated expressions of CD206 and arginase-1 (Arg1). In macrophages/goblet-like LS174T cells co-culture systems, butyrate-primed M2 macrophages significantly enhanced the expression of mucin-2 (MUC2) and SPDEF (goblet cell marker genes) than butyrate alone, while blockade of WNTs secretion or ERK1/2 activation significantly decreased the beneficial effect of butyrate-primed macrophages on goblet cell function. Additionally, the adoptive transfer of butyrate-induced M2 macrophages facilitated the generation of goblet cells and mucus restoration following dextran sulfate sodium (DSS) insult. Taken together, our results revealed a novel mediator of macrophage-goblet cell cross-talk associated with the regulation of epithelial barrier integrity, implying that the microbial metabolite butyrate may serve as a candidate therapeutic target for UC.


Assuntos
Butiratos/farmacologia , Colite Ulcerativa/terapia , Células Caliciformes/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Via de Sinalização Wnt , Transferência Adotiva , Animais , Estudos de Casos e Controles , Linhagem Celular , Colite Ulcerativa/imunologia , Microbioma Gastrointestinal , Humanos , Macrófagos/metabolismo , Macrófagos/transplante , Camundongos Endogâmicos BALB C
3.
Pharm Biol ; 60(1): 384-393, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35188856

RESUMO

CONTEXT: Therapeutic lymphangiogenesis is a new treatment for cardiovascular diseases. Our previous study showed M2b macrophages can alleviate myocardial ischaemia/reperfusion injury (MI/RI). However, the relation between M2b macrophages and lymphangiogenesis is not clear. OBJECTIVE: To investigate the effects of M2b macrophages on lymphangiogenesis after MI/RI. MATERIALS AND METHODS: Forty male Sprague-Dawley (SD) rats were randomized into Sham operation group (control, n = 8), MI/RI group (n = 16) and M2b macrophage transplantation group (n = 16). M2b macrophages (1 × 106) in 100 µL of normal saline or the same volume of vehicle was injected into the cardiac ischaemic zone. Two weeks later, echocardiography and lymphatic counts were performed, and the extent of myocardial fibrosis and the expression of vascular endothelial growth factor C (VEGFC) and VEGF receptor 3 (VEGFR3) were determined. In vitro, lymphatic endothelial cells (LECs) were cultured with M2b macrophages for 6-24 h, and the proliferation, migration and tube formation of the LECs were assessed. RESULTS: In vivo, M2b macrophage transplantation increased the level of lymphangiogenesis 2.11-fold, reduced 4.42% fibrosis, improved 18.65% left ventricular ejection fraction (LVEF) and upregulated the expressions of VEGFC and VEGFR3. In vitro, M2b macrophage increased the proliferation, migration, tube formation and VEGFC expression of LECs. M2b macrophage supernatant upregulated VEGFR3 expression of LECs. DISCUSSION AND CONCLUSIONS: Our study shows that M2b macrophages can promote lymphangiogenesis to reduce myocardial fibrosis and improve heart function, suggesting the possible use of M2b macrophage for myocardial protection therapy.


Assuntos
Linfangiogênese/fisiologia , Macrófagos/transplante , Traumatismo por Reperfusão Miocárdica/terapia , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Ecocardiografia , Células Endoteliais/metabolismo , Fibrose , Masculino , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Volume Sistólico/fisiologia , Função Ventricular Esquerda/fisiologia
4.
STAR Protoc ; 2(4): 101004, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34917981

RESUMO

Macrophages are key innate immune cells involved in a broad spectrum of physiological and pathological processes. Macrophage depletion with clodronate-liposomes is commonly used to investigate in vivo functions of macrophages in mice. Here, we describe a protocol that combines the depletion of resident macrophages with the reconstitution of the mice with in vitro differentiated, lentivirus-transduced bone marrow-derived macrophages (BMDMs) in the context of an experimental sepsis model. This experimental strategy is easily adapted to other experimental designs. For complete details on the use and execution of this protocol, please refer to Du et al. (2020).


Assuntos
Modelos Animais de Doenças , Macrófagos , Sepse/imunologia , Animais , Diferenciação Celular , Terapia de Imunossupressão , Lentivirus/genética , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/transplante , Camundongos
5.
Front Immunol ; 12: 783305, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899748

RESUMO

Cellular immunotherapies represent a promising approach for the treatment of cancer. Engineered adoptive cell therapies redirect and augment a leukocyte's inherent ability to mount an immune response by introducing novel anti-tumor capabilities and targeting moieties. A prominent example of this approach is the use of T cells engineered to express chimeric antigen receptors (CARs), which have demonstrated significant efficacy against some hematologic malignancies. Despite increasingly sophisticated strategies to harness immune cell function, efficacy against solid tumors has remained elusive for adoptive cell therapies. Amongst cell types used in immunotherapies, however, macrophages have recently emerged as prominent candidates for the treatment of solid tumors. In this review, we discuss the use of monocytes and macrophages as adoptive cell therapies. Macrophages are innate immune cells that are intrinsically equipped with broad therapeutic effector functions, including active trafficking to tumor sites, direct tumor phagocytosis, activation of the tumor microenvironment and professional antigen presentation. We focus on engineering strategies for manipulating macrophages, with a specific focus on CAR macrophages (CAR-M). We highlight CAR design for macrophages, the production of CAR-M for adoptive cell transfer, and clinical considerations for their use in treating solid malignancies. We then outline recent progress and results in applying CAR-M as immunotherapies. The recent development of engineered macrophage-based therapies holds promise as a key weapon in the immune cell therapy armamentarium.


Assuntos
Terapia Genética , Imunoterapia Adotiva , Macrófagos/transplante , Monócitos/transplante , Neoplasias/terapia , Receptores de Antígenos Quiméricos/genética , Animais , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Fenótipo , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/metabolismo , Microambiente Tumoral/imunologia
6.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34638941

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide. An accumulation of fat, followed by inflammation, is the major cause of NAFLD progression. During inflammation, macrophages are the most abundant immune cells recruited to the site of injury. Macrophages are classified into "proinflammatory" M1 macrophages, and "anti-inflammatory" M2 macrophages. In NAFLD, M1 macrophages are the most prominent macrophages that lead to an excessive inflammatory response. Previously, we found that baicalin could polarize macrophages into anti-inflammatory M2c subtype macrophages with an increased level of MERTK expression. Several studies have also shown a strong correlation between MERTK expression and cholesterol efflux, efferocytosis, as well as phagocytosis capability. Therefore, in this study, we aim to elucidate the potential and efficacy of mononuclear-cell (MNC)-derived MERTK+/hi M2c macrophages induced by baicalin as a cell-based therapy for NAFLD treatment. In our results, we have demonstrated that a MERTK+/hi M2c macrophage injection to NAFLD mice contributes to an increased level of serum HDL secretion in the liver, a decline in the circulating CD4+CD25- and CD8+CD25- T cells and lowers the total NAFLD pathological score by lessening the inflammation, necrosis, and fibrosis. In the liver, profibrotic COL1A1 and FN, proinflammation TNFα, as well as the regulator of lipid metabolism PPARÉ£ expression, were also downregulated after injection. In parallel, the transcriptomic profiles of the injected MERTK+/hi M2c macrophages showed that the various genes directly or indirectly involved in NAFLD progression (e.g., SERPINE1, FADS2) were also suppressed. Downregulation of cytokines and inflammation-associated genes, such as CCR5, may promote a pro-resolving milieu in the NAFLD liver. Altogether, cell-based therapy using MERTK+/hi M2c macrophages is promising, as it ameliorates NAFLD in mice.


Assuntos
Transferência Adotiva/métodos , Transplante de Células/métodos , Flavonoides/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/transplante , Hepatopatia Gordurosa não Alcoólica/terapia , Transdução de Sinais/efeitos dos fármacos , c-Mer Tirosina Quinase/metabolismo , Animais , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Lipoproteínas HDL/sangue , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/sangue , Hepatopatia Gordurosa não Alcoólica/genética , PPAR gama/metabolismo , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Transcriptoma , Resultado do Tratamento , Fator de Necrose Tumoral alfa/metabolismo
7.
Biomed Pharmacother ; 144: 112311, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34653754

RESUMO

Although accumulating evidence indicated that modulating macrophage polarization could ameliorate the immune microenvironment and facilitate the repair of spinal cord injury (SCI), the underlying mechanism of macrophage phenotypic switch is still poorly understood. Exosomes (Exos), a potential tool of cell-to-cell communication, may play important roles in cell reprogramming. Herein, we investigated the roles of macrophages-derived exosomes played for macrophage polarization in the SCI immune microenvironment. In this study, we found the fraction of M2 macrophages was markedly decreased after SCI. Moreover, the M2 macrophages-derived exosomes could increase the percentage of M2 macrophages, decrease that of M1 macrophages while the M1 macrophages-derived exosomes acted oppositely. According to the results of in silico analyses and molecular experiments verification, this phenotypic switch might be mediated by the exosomal miRNA-mRNA network, in which the miR-23a-3p/PTEN/PI3K/AKT axis might play an important role. In conclusion, our study suggests macrophage polarization that regulated by various interventions might be mediated by their own exosomes at last. Moreover, M2 macrophages-derived exosomes could promote M2 macrophage polarization via the potential miRNA-mRNA network. Considering its potential of modulating polarization, M2 macrophages-derived exosomes may be a promising therapeutic agent for SCI repair.


Assuntos
Plasticidade Celular , Microambiente Celular , Exossomos/metabolismo , Macrófagos/metabolismo , Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Animais , Microambiente Celular/imunologia , Modelos Animais de Doenças , Exossomos/imunologia , Exossomos/transplante , Macrófagos/imunologia , Macrófagos/transplante , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fenótipo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células RAW 264.7 , Ratos Wistar , Transdução de Sinais , Medula Espinal/imunologia , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/imunologia , Traumatismos da Medula Espinal/cirurgia
8.
J Neuroinflammation ; 18(1): 232, 2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34654458

RESUMO

BACKGROUND: Brain-resident microglia have a distinct origin compared to macrophages in other organs. Under physiological conditions, microglia are maintained by self-renewal from the local pool, independent of hematopoietic progenitors. Pharmacological depletion of microglia during whole-brain radiotherapy prevents synaptic loss and long-term recognition memory deficits. However, the origin or repopulated cells and the mechanisms behind these protective effects are unknown. METHODS: CD45low/int/CD11b+ cells from naïve brains, irradiated brains, PLX5622-treated brains and PLX5622 + whole-brain radiotherapy-treated brains were FACS sorted and sequenced for transcriptomic comparisons. Bone marrow chimeras were used to trace the origin and long-term morphology of repopulated cells after PLX5622 and whole-brain radiotherapy. FACS analyses of intrinsic and exotic synaptic compartments were used to measure phagocytic activities of microglia and repopulated cells. In addition, concussive brain injuries were given to PLX5622 and brain-irradiated mice to study the potential protective functions of repopulated cells after PLX5622 + whole-brain radiotherapy. RESULTS: After a combination of whole-brain radiotherapy and microglia depletion, repopulated cells are brain-engrafted macrophages that originate from circulating monocytes. Comparisons of transcriptomes reveal that brain-engrafted macrophages have an intermediate phenotype that resembles both monocytes and embryonic microglia. In addition, brain-engrafted macrophages display reduced phagocytic activity for synaptic compartments compared to microglia from normal brains in response to a secondary concussive brain injury. Importantly, replacement of microglia by brain-engrafted macrophages spare mice from whole-brain radiotherapy-induced long-term cognitive deficits, and prevent concussive injury-induced memory loss. CONCLUSIONS: Brain-engrafted macrophages prevent radiation- and concussion-induced brain injuries and cognitive deficits.


Assuntos
Lesões Encefálicas/prevenção & controle , Encéfalo/fisiologia , Encéfalo/efeitos da radiação , Fracionamento da Dose de Radiação , Macrófagos/fisiologia , Macrófagos/transplante , Animais , Lesões Encefálicas/radioterapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
9.
Front Immunol ; 12: 744738, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34691050

RESUMO

The murine interleukin-4 treated macrophage (MIL4) exerts anti-inflammatory and pro-healing effects and has been shown to reduce the severity of chemical-induced colitis. Positing M(IL4) transfer as an anti-inflammatory therapy, the possibility of side-effects must be considered. Consequently, bone marrow-derived M(IL4)s were administered via intraperitoneal injection to mice concomitant with Citrobacter rodentium infection (infections colitis), azoxymethane/dextran sodium sulphate (AOM/DSS) treatment [a model of colorectal cancer (CRC)], or ovalbumin sensitization (airway inflammation). The impact of M(IL4) treatment on C. rodentium infectivity, colon histopathology, tumor number and size and tissue-specific inflammation was examined in these models. The anti-colitic effect of the M(IL4)s were confirmed in the di-nitrobenzene sulphonic acid model of colitis and the lumen-to-blood movement of 4kDa FITC-dextran and bacterial translocation to the spleen and liver was also improved by M(IL4) treatment. Analysis of the other models of disease, that represent comorbidities that can occur in human inflammatory bowel disease (IBD), revealed that M(IL4) treatment did not exaggerate the severity of any of the conditions. Rather, there was reduction in the size (but not number) of polyps in the colon of AOM/DSS-mice and reduced infectivity and inflammation in C. rodentium-infected mice in M(IL4)-treated mice. Thus, while any new therapy can have unforeseen side effects, our data confirm and extend the anti-colitic capacity of murine M(IL4)s and indicate that systemic delivery of one million M(IL4)s did not exaggerate disease in models of colonic or airways inflammation or colonic tumorigenesis.


Assuntos
Colite/patologia , Neoplasias do Colo/patologia , Interleucina-4/imunologia , Macrófagos/transplante , Hipersensibilidade Respiratória/patologia , Animais , Inflamação/patologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
10.
Am J Physiol Lung Cell Mol Physiol ; 321(5): L872-L884, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34523355

RESUMO

The influenza virus infection poses a serious health threat worldwide. Myeloid cells play pivotal roles in regulating innate and adaptive immune defense. A disintegrin and metalloproteinase (ADAM) family of proteins contributes to various immune responses; however, the role of a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in influenza virus infection remains largely unknown. Herein, we investigated its role, focusing on myeloid cells, during influenza virus infection in mice. ADAM10 gene (Adam10)flox/flox/Lyz2-Cre (Adam10ΔLyz2) and control Adam10flox/flox mice were intranasally infected with 200 plaque-forming units of influenza virus A/H1N1/PR8/34. Adam10ΔLyz2 mice exhibited a significantly higher mortality rate, stronger lung inflammation, and a higher virus titer in the lungs than control mice. Macrophages and inflammatory cytokines, such as TNF-α, IL-1ß, and CCL2, were increased in bronchoalveolar lavage fluid from Adam10ΔLyz2 mice following infection. CD11b+Ly6G-F4/80+ myeloid cells, which had an inflammatory monocyte/macrophage-like phenotype, were significantly increased in the lungs of Adam10ΔLyz2 mice. Adoptive transfer experiments suggested that these cells likely contributed to the poorer prognosis in Adam10ΔLyz2 mice. Seven days after infection, CD11b+Ly6G-F4/80+ lung cells exhibited significantly higher arginase-1 expression levels in Adam10ΔLyz2 mice than in control mice, whereas an arginase-1 inhibitor improved the prognosis of Adam10ΔLyz2 mice. Enhanced granulocyte-macrophage colony-stimulating factor (GM-CSF)/GM-CSF receptor signaling likely contributed to this process. Collectively, these results indicate that myeloid ADAM10 protects against influenza virus pneumonia and may be a promising therapeutic target.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Arginase/biossíntese , Vírus da Influenza A Subtipo H1N1/metabolismo , Macrófagos/imunologia , Proteínas de Membrana/metabolismo , Células Mieloides/imunologia , Infecções por Orthomyxoviridae/patologia , Proteína ADAM10/genética , Transferência Adotiva/métodos , Secretases da Proteína Precursora do Amiloide/genética , Animais , Arginase/antagonistas & inibidores , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/citologia , Citocinas/análise , Imunidade Inata/imunologia , Macrófagos/transplante , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/transplante , Infecções por Orthomyxoviridae/mortalidade , Infecções por Orthomyxoviridae/prevenção & controle , Prognóstico , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo
11.
Int J Mol Sci ; 22(15)2021 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-34360736

RESUMO

Myeloid regulatory cell-based therapy has been shown to be a promising cell-based medicinal approach in organ transplantation and for the treatment of autoimmune diseases, such as type 1 diabetes, rheumatoid arthritis, Crohn's disease and multiple sclerosis. Dendritic cells (DCs) are the most efficient antigen-presenting cells and can naturally acquire tolerogenic properties through a variety of differentiation signals and stimuli. Several subtypes of DCs have been generated using additional agents, including vitamin D3, rapamycin and dexamethasone, or immunosuppressive cytokines, such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-ß). These cells have been extensively studied in animals and humans to develop clinical-grade tolerogenic (tol)DCs. Regulatory macrophages (Mregs) are another type of protective myeloid cell that provide a tolerogenic environment, and have mainly been studied within the context of research on organ transplantation. This review aims to thoroughly describe the ex vivo generation of tolDCs and Mregs, their mechanism of action, as well as their therapeutic application and assessment in human clinical trials.


Assuntos
Artrite Reumatoide , Terapia Baseada em Transplante de Células e Tecidos , Células Dendríticas , Diabetes Mellitus Tipo 1 , Tolerância Imunológica , Macrófagos , Animais , Artrite Reumatoide/imunologia , Artrite Reumatoide/terapia , Colecalciferol/farmacologia , Células Dendríticas/imunologia , Células Dendríticas/transplante , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Humanos , Interleucina-10/farmacologia , Macrófagos/imunologia , Macrófagos/transplante , Fator de Crescimento Transformador beta/farmacologia
12.
Front Immunol ; 12: 670776, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34413847

RESUMO

Despite significant therapeutic advances, graft-versus-host disease (GvHD) remains the main life-threatening complication following allogeneic hematopoietic stem cell transplantation. The pathogenesis of GvHD is dominated by a dysregulated allogeneic immune response that drives fibrosis and autoimmunity in chronic forms. A multitude of cell therapy approaches, including infusion of myeloid cells, has been proposed to prevent GvHD through tolerance induction but yielded variable results. Myeloid cells like macrophages can be reprogrammed to develop adaptive-like features following antigenic challenge to reinforce or inhibit a subsequent immune response; a phenomenon termed 'trained immunity'. Here we report that, whereas LPSlow-trained macrophages elicit a suppressor effect on allogeneic T cell proliferation and function in vitro in an IL-10-dependent manner, Bacille Calmette et Guérin (BCG)-trained macrophages exert an opposite effect. In a murine model of sclerodermatous chronic GvHD, LPSlow-trained macrophages attenuate clinical signs of GvHD with significant effects on T cell phenotype and function, autoantibodies production, and tissue fibrosis. Furthermore, infusion of LPSlow-macrophages significantly improves survival in mice with acute GvHD. Importantly, we also provide evidence that LPSlow-macrophages do not accelerate A20-lymphoma tumor growth, which is significantly reduced upon transfer of BCG-macrophages. Collectively, these data indicate that macrophages can be trained to significantly inhibit in vitro and in vivo allo-reactive T cell proliferation without exhibiting pro-tumoral effect, thereby opening the way to promising clinical applications.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas , Linfoma/imunologia , Macrófagos/imunologia , Linfócitos T/imunologia , Animais , Carcinogênese , Proliferação de Células , Células Cultivadas , Reprogramação Celular , Feminino , Tolerância Imunológica , Interleucina-10/metabolismo , Lipopolissacarídeos/metabolismo , Macrófagos/transplante , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante Homólogo
13.
Am J Physiol Heart Circ Physiol ; 321(4): H633-H649, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34415184

RESUMO

ß-Adrenergic receptors (ßARs) regulate normal and pathophysiological heart function through their impact on contractility. ßARs are also regulators of immune function where they play a unique role depending on the disease condition and immune cell type. Emerging evidence suggests an important role for the ß2AR subtype in regulating remodeling in the pathological heart; however, the importance of these responses has never been examined. In heart failure, catecholamines are elevated, leading to chronic ßAR activation and contributing to the detrimental effects in the heart. We hypothesized that immune cell ß2AR plays a critical role in the development of heart failure in response to chronic catecholamine elevations through their regulation of immune cell infiltration. To test this, chimeric mice were generated by performing bone marrow transplant (BMT) experiments using wild-type (WT) or ß2AR knockout (KO) donors. WT and ß2ARKO BMT mice were chronically administered the ßAR agonist isoproterenol. Immune cell recruitment to the heart was examined by histology and flow cytometry. Numerous changes in immune cell recruitment were observed with isoproterenol administration in WT BMT mice including proinflammatory myeloid populations and lymphocytes with macrophages made up the majority of immune cells in the heart and which were absent in ß2ARKO BMT animal. ß2ARKO BMT mice had decreased cardiomyocyte death, hypertrophy, and interstitial fibrosis following isoproterenol treatment, culminating in improved function. These findings demonstrate an important role for immune cell ß2AR expression in the heart's response to chronically elevated catecholamines.NEW & NOTEWORTHY Immune cell ß2-adrenergic receptors (ß2ARs) are important for proinflammatory macrophage infiltration to the heart in a chronic isoproterenol administration model of heart failure. Mice lacking immune cell ß2AR have decreased immune cell infiltration to their heart, primarily proinflammatory macrophage populations. This decrease culminated to decreased cardiac injury with lessened cardiomyocyte death, decreased interstitial fibrosis and hypertrophy, and improved function demonstrating that ß2AR regulation of immune responses plays an important role in the heart's response to persistent ßAR stimulation.


Assuntos
Insuficiência Cardíaca/metabolismo , Macrófagos/metabolismo , Miocárdio/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transferência Adotiva , Animais , Transplante de Medula Óssea , Morte Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibrose , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Mediadores da Inflamação/metabolismo , Isoproterenol , Linfócitos/imunologia , Linfócitos/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Macrófagos/transplante , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , Miocárdio/imunologia , Miocárdio/patologia , Fenótipo , Receptores Adrenérgicos beta 2/genética , Transdução de Sinais , Remodelação Ventricular
14.
Nat Commun ; 12(1): 4310, 2021 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34262026

RESUMO

Patients with primary and bone metastatic breast cancer have significantly reduced survival and life quality. Due to the poor drug delivery efficiency of anti-metastasis therapy and the limited response rate of immunotherapy for breast cancer, effective treatment remains a formidable challenge. In this work, engineered macrophages (Oxa(IV)@ZnPc@M) carrying nanomedicine containing oxaliplatin prodrug and photosensitizer are designed as near-infrared (NIR) light-activated drug vectors, aiming to achieve enhanced chemo/photo/immunotherapy of primary and bone metastatic tumors. Oxa(IV)@ZnPc@M exhibits an anti-tumor M1 phenotype polarization and can efficiently home to primary and bone metastatic tumors. Additionally, therapeutics inside Oxa(IV)@ZnPc@M undergo NIR triggered release, which can kill primary tumors via combined chemo-photodynamic therapy and induce immunogenic cell death simultaneously. Oxa(IV)@ZnPc@M combined with anti-PD-L1 can eliminate primary and bone metastatic tumors, activate tumor-specific antitumor immune response, and improve overall survival with limited systemic toxicity. Therefore, this all-in-one macrophage provides a treatment platform for effective therapy of primary and bone metastatic tumors.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Macrófagos/transplante , Fotoquimioterapia/métodos , Animais , Apoptose/efeitos dos fármacos , Antígeno B7-H1/antagonistas & inibidores , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/secundário , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Portadores de Fármacos/química , Feminino , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Morte Celular Imunogênica/efeitos dos fármacos , Memória Imunológica/efeitos dos fármacos , Indóis/administração & dosagem , Indóis/química , Indóis/farmacologia , Raios Infravermelhos , Macrófagos/química , Nanomedicina , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/química , Compostos Organometálicos/farmacologia , Oxaliplatina/administração & dosagem , Oxaliplatina/química , Oxaliplatina/farmacologia , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Pró-Fármacos/farmacologia
15.
Cells ; 10(7)2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34209378

RESUMO

Phagocytosis is an inherent function of tissue macrophages for the removal of apoptotic cells and cellular debris during acute and chronic injury; however, the dynamics of this event during fibrosis development is unknown. We aim to prove that during the development of kidney fibrosis in the unilateral ureteral obstruction (UUO) model, there are some populations of macrophage with a reduced ability to phagocytose, and whether the infusion of a population of phagocytic macrophages could reduce fibrosis in the murine model UUO. For this purpose, we have identified the macrophage populations during the development of fibrosis and have characterized their phagocytic ability and their expression of CPT1a. Furthermore, we have evaluated the therapeutic effect of macrophages overexpressing CPT1a with high phagocytic skills. We evidenced that the macrophage population which exhibits high phagocytic ability (F4/80low-CD11b) in fibrotic animals decreases during the progression of fibrosis while the macrophage population with lower phagocytic ability (F4/80high-CD11b) in fibrotic conditions, conversely, increases and CPT1a macrophage cell therapy with a strengthening phagocytic ability is associated with a therapeutic effect on kidney fibrosis. We have developed a therapeutic approach to reduce fibrosis in the UUO model by enrichment of the kidney resident macrophage population with a higher proportion of exogenous phagocytic macrophages overexpressing CPT1a.


Assuntos
Carnitina O-Palmitoiltransferase/metabolismo , Rim/patologia , Macrófagos/transplante , Fagocitose , Obstrução Ureteral/complicações , Animais , Antígeno CD11b/metabolismo , Carnitina O-Palmitoiltransferase/genética , Terapia Baseada em Transplante de Células e Tecidos , Modelos Animais de Doenças , Progressão da Doença , Fibrose , Perfilação da Expressão Gênica , Masculino , Camundongos , Células RAW 264.7
16.
Nat Rev Mol Cell Biol ; 22(9): 608-624, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34079104

RESUMO

Liver regeneration is a complex process involving the crosstalk of multiple cell types, including hepatocytes, hepatic stellate cells, endothelial cells and inflammatory cells. The healthy liver is mitotically quiescent, but following toxic damage or resection the cells can rapidly enter the cell cycle to restore liver mass and function. During this process of regeneration, epithelial and non-parenchymal cells respond in a tightly coordinated fashion. Recent studies have described the interaction between inflammatory cells and a number of other cell types in the liver. In particular, macrophages can support biliary regeneration, contribute to fibrosis remodelling by repressing hepatic stellate cell activation and improve liver regeneration by scavenging dead or dying cells in situ. In this Review, we describe the mechanisms of tissue repair following damage, highlighting the close relationship between inflammation and liver regeneration, and discuss how recent findings can help design novel therapeutic approaches.


Assuntos
Cirrose Hepática/patologia , Cirrose Hepática/terapia , Regeneração Hepática/fisiologia , Transplante de Células , Células Epiteliais/citologia , Células Epiteliais/transplante , Células Estreladas do Fígado/metabolismo , Hepatócitos/citologia , Hepatócitos/patologia , Hepatócitos/transplante , Humanos , Inflamação , Macrófagos/citologia , Macrófagos/patologia , Macrófagos/transplante , Transdução de Sinais
17.
Biochim Biophys Acta Rev Cancer ; 1876(1): 188547, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33932561

RESUMO

Chimeric antigen receptor (CAR) T cell treatment has provided notable results in hematological tumors. Unfortunately, this evidence has not been translated into improved outcomes in solid malignancies so far, where several reports have suggested that T cells encounter substantial difficulties in penetrating and surviving in the tumor microenvironment (TME). Thus, researchers have recently investigated other immune cell types as CAR platforms, in order to overcome the limitations of CAR T cells. Among them, CAR-macrophages (M) technology has emerged as a novel perspective for cancer patients, on the basis of preclinical studies observing that CAR expression in human macrophages could play a crucial role in enhancing phagocytosis, polarizing M2 to M1 phenotype, and stimulating T cell anti-tumor activity. Herein, we provide an overview of current scenario of CAR-Ms in several solid tumors, also focusing on the biological rationale behind this promising therapeutic approach and future research directions in this setting.


Assuntos
Plasticidade Celular , Terapia Genética , Imunoterapia Adotiva , Macrófagos/transplante , Nanomedicina , Neoplasias/terapia , Receptores de Antígenos Quiméricos/metabolismo , Macrófagos Associados a Tumor/metabolismo , Animais , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Nanopartículas , Neoplasias/genética , Neoplasias/imunologia , Neoplasias/metabolismo , Fenótipo , Receptores de Antígenos Quiméricos/genética , Microambiente Tumoral , Macrófagos Associados a Tumor/imunologia
18.
Cell Prolif ; 54(6): e13050, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33960559

RESUMO

OBJECTIVES: In this study, we study the transplantation of tauroursodeoxycholic acid (TUDCA)-induced M2-phenotype (M2) macrophages and their ability to promote anti-neuroinflammatory effects and functional recovery in a spinal cord injury (SCI) model. METHODS: To this end, compared to the granulocyte-macrophage colony-stimulating factor (GM-CSF), we evaluated whether TUDCA effectively differentiates bone marrow-derived macrophages (BMDMs) into M2 macrophages. RESULTS: The M2 expression markers in the TUDCA-treated BMDM group were increased more than those in the GM-CSF-treated BMDM group. After the SCI and transplantation steps, pro-inflammatory cytokine levels and the mitogen-activated protein kinase (MAPK) pathway were significantly decreased in the TUDCA-induced M2 group more than they were in the GM-CSF-induced M1 group and in the TUDCA group. Moreover, the TUDCA-induced M2 group showed significantly enhanced tissue volumes and improved motor functions compared to the GM-CSF-induced M1 group and the TUDCA group. In addition, biotinylated dextran amine (BDA)-labelled corticospinal tract (CST) axons and neuronal nuclei marker (NeuN) levels were increased in the TUDCA-induced M2 group more than those in the GM-CSF-induced M1 group and the TUDCA group. CONCLUSIONS: This study demonstrates that the transplantation of TUDCA-induced M2 macrophages promotes an anti-neuroinflammatory effect and motor function recovery in SCI. Therefore, we suggest that the transplantation of TUDCA-induced M2 macrophages represents a possible alternative cell therapy for SCI.


Assuntos
Macrófagos/transplante , Traumatismos da Medula Espinal/terapia , Ácido Tauroquenodesoxicólico/metabolismo , Animais , Células Cultivadas , Feminino , Inflamação/metabolismo , Inflamação/fisiopatologia , Inflamação/terapia , Macrófagos/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia
19.
Front Immunol ; 12: 682871, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34040616

RESUMO

Macrophages are cells that mediate both innate and adaptive immunity reactions, playing a major role in both physiological and pathological processes. Systemic SARS-CoV-2-associated complications include acute respiratory distress syndrome (ARDS), disseminated intravascular coagulation syndrome, edema, and pneumonia. These are predominantly effects of massive macrophage activation that collectively can be defined as macrophage activation syndrome. In this review we focus on the role of macrophages in COVID-19, as pathogenesis of the new coronavirus infection, especially in cases complicated by ARDS, largely depends on macrophage phenotypes and functionalities. We describe participation of monocytes, monocyte-derived and resident lung macrophages in SARS-CoV-2-associated ARDS and discuss possible utility of cell therapies for its treatment, notably the use of reprogrammed macrophages with stable pro- or anti-inflammatory phenotypes.


Assuntos
COVID-19/patologia , Macrófagos/imunologia , Síndrome do Desconforto Respiratório/patologia , COVID-19/complicações , COVID-19/imunologia , COVID-19/terapia , Terapia Baseada em Transplante de Células e Tecidos , Humanos , Inflamação , Pulmão/imunologia , Pulmão/patologia , Ativação de Macrófagos , Macrófagos/transplante , Macrófagos Alveolares/imunologia , Monócitos/imunologia , Síndrome do Desconforto Respiratório/etiologia , Síndrome do Desconforto Respiratório/imunologia , Síndrome do Desconforto Respiratório/terapia , SARS-CoV-2
20.
Microvasc Res ; 137: 104178, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34015275

RESUMO

OBJECTIVE: To examine the role of YTHDF1 knock-down macrophages on the immunity of severe sepsis rats with ECMO. METHODS: 15 SD rats were randomly allocated into 3 groups: mild sepsis (I), severe sepsis with ECMO (II), and YTHDF1 knock-down macrophages treatment groups (III). Blood biochemical indexes, different immune factors and brain changes were detected by RT-PCR, ELISA, ELISPOT and HE staining. Isolated macrophages subtypes and signal proteins were detected by flow cytometry, western blot and m6A RNA methylation test. RESULTS: The levels of HMGB1, RAGE, YTHDF1 and IL-17 in peripheral blood were significantly higher (p < 0.01), while the level of CXCL9 and TNF-α, and LPS-specific CD8+CTL function were significantly decreased in group II compared with group I (p < 0.01). The ratio of CD63+ macrophages (p < 0.05) and CD64+ macrophages (p< 0.05) decreased and the level of elastase (p < 0.01) and CCR2highCX3CR1low/CCR2lowCX3CR1high (p < 0.01) of macrophages increased in group II. The above were consistent with the severity of biochemical indicators, the increasing endothelial injury factor (Ang2/Ang1), lower endothelial protective factor (sTie2), severer brain injury in group II. After YTHDF1 knock-down macrophages treatment, the above indexes' changes were opposite when Group III versus Group II through the down-regulation of m6A RNA methylation of JAK2/STAT3 (p < 0.01) and protein expression of PJAK2/PSTAT3 (p < 0.05) in isolated macrophages. CONCLUSIONS: YTHDF1 knock-down macrophages improved the immune paralysis of macrophages, Th1/Th17 and CTL and reduced the entry of macrophages into the brain to cause endothelial damage of severe sepsis rats with ECMO through the inhibition of HMGB1/RAGE and YTHDF1, m6A RNA methylation of JAK2/STAT3 and PJAK2/PSTAT3 proteins expression in macrophages.


Assuntos
Lesões Encefálicas/prevenção & controle , Encéfalo/imunologia , Oxigenação por Membrana Extracorpórea , Macrófagos/transplante , Pneumonia/terapia , Proteínas de Ligação a RNA/fisiologia , Sepse/terapia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/imunologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Células Cultivadas , Citocinas/sangue , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Técnicas de Silenciamento de Genes , Mediadores da Inflamação/sangue , Macrófagos/imunologia , Macrófagos/metabolismo , Fenótipo , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/patologia , Proteínas de Ligação a RNA/genética , Ratos Sprague-Dawley , Sepse/imunologia , Sepse/metabolismo , Sepse/patologia , Transdução de Sinais , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th17/imunologia , Células Th17/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...