Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Sci Immunol ; 7(67): eabf7777, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34995099

RESUMO

Resident macrophages orchestrate homeostatic, inflammatory, and reparative activities. It is appreciated that different tissues instruct specialized macrophage functions. However, individual tissues contain heterogeneous subpopulations, and how these subpopulations are related is unclear. We asked whether common transcriptional and functional elements could reveal an underlying framework across tissues. Using single-cell RNA sequencing and random forest modeling, we observed that four genes could predict three macrophage subsets that were present in murine heart, liver, lung, kidney, and brain. Parabiotic and genetic fate mapping studies revealed that these core markers predicted three unique life cycles across 17 tissues. TLF+ (expressing TIMD4 and/or LYVE1 and/or FOLR2) macrophages were maintained through self-renewal with minimal monocyte input; CCR2+ (TIMD4−LYVE1−FOLR2−) macrophages were almost entirely replaced by monocytes, and MHC-IIhi macrophages (TIMD4−LYVE1−FOLR2−CCR2−), while receiving modest monocyte contribution, were not continually replaced. Rather, monocyte-derived macrophages contributed to the resident macrophage population until they reached a defined upper limit after which they did not outcompete pre-existing resident macrophages. Developmentally, TLF+ macrophages were first to emerge in the yolk sac and early fetal organs. Fate mapping studies in the mouse and human single-cell RNA sequencing indicated that TLF+ macrophages originated from both yolk sac and fetal monocyte precursors. Furthermore, TLF+ macrophages were the most transcriptionally conserved subset across mouse tissues and between mice and humans, despite organ- and species-specific transcriptional differences. Here, we define the existence of three murine macrophage subpopulations based on common life cycle properties and core gene signatures and provide a common starting point to understand tissue macrophage heterogeneity.


Assuntos
Receptor 2 de Folato/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Macrófagos/imunologia , Proteínas de Membrana/imunologia , Receptores CCR2/imunologia , Proteínas de Transporte Vesicular/imunologia , Animais , Estágios do Ciclo de Vida/imunologia , Ativação de Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptores CCR2/deficiência
3.
Front Immunol ; 12: 771054, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745147

RESUMO

The placenta is a fetal-derived organ whose function is crucial for both maternal and fetal health. The human placenta contains a population of fetal macrophages termed Hofbauer cells. These macrophages play diverse roles, aiding in placental development, function and defence. The outer layer of the human placenta is formed by syncytiotrophoblast cells, that fuse to form the syncytium. Adhered to the syncytium at sites of damage, on the maternal side of the placenta, is a population of macrophages termed placenta associated maternal macrophages (PAMM1a). Here we discuss recent developments that have led to renewed insight into our understanding of the ontogeny, phenotype and function of placental macrophages. Finally, we discuss how the application of new technologies within placental research are helping us to further understand these cells.


Assuntos
Desenvolvimento Fetal/imunologia , Feto/imunologia , Imunidade Inata/imunologia , Macrófagos/imunologia , Placenta/imunologia , Animais , Movimento Celular/imunologia , Movimento Celular/fisiologia , Vilosidades Coriônicas/imunologia , Vilosidades Coriônicas/metabolismo , Feminino , Feto/citologia , Feto/fisiologia , Receptor 2 de Folato/imunologia , Receptor 2 de Folato/metabolismo , Antígenos HLA-DR/imunologia , Antígenos HLA-DR/metabolismo , Humanos , Macrófagos/metabolismo , Macrófagos/fisiologia , Fagocitose/imunologia , Fagocitose/fisiologia , Placenta/citologia , Placenta/fisiologia , Gravidez
4.
Cell Rep Med ; 2(10): 100422, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34755134

RESUMO

Provoked by sterile/nonsterile insults, prolonged monocyte mobilization and uncontrolled monocyte/macrophage activation can pose imminent or impending harm to the affected organs. Curiously, folate receptor beta (FRß), with subnanomolar affinity for the vitamin folic acid (FA), is upregulated during immune activation in hematopoietic cells of the myeloid lineage. This phenomenon has inspired a strong interest in exploring FRß-directed diagnostics/therapeutics. Previously, we have reported that FA-targeted aminopterin (AMT) therapy can modulate macrophage function and effectively treat animal models of inflammation. Our current investigation of a lead compound (EC2319) leads to discovery of a highly FR-specific mechanism of action independent of the root causes against inflammatory monocytes. We further show that EC2319 suppresses interleukin-6/interleukin-1ß release by FRß+ monocytes in a triple co-culture leukemic model of cytokine release syndrome with anti-CD19 chimeric antigen receptor T cells. Because of its chemical stability and metabolically activated linker, EC2319 demonstrates favorable pharmacokinetic characteristics and cross-species translatability to support future pre-clinical and clinical development.


Assuntos
Aminopterina/farmacologia , Síndrome da Liberação de Citocina/prevenção & controle , Receptor 2 de Folato/genética , Antagonistas do Ácido Fólico/farmacologia , Ácido Fólico/metabolismo , Macrófagos/efeitos dos fármacos , Animais , Antígenos CD19/genética , Antígenos CD19/imunologia , Células CHO , Cricetulus , Síndrome da Liberação de Citocina/genética , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/patologia , Feminino , Receptor 1 de Folato/antagonistas & inibidores , Receptor 1 de Folato/genética , Receptor 1 de Folato/imunologia , Receptor 2 de Folato/antagonistas & inibidores , Receptor 2 de Folato/imunologia , Humanos , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/patologia , Camundongos , Modelos Biológicos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Monócitos/patologia , Células RAW 264.7 , Ratos , Ratos Endogâmicos Lew , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/patologia
5.
Int J Mol Sci ; 22(11)2021 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-34070369

RESUMO

Folate receptor beta (FRß) is a folate binding receptor expressed on myeloid lineage hematopoietic cells. FRß is commonly expressed at high levels on malignant blasts in patients with acute myeloid leukemia (AML), as well as on M2 polarized tumor-associated macrophages (TAMs) in the tumor microenvironment of many solid tumors. Therefore, FRß is a potential target for both direct and indirect cancer therapy. We demonstrate that FRß is expressed in both AML cell lines and patient-derived AML samples and that a high-affinity monoclonal antibody against FRß (m909) has the ability to cause dose- and expression-dependent ADCC against these cells in vitro. Importantly, we find that administration of m909 has a significant impact on tumor growth in a humanized mouse model of AML. Surprisingly, m909 functions in vivo with and without the infusion of human NK cells as mediators of ADCC, suggesting potential involvement of mouse macrophages as effector cells. We also found that TAMs from primary ovarian ascites samples expressed appreciable levels of FRß and that m909 has the ability to cause ADCC in these samples. These results indicate that the targeting of FRß using m909 has the potential to limit the outgrowth of AML in vitro and in vivo. Additionally, m909 causes cytotoxicity to TAMs in the tumor microenvironment of ovarian cancer warranting further investigation of m909 and its derivatives as therapeutic agents in patients with FRß-expressing cancers.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Receptor 2 de Folato , Imunoterapia , Leucemia Mieloide Aguda , Proteínas de Neoplasias , Neoplasias Ovarianas , Animais , Células CHO , Cricetulus , Feminino , Receptor 2 de Folato/antagonistas & inibidores , Receptor 2 de Folato/imunologia , Células HL-60 , Humanos , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/terapia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Células THP-1 , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Nat Commun ; 12(1): 877, 2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33563975

RESUMO

The immunosuppressive tumor microenvironment (TME) represents a major barrier for effective immunotherapy. Tumor-associated macrophages (TAMs) are highly heterogeneous and plastic cell components of the TME which can either promote tumor progression (M2-like) or boost antitumor immunity (M1-like). Here, we demonstrate that a subset of TAMs that express folate receptor ß (FRß) possess an immunosuppressive M2-like profile. In syngeneic tumor mouse models, chimeric antigen receptor (CAR)-T cell-mediated selective elimination of FRß+ TAMs in the TME results in an enrichment of pro-inflammatory monocytes, an influx of endogenous tumor-specific CD8+ T cells, delayed tumor progression, and prolonged survival. Preconditioning of the TME with FRß-specific CAR-T cells also improves the effectiveness of tumor-directed anti-mesothelin CAR-T cells, while simultaneous co-administration of both CAR products does not. These results highlight the pro-tumor role of FRß+ TAMs in the TME and the therapeutic implications of TAM-depleting agents as preparative adjuncts to conventional immunotherapies that directly target tumor antigens.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias/terapia , Receptores de Antígenos Quiméricos/imunologia , Macrófagos Associados a Tumor/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Receptor 2 de Folato/imunologia , Receptor 2 de Folato/metabolismo , Humanos , Terapia de Imunossupressão , Mesotelina , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Neoplasias/imunologia , Células Tumorais Cultivadas , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/metabolismo
7.
J Exp Med ; 218(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33075123

RESUMO

Hofbauer cells (HBCs) are a population of macrophages found in high abundance within the stroma of the first-trimester human placenta. HBCs are the only fetal immune cell population within the stroma of healthy placenta. However, the functional properties of these cells are poorly described. Aligning with their predicted origin via primitive hematopoiesis, we find that HBCs are transcriptionally similar to yolk sac macrophages. Phenotypically, HBCs can be identified as HLA-DR-FOLR2+ macrophages. We identify a number of factors that HBCs secrete (including OPN and MMP-9) that could affect placental angiogenesis and remodeling. We determine that HBCs have the capacity to play a defensive role, where they are responsive to Toll-like receptor stimulation and are microbicidal. Finally, we also identify a population of placenta-associated maternal macrophages (PAMM1a) that adhere to the placental surface and express factors, such as fibronectin, that may aid in repair.


Assuntos
Macrófagos/imunologia , Placenta/imunologia , Primeiro Trimestre da Gravidez/imunologia , Gravidez/imunologia , Adulto , Feminino , Receptor 2 de Folato/imunologia , Antígenos HLA-DR/imunologia , Humanos , Metaloproteinase 9 da Matriz/imunologia
8.
Front Immunol ; 10: 2724, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31824505

RESUMO

Background: Interstitial lung disease (ILD) is a common and severe complication in rheumatic diseases. Folate receptor-ß is expressed on activated, but not resting macrophages which play a key role in dysregulated tissue repair including ILD. We therefore aimed to pre-clinically evaluate the potential of 18F-AzaFol-based PET/CT (positron emission computed tomography/computed tomography) for the specific detection of macrophage-driven pathophysiologic processes in experimental ILD. Methods: The pulmonary expression of folate receptor-ß was analyzed in patients with different subtypes of ILD as well as in bleomycin (BLM)-treated mice and respective controls using immunohistochemistry. PET/CT was performed at days 3, 7, and 14 after BLM instillation using the 18F-based folate radiotracer 18F-AzaFol. The specific pulmonary accumulation of the radiotracer was assessed by ex vivo PET/CT scans and quantified by ex vivo biodistribution studies. Results: Folate receptor-ß expression was 3- to 4-fold increased in patients with fibrotic ILD, including idiopathic pulmonary fibrosis and connective tissue disease-related ILD, and significantly correlated with the degree of lung remodeling. A similar increase in the expression of folate receptor-ß was observed in experimental lung fibrosis, where it also correlated with disease extent. In the mouse model of BLM-induced ILD, pulmonary accumulation of 18F-AzaFol reflected macrophage-related disease development with good correlation of folate receptor-ß positivity with radiotracer uptake. In the ex vivo imaging and biodistribution studies, the maximum lung accumulation was observed at day 7 with a mean accumulation of 1.01 ± 0.30% injected activity/lung in BLM-treated vs. control animals (0.31 ± 0.06% % injected activity/lung; p < 0.01). Conclusion: Our preclinical proof-of-concept study demonstrated the potential of 18F-AzaFol as a novel imaging tool for the visualization of macrophage-driven fibrotic lung diseases.


Assuntos
Radioisótopos de Flúor , Receptor 2 de Folato/imunologia , Ácido Fólico , Doenças Pulmonares Intersticiais , Macrófagos/imunologia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Compostos Radiofarmacêuticos , Animais , Bleomicina/efeitos adversos , Bleomicina/farmacologia , Feminino , Radioisótopos de Flúor/química , Radioisótopos de Flúor/farmacologia , Ácido Fólico/análogos & derivados , Ácido Fólico/química , Ácido Fólico/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Humanos , Doenças Pulmonares Intersticiais/induzido quimicamente , Doenças Pulmonares Intersticiais/diagnóstico por imagem , Doenças Pulmonares Intersticiais/imunologia , Camundongos , Estudo de Prova de Conceito , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacologia
9.
Arthritis Res Ther ; 21(1): 143, 2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31174578

RESUMO

OBJECTIVES: Most therapies for autoimmune and inflammatory diseases either neutralize or suppress production of inflammatory cytokines produced by activated macrophages (e.g., TNFα, IL-1, IL-6, IL-17, GM-CSF). However, no approved therapies directly target this activated subset of macrophages. METHODS: First, we undertook to examine whether the folate receptor beta (FR-ß) positive subpopulation of macrophages, which marks the inflammatory subset in animal models of rheumatoid arthritis, might constitute the prominent population of macrophages in inflamed lesions in humans. Next, we utilized anti-FR-ß monoclonal antibodies capable of mediating antibody-dependent cell cytotoxicity (ADCC) to treat animal models of rheumatoid arthritis and peritonitis. RESULTS: Human tissue samples of rheumatoid arthritis, Crohn's disease, ulcerative colitis, idiopathic pulmonary fibrosis, nonspecific interstitial pneumonia, chronic obstructive pulmonary disease, systemic lupus erythematosus, psoriasis, and scleroderma are all characterized by dramatic accumulation of macrophages that express FR-ß, a protein not expressed on resting macrophages or any other healthy tissues. A monoclonal antibody to FR-ß accumulates specifically in inflamed lesions of murine inflammatory disease models and successfully treats such models of rheumatoid arthritis and peritonitis. More importantly, elimination of FR-ß-positive macrophages upon treatment with an anti-FR-ß monoclonal antibody promotes the departure of other immune cells, including T cells, B cells, neutrophils, and dendritic cells from the inflamed lesions. CONCLUSIONS: These data suggest that specific elimination of FR-ß-expressing macrophages may constitute a highly specific therapy for multiple autoimmune and inflammatory diseases and that a recently developed human anti-human FR-ß monoclonal antibody (m909) might contribute to suppression of this subpopulation of macrophages.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Artrite Reumatoide/imunologia , Receptor 2 de Folato/imunologia , Imunidade Celular , Ativação de Macrófagos , Macrófagos/metabolismo , Animais , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/patologia , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Macrófagos/imunologia , Macrófagos/patologia , Camundongos
10.
Leukemia ; 30(6): 1355-64, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26898190

RESUMO

Acute myeloid leukemia (AML) is an aggressive malignancy, and development of new treatments to prolong remissions is warranted. Chimeric antigen receptor (CAR) T-cell therapies appear promising but on-target, off-tumor recognition of antigen in healthy tissues remains a concern. Here we isolated a high-affinity (HA) folate receptor beta (FRß)-specific single-chain variable fragment (2.48 nm KD) for optimization of FRß-redirected CAR T-cell therapy for AML. T cells stably expressing the HA-FRß CAR exhibited greatly enhanced antitumor activity against FRß(+) AML in vitro and in vivo compared with a low-affinity FRß CAR (54.3 nm KD). Using the HA-FRß immunoglobulin G, FRß expression was detectable in myeloid-lineage hematopoietic cells; however, expression in CD34(+) hematopoietic stem cells (HSCs) was nearly undetectable. Accordingly, HA-FRß CAR T cells lysed mature CD14(+) monocytes, while HSC colony formation was unaffected. Because of the potential for elimination of mature myeloid lineage, mRNA CAR electroporation for transient CAR expression was evaluated. mRNA-electroporated HA-FRß CAR T cells retained effective antitumor activity in vitro and in vivo. Together, our results highlight the importance of antibody affinity in target protein detection and CAR development and suggest that transient delivery of potent HA-FRß CAR T cells is highly effective against AML and reduces the risk for long-term myeloid toxicity.


Assuntos
Receptor 2 de Folato/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Animais , Linhagem Celular , Linhagem da Célula , Células-Tronco Hematopoéticas , Humanos , Leucemia Mieloide Aguda , Camundongos , Camundongos Transgênicos , Monócitos , Células Mieloides , Anticorpos de Cadeia Única , Linfócitos T/imunologia
11.
Monoclon Antib Immunodiagn Immunother ; 34(3): 181-90, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26090596

RESUMO

Folate receptors α (FRα) and ß (FRß) are two isoforms of the cell surface glycoprotein that binds folate. The expression of FRα is rare in normal cells and elevated in cancer cells. Thus, FRα-based tumor-targeted therapy has been a focus area of laboratory research and clinical trials. Recently, it was shown that a significant fraction of tumor-associated macrophages expresses FRß and that these cells can enhance tumor growth. Although FRα and FRß share 70% identity in their deduced amino acid sequence, a monoclonal antibody (MAb) reactive with both receptors has not been developed. A MAb that can target both FRα-expressing cancer cells and FRß-expressing tumor-associated macrophages may provide a more potent therapeutic tool for cancer than individual anti-FRα or anti-FRß MAbs. In this study, we developed a MAb that recognizes both FRα and FRß (anti-FRαß). The anti-FRαß specifically stained trophoblasts and macrophages from human placenta, synovial macrophages from rheumatoid arthritis patient, liver macrophages from cynomolgus monkey and common marmoset, and cancer cells and tumor-associated macrophages from ovary and lung carcinomas. Surface plasmon resonance showed that the anti-FRαß bound to soluble forms of the FRα and FRß proteins with high affinity (KD=6.26×10(-9) M and 4.33×10(-9) M, respectively). In vitro functional analysis of the anti-FRαß showed that this MAb mediates complement-dependent cytotoxicity, antibody-dependent cellular cytotoxicity, and antibody-dependent cellular phagocytosis of FRα-expressing and FRß-expressing cell lines. The anti-FRαß MAb is a promising therapeutic candidate for cancers in which macrophages promote tumor progression.


Assuntos
Anticorpos Monoclonais/imunologia , Artrite Reumatoide/diagnóstico , Receptor 1 de Folato/imunologia , Receptor 2 de Folato/imunologia , Neoplasias Pulmonares/diagnóstico , Macrófagos/imunologia , Neoplasias Ovarianas/diagnóstico , Trofoblastos/imunologia , Animais , Anticorpos Monoclonais/biossíntese , Formação de Anticorpos , Citotoxicidade Celular Dependente de Anticorpos , Artrite Reumatoide/imunologia , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Neoplasias Pulmonares/imunologia , Macaca fascicularis , Camundongos , Neoplasias Ovarianas/imunologia , Placenta/imunologia , Gravidez , Ratos , Ratos Wistar , Ressonância de Plasmônio de Superfície , Linfócitos T Citotóxicos/imunologia
12.
Mod Rheumatol ; 24(5): 816-22, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24498991

RESUMO

OBJECTIVES: Folate receptor ß (FRß)-expressing macrophages have been identified as activated macrophages. Here, we investigated the infiltration of FRß-expressing macrophages in a murine model of bleomycin (BLM)-induced skin fibrosis and assessed the antifibrotic effects of depletion of FRß-expressing macrophages in this model using a recombinant immunotoxin to FRß. METHODS: A recombinant immunotoxin (anti-FRß-PE38) was prepared by conjugating the Fv portion of the anti-mouse FRß heavy chain with truncated Pseudomonas exotoxin A (VH-PE38) and the Fv portion of the anti-mouse FRß light chain. BLM-induced skin fibrosis mice were intravenously treated with either anti-FRß-PE38 or VH-PE38 as a control protein. Skin fibrosis was evaluated by the change of skin thickness and hydroxyproline content on Day 29. The TGFß1 mRNA levels in the treated skin were assessed by quantitative real-time RT-PCR on Day 9. RESULTS: Numbers of FRß-expressing macrophages increased in BLM-injected skin. Anti-FRß-PE38 treatment led to a dramatic reduction in the number of FRß-expressing macrophages. Additionally, skin thickness and hydroxyproline content, were markedly reduced. TGFß1 mRNA levels were also down-regulated after the treatment. TGFß1 expression was enriched in FRß-expressing macrophages compared with FRß-negative macrophages. CONCLUSION: These results indicated that anti-FRß-PE38 treatment efficiently depleted FRß-expressing macrophages and consequently alleviated BLM-induced skin fibrosis.


Assuntos
Fibrose/terapia , Receptor 2 de Folato/imunologia , Receptor 2 de Folato/metabolismo , Macrófagos/metabolismo , Dermatopatias/terapia , ADP Ribose Transferases , Animais , Toxinas Bacterianas , Bleomicina , Exotoxinas , Fibrose/induzido quimicamente , Fibrose/metabolismo , Fibrose/patologia , Imunotoxinas , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Dermatopatias/induzido quimicamente , Dermatopatias/metabolismo , Dermatopatias/patologia , Fatores de Virulência , Exotoxina A de Pseudomonas aeruginosa
13.
J Leukoc Biol ; 95(5): 797-808, 2014 05.
Artigo em Inglês | MEDLINE | ID: mdl-24399840

RESUMO

Vitamin B9, commonly known as folate, is an essential cofactor for one-carbon metabolism that enters cells through three major specialized transporter molecules (RFC, FR, and PCFT), which differ in expression pattern, affinity for substrate, and ligand-binding pH dependency. We now report that the expression of the folate transporters differs between macrophage subtypes and explains the higher accumulation of 5-MTHF-the major folate form found in serum-in M2 macrophages in vitro and in vivo. M1 macrophages display a higher expression of RFC, whereas FRß and PCFT are preferentially expressed by anti-inflammatory and homeostatic M2 macrophages. These differences are also seen in macrophages from normal tissues involved in folate transit (placenta, liver, colon) and inflamed tissues (ulcerative colitis, RA), as M2-like macrophages from normal tissues express FRß and PCFT, whereas TNF-α-expressing M1 macrophages from inflamed tissues are RFC+. Besides, we provide evidences that activin A is a critical factor controlling the set of folate transporters in macrophages, as it down-regulates FRß, up-regulates RFC expression, and modulates 5-MTHF uptake. All of these experiments support the notion that folate handling is dependent on the stage of macrophage polarization.


Assuntos
Ativinas/imunologia , Polaridade Celular/efeitos dos fármacos , Receptor 2 de Folato/imunologia , Transportadores de Ácido Fólico/imunologia , Ácido Fólico , Macrófagos/imunologia , Polaridade Celular/imunologia , Ácido Fólico/farmacocinética , Ácido Fólico/farmacologia , Humanos
14.
Clin Immunol ; 150(1): 64-77, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24333534

RESUMO

EC0746 is a rationally designed anti-inflammatory drug conjugate consisting of a modified folic acid-based ligand linked to a γ-hydrazide analog of aminopterin. In this report, EC0746's effectiveness was evaluated against experimental retinal S-antigen (PDSAg) induced autoimmune uveitis (EAU) and myelin-basic-protein induced autoimmune encephalomyelitis (EAE). In both models, functional FR-ß was detected on activated macrophages in local (retinal or central-nervous-system, respectively) and systemic (peritoneal cavity) sites of inflammation. In myelin-rich regions of EAE rats, an increased uptake of (99m)Tc-EC20 (etarfolatide; a FR-specific radioimaging agent) was also observed. EC0746 treatment at disease onset suppressed the clinical severity of both EAU and EAE, and it strongly attenuated progressive histopathological changes in the affected organs. In all parameters assessed, EC0746 activity was completely blocked by a benign folate competitor, suggesting that these therapeutic outcomes were specifically FR-ß mediated. EC0746 may emerge as a useful macrophage-modulating agent for treating inflammatory episodes of organ-specific autoimmunity.


Assuntos
Aminopterina/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Antagonistas do Ácido Fólico/uso terapêutico , Uveíte/tratamento farmacológico , Aminopterina/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Receptor 2 de Folato/imunologia , Antagonistas do Ácido Fólico/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Ratos , Ratos Endogâmicos Lew , Medula Espinal/efeitos dos fármacos , Medula Espinal/imunologia , Uveíte/imunologia , Uveíte/patologia
15.
Arthritis Res Ther ; 14(3): R106, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22551402

RESUMO

INTRODUCTION: We previously demonstrated that synovial sublining macrophages express folate receptor beta (FRß). The aim of this study was to evaluate the efficacy of intra-articular administration of a recombinant immunotoxin to FRß for treating rat antigen-induced arthritis. METHODS: A monoclonal antibody (mAb) to rat FRß was produced by immunizing mice with B300-19 cells (murine pre-B cells) transfected with the rat FRß gene. Recombinant immunotoxin was prepared by conjugating the Fv portion of the anti-rat FRß mAb heavy chain with a truncated Pseudomonas exotoxin A and the Fv portion of the anti-rat FRß mAb light chain. Antigen-induced arthritis was induced through intra-articular injection of methylated bovine serum albumin (mBSA) after two subcutaneous injections of mBSA and complete Freund's adjuvant. Immunotoxin was intra-articularly injected into the arthritis joint every other day for seven days after arthritis onset. Joint swelling was measured and histological scores of inflammation, synovial thickness, cartilage, and bone destruction were determined. Immunohistochemistry was performed to detect osteoclast and osteoclast precursor FRß-expressing macrophages and cathepsin K-positive cells on day 21. RESULTS: Intra-articular administration of the immunotoxin attenuated joint swelling (61% suppression; P < 0.01 compared to the control on day 21) and improved histological findings, particularly cartilage and bone destruction (scores of rats treated with control versus the immunotoxin: 2.2 versus 0.5; P < 0.01), by reducing the number of FRß-expressing macrophages and cathepsin K-positive cells. CONCLUSIONS: Intra-articular administration of an immunotoxin to FRß is effective for improving rat antigen-induced arthritis.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Receptor 2 de Folato/antagonistas & inibidores , Receptor 2 de Folato/imunologia , Imunotoxinas/administração & dosagem , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Artrite Reumatoide/patologia , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Imunotoxinas/imunologia , Injeções Intra-Articulares , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Ratos , Ratos Endogâmicos Lew
16.
Autoimmun Rev ; 11(9): 621-6, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22094710

RESUMO

In patients with systemic auto-immune inflammatory rheumatic diseases (AIIRD) like rheumatoid arthritis the prevalence of cardiovascular disease (CVD) is increased. In the pathogenesis of AIIRD and atherosclerosis many similarities can be found in the process underlying CVD. Accumulation of inflammatory cells, in particular macrophages at the site of inflammation producing inflammatory mediators serve as a prominent feature in both systemic inflammation and atherosclerosis. Two different subtypes of macrophages have been described in recent literature namely classically activated macrophages (M1) and alternatively activated macrophages (M2). Alternatively activated macrophages are characterized by low CD14 and high CD163 expression. Macrophages expressing CD14 (M1) have been identified within atherosclerotic plaques, whereas CD14 low macrophages are abundant in vessels without atherosclerosis. Depending on the environment and responses to different stimuli, macrophages in plaques can express diverse pro and anti-atherogenic functions. The balance of these different activation profiles influences atheroma evolution and outcome. Nowadays, influx of macrophages is recognized as a very important feature of the pathogenesis of plaque formation. Activated macrophages accumulate at the sites of inflammation and can therefore be exploited to better visualize inflammatory responses in atherosclerosis. Furthermore, activated (but not resting) macrophages possess a functionally active receptor for folate (FR-ß), but it is not completely clear which subtype of this activated macrophages expresses this receptor and whether the expression of FR-ß is restricted to only one of the macrophage subsets. Although future research needs to be done to investigate FR-ß expression and function within inflamed tissues, the expression of functional FR-ß on tissue macrophages likely occurs during activation. Therefore, expression of FR-ß on activated macrophages holds a promising potential for early diagnosis and better analysis of optimal treatment regiments of vascular diseases in association with systemic diseases.


Assuntos
Artrite Reumatoide/imunologia , Aterosclerose/imunologia , Receptor 2 de Folato/genética , Macrófagos/metabolismo , Artrite Reumatoide/complicações , Artrite Reumatoide/genética , Aterosclerose/diagnóstico , Aterosclerose/etiologia , Aterosclerose/genética , Biomarcadores/metabolismo , Linhagem da Célula/imunologia , Citocinas/imunologia , Receptor 2 de Folato/imunologia , Expressão Gênica , Humanos , Inflamação/complicações , Inflamação/genética , Inflamação/imunologia , Macrófagos/patologia
17.
Arthritis Res Ther ; 13(2): R59, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21477314

RESUMO

INTRODUCTION: Folate receptor beta (FRß) is only detectable in placenta and limited to some hematopoietic cells of myeloid lineage in healthy people. Studies have indicated that FRß is over-expressed in activated macrophages in autoimmune diseases and some cancer cells. In this study we aimed to develop an FRß-specific human monoclonal antibody (mAb) that could be used as a therapeutic agent to treat rheumatoid arthritis and other autoimmune diseases, as well as FRß positive cancers. METHODS: Functional recombinant FRß protein was produced in insect cells and used as antigen to isolate a mAb, m909, from a human naïve Fab phage display library. Binding of Fab and IgG1 m909 to FRß was measured by ELISA, surface plasmon resonance, immune fluorescence staining, and flow cytometry. Antibody-dependent cell-mediated cytotoxicity (ADCC) was evaluated with FRß positive CHO cells as target cells and isolated peripheral blood monocytes as effector cells in an in vitro assay. RESULTS: Fab m909 bound with relatively high affinity (equilibrium dissociation constant 57 nM) to FRß. The IgG1 m909 showed much higher (femtomolar) avidity as measured by ELISA, and it bound to FRß positive cells in a dose-dependent manner, but not to parental FRß negative cells. m909 did not compete with folate for the binding to FRß on cells. m909 was not only able to select FRß positive, activated macrophages from synovial fluid cells of arthritis patients as efficiently as folate, but also able to mediate ADCC in FRß positive cells. CONCLUSIONS: Unlike folate-drug conjugates, m909 selectively binds to FRß, does not recognize FRα, and has at least one effector function. m909 alone has potential to eliminate FRß positive cells. Because m909 does not compete with folate for receptor binding, it can be used with folate-drug conjugates in a combination therapy. m909 can also be a valuable research reagent.


Assuntos
Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Artrite Reumatoide/imunologia , Receptor 2 de Folato/imunologia , Macrófagos/imunologia , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , Humanos , Ativação de Macrófagos/imunologia , Microscopia Confocal , Biblioteca de Peptídeos , Proteínas Recombinantes/imunologia , Ressonância de Plasmônio de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA