Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
CPT Pharmacometrics Syst Pharmacol ; 13(6): 994-1005, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38532525

RESUMO

Trastuzumab deruxtecan (T-DXd; DS-8201; ENHERTU®) is a human epithelial growth factor receptor 2 (HER2)-directed antibody drug conjugate (ADC) with demonstrated antitumor activity against a range of tumor types. Aiming to understand the relationship between antigen expression and downstream efficacy outcomes, T-DXd was administered in tumor-bearing mice carrying NCI-N87, Capan-1, JIMT-1, and MDA-MB-468 xenografts, characterized by varying HER2 levels. Plasma pharmacokinetics (PK) of total antibody, T-DXd, and released DXd and tumor concentrations of released DXd were evaluated, in addition to monitoring γΗ2AX and pRAD50 pharmacodynamic (PD) response. A positive relationship was observed between released DXd concentrations in tumor and HER2 expression, with NCI-N87 xenografts characterized by the highest exposures compared to the remaining cell lines. γΗ2AX and pRAD50 demonstrated a sustained increase over several days occurring with a time delay relative to tumoral-released DXd concentrations. In vitro investigations of cell-based DXd disposition facilitated the characterization of DXd kinetics across tumor cells. These outputs were incorporated into a mechanistic mathematical model, utilized to describe PK/PD trends. The model captured plasma PK across dosing arms as well as tumor PK in NCI-N87, Capan-1, and MDA-MB-468 models; tumor concentrations in JIMT-1 xenografts required additional parameter adjustments reflective of complex receptor dynamics. γΗ2AX longitudinal trends were well characterized via a unified PD model implemented across xenografts demonstrating the robustness of measured PD trends. This work supports the application of a mechanistic model as a quantitative tool, reliably projecting tumor payload concentrations upon T-DXd administration, as the first step towards preclinical-to-clinical translation.


Assuntos
Imunoconjugados , Receptor ErbB-2 , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Trastuzumab/farmacocinética , Trastuzumab/farmacologia , Receptor ErbB-2/metabolismo , Camundongos , Humanos , Imunoconjugados/farmacocinética , Imunoconjugados/farmacologia , Linhagem Celular Tumoral , Feminino , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Camptotecina/farmacologia , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/administração & dosagem , Camundongos Nus
2.
Clin Cancer Res ; 30(8): 1567-1581, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-37882675

RESUMO

PURPOSE: Platinum and PARP inhibitors (PARPi) demonstrate activity in breast and ovarian cancers, but drug resistance ultimately emerges. Here, we examine B7-H4 expression in primary and recurrent high-grade serous ovarian carcinoma (HGSOC) and the activity of a B7-H4-directed antibody-drug conjugate (B7-H4-ADC), using a pyrrolobenzodiazepine-dimer payload, in PARPi- and platinum-resistant HGSOC patient-derived xenograft (PDX) models. EXPERIMENTAL DESIGN: B7-H4 expression was quantified by flow cytometry and IHC. B7-H4-ADC efficacy was tested against multiple cell lines in vitro and PDX in vivo. The effect of B7-H4-ADC on cell cycle, DNA damage, and apoptosis was measured using flow cytometry. RESULTS: B7-H4 is overexpressed in 92% of HGSOC tumors at diagnosis (n = 12), persisted in recurrent matched samples after platinum treatment, and was expressed at similar levels across metastatic sites after acquired multi-drug resistance (n = 4). Treatment with B7-H4-ADC resulted in target-specific growth inhibition of multiple ovarian and breast cancer cell lines. In platinum- or PARPi-resistant ovarian cancer cells, B7-H4-ADC significantly decreased viability and colony formation while increasing cell-cycle arrest and DNA damage, ultimately leading to apoptosis. Single-dose B7-H4-ADC led to tumor regression in 65.5% of breast and ovarian PDX models (n = 29), with reduced activity in B7-H4 low or negative models. In PARPi and platinum-resistant HGSOC PDX models, scheduled B7-H4-ADC dosing led to sustained tumor regression and increased survival. CONCLUSIONS: These data support B7-H4 as an attractive ADC target for treatment of drug-resistant HGSOC and provide evidence for activity of an ADC with a DNA-damaging payload in this population. See related commentary by Veneziani et al., p. 1434.


Assuntos
Imunoconjugados , Neoplasias Ovarianas , Feminino , Humanos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Platina/farmacologia , Platina/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Apoptose , Carcinoma Epitelial do Ovário/tratamento farmacológico , Linhagem Celular Tumoral
3.
Immunobiology ; 222(1): 45-54, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26686908

RESUMO

Constituting a part of the innate immune system, the complement system consists of over 50 proteins either acting as part of a 3-branch activation cascade, a well-differentiated regulatory system in fluid phase or on each tissue, or as receptors translating the activation signal to multiple cellular effector functions. Complement serves as first line of defence against infections from bacteria, viruses and parasites by orchestrating the immune response through opsonisation, recruitment of immune cells to the site of infection and direct cell lysis. Complement is generally recognised as a protective mechanism against the formation of tumours in humans, but is often limited by various resistance mechanisms interfering with its cytotoxic action, now considered as a great barrier of successful antibody-based immunotherapy. However, recent studies also indicate a pro-tumourigenic potential of complement in certain cancers and under certain conditions. In this review, we present recent findings on the possible dual role of complement in destroying cancer, especially if resistance mechanisms are blocked, but also under certain inflammatory conditions-promoting tumour development.


Assuntos
Proteínas do Sistema Complemento/imunologia , Neoplasias/imunologia , Neoplasias/patologia , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Citotoxicidade Imunológica , Técnicas de Silenciamento de Genes , Humanos , Imunomodulação , Imunoterapia/métodos , Neoplasias/metabolismo , Neoplasias/terapia , Engenharia de Proteínas
4.
Int J Cancer ; 138(12): 2963-73, 2016 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26830059

RESUMO

A small percentage of healthy donors identified in the Western population carry antibodies in their peripheral blood which convey cytotoxic activity against certain human melanoma and neuroblastoma cell lines. We measured the cytotoxic activity of sera and plasmas from healthy donors on the human neuroblastoma cell line Kelly and various melanoma cell lines. Antibodies of IgM isotype, presumably belonging to the class of naturally occurring antibodies, exerted cytotoxic activity in a complement-dependent fashion. Apart from complement-dependent tumor cell lysis, we observed C3 opsonization in all tumor cell lines upon treatment with cytotoxic plasmas. Cell lines tested primarily expressed membrane complement regulatory proteins (mCRP) CD46, CD55 and CD59 to various extents. Blocking of mCRPs by monoclonal antibodies enhanced cell lysis and opsonization, though some melanoma cells remained resistant to complement attack. Epitopes recognized by cytotoxic antibodies were represented by gangliosides such as GD2 and GD3, as evidenced by cellular sialidase pretreatment and enhanced expression of distinct gangliosides. It remains to be clarified why only a small fraction of healthy persons carry these antitumor cytotoxic antibodies.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Imunoglobulina M/imunologia , Melanoma/imunologia , Neuroblastoma/imunologia , Biomarcadores Tumorais/imunologia , Linhagem Celular Tumoral , Epitopos/imunologia , Gangliosídeos/imunologia , Voluntários Saudáveis , Humanos , Imunoglobulina M/sangue
5.
Oncoimmunology ; 4(3): e979688, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25949896

RESUMO

Complement-dependent cytotoxicity (CDC) is one of the effector mechanisms mediated by therapeutic anticancer monoclonal antibodies (mAbs). However, the efficacy of antibodies is limited by the resistance of malignant cells to complement attack, primarily due to the over-expression of one or more membrane complement regulatory proteins (mCRPs) CD46, CD55, and CD59. CD20-positive Burkitt lymphoma Raji cells and primary CLL cells are resistant to rituximab (RTX)-induced CDC whereas ofatumumab (OFA) proved to be more efficient in cell killing. Primary CLL cells but not CD52-positive acute lymphoblastic leukemia (ALL) REH cells were sensitive to alemtuzumab (ALM)-induced CDC. Upon combined inhibition on Raji and CLL cells by mCRPs-specific siRNAs or neutralizing antibodies, CDC induced by RTX and by OFA was augmented. Similarly, CDC of REH cells was enhanced after mCRPs were inhibited upon treatment with ALM. All mAbs induced C3 opsonization, which was significantly augmented upon blocking mCRPs. C3 opsonization led to enhanced cell-mediated cytotoxicity of leukemia cells exposed to PBLs or macrophages. Furthermore, opsonized CLL cells were efficiently phagocytized by macrophages. Our results provide conclusive evidence that inhibition of mCRPs expression sensitizes leukemic cells to complement attack thereby enhancing the therapeutic effect of mAbs targeting leukemic cells.

6.
Target Oncol ; 10(3): 405-13, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25395366

RESUMO

The overexpression of membrane-bound complement regulatory proteins (mCRP; CD46, CD55, CD59) preventing opsonization and complement-dependent cytotoxicity (CDC) is considered a major barrier for successful antibody-based cancer immunotherapy. To avoid a potential deleterious effect of mCRP neutralization on normal tissue cells, complement regulation has to be selectively targeted to the malignant cells. In this study, anti-mCRP small interfering RNAs (siRNAs) were encapsulated in transferrin-coupled lipoplexes for the specific delivery to transferrin receptor CD71(high) expressing BT474, DU145, and SW480 as well as corresponding CD71-knockdown (CD71(low)) tumor cells. Targeted delivery with transferrin-siRNA-lipoplexes became possible by charge neutralization and resulted in efficient silencing of all three mCRPs up to 90%, which is dependent on their CD71 expression. The mCRP knockdown led to a significant increase of CDC on CD71(high) tumor cells by 68% in BT474, 58% in DU145, and 40% in SW480 cells but only slightly increased on CD71(low) cells. Downregulation of CD46 and CD55 significantly increased C3 opsonization only on CD71(high) tumor cells. Our results demonstrate for the first time that by specific delivery of anti-mCRP siRNA through transferrin receptor, complement regulation can be selectively neutralized, allowing specific antibody-mediated killing of tumor cells without affecting healthy bystander cells, which appears to be a suited strategy to improve antibody-based cancer immunotherapy.


Assuntos
Anticorpos/química , Antígenos CD/metabolismo , Regulação Neoplásica da Expressão Gênica , RNA Interferente Pequeno/metabolismo , Receptores da Transferrina/metabolismo , Antígenos CD/genética , Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Linhagem Celular Tumoral , Complemento C3/metabolismo , Proteínas do Sistema Complemento , Regulação para Baixo , Feminino , Citometria de Fluxo , Humanos , Imunoterapia/métodos , Masculino , Proteína Cofatora de Membrana/metabolismo , RNA Mensageiro/metabolismo , Receptores da Transferrina/genética , Transferrina/metabolismo
7.
Mol Oncol ; 7(3): 580-94, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23474221

RESUMO

The therapeutic potential of anticancer antibodies is limited by the resistance of tumor cells to complement-mediated attack, primarily through the over-expression of membrane complement regulatory proteins (mCRPs: CD46, CD55 and CD59). Trastuzumab, an anti- HER2 monoclonal antibody, approved for the treatment of HER2-positive breast and gastric cancers, exerts only minor complement-mediated cytotoxicity (CDC). Pertuzumab is a novel anti-HER2 monoclonal antibody, which blocks HER2 dimerization with other ligand-activated HER family members. Here, we explored the complement-mediated anti-tumor effects of trastuzumab and pertuzumab on HER2-positive tumor cells of various histological origins. Delivery of chemically stabilized anti-mCRP siRNAs using cationic lipoplexes, AtuPLEXes, to HER2-over-expressing BT474, SK-BR-3 (breast), SKOV3 (ovarian) and Calu-3 (lung) cancer cells reduced mCRPs expression by 85-95%. Knockdown of individual complement regulators variably led to increased CDC only upon combined treatment with trastuzumab and pertuzumab. The combined down-regulation of all the three regulators augmented CDC by 48% in BT474, 46% in SK-BR-3 cells, 78% in SKOV3 cells and by 30% in Calu-3 cells and also increased complement-induced apoptosis and caspase activity on mCRP neutralized tumor cells. In addition, antibody-induced C3 opsonization of tumor cells was significantly enhanced after mCRP silencing and further augmented tumor cell killing by macrophages. Our findings suggest that siRNA-induced inhibition of complement regulator expression clearly enhances complement- and macrophage-mediated anti-tumor activity of trastuzumab and pertuzumab on HER2-positive tumor cells. Thus - if selectively targeted to the tumor - siRNA-induced inhibition of complement regulation may serve as an innovative strategy to potentiate the efficacy of antibody-based immunotherapy.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Antígenos CD55/genética , Antígenos CD59/genética , Proteína Cofatora de Membrana/genética , Neoplasias/tratamento farmacológico , RNA Interferente Pequeno/administração & dosagem , Caspases/imunologia , Linhagem Celular Tumoral , Ativação do Complemento/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Neoplasias/genética , Neoplasias/imunologia , Interferência de RNA , RNA Interferente Pequeno/genética , Receptor ErbB-2/análise , Receptor ErbB-2/antagonistas & inibidores , Trastuzumab
8.
Immunobiology ; 217(6): 593-600, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22204820

RESUMO

All-trans retinoic acid (ATRA) is a potent retinoid, which has been used successfully in different clinical settings as a potential drug to treat COPD and emphysema. In the present study, we analyzed genes modulated by ATRA by performing mRNA expression array analysis on alveolar macrophages after treatment with ATRA. Here we observed a 375-fold up-regulation of Prostaglandin-E Synthase (microsomal PGES-1, NM_004878 PTGES) which mediates the conversion of prostaglandin H(2) (PGH(2)) to Prostaglandin E(2) (PGE(2)). We furthermore studied the expression of PTGES after treatment with ATRA in human monocyte-derived macrophages (MDMs) and bronchoalveolar lavage (BAL) cells. ATRA up-regulated PTGES mRNA expression in MDMs generated with M-CSF by 2500-fold whereas in M-CSF+IL-13 macrophages the up-regulation was only 20-fold. Similarly, ATRA up-regulated PTGES mRNA expression by factor 1524 in BAL cells. The up-regulation of PTGES mRNA expression by ATRA is both time and dose dependent. IL-13 suppressed the ATRA induced PTGES expression at both mRNA and protein level in MDM and BAL cells. We also observed that LPS acts synergistically with ATRA in MDMs and strongly induces PTGES expression. ATRA had little impact on cyclooxygenase-1 and -2 (COX-1 and -2) expression as compared to PTGES expression under the same experimental conditions. Furthermore, we observed an induction of PGE(2) levels by ATRA in BAL cells. These data indicate that ATRA is a potent inducer of PTGES expression in human macrophages but not in alternatively activated macrophages and suggest that the eicosanoid pathway is important for ATRA action in macrophages.


Assuntos
Oxirredutases Intramoleculares/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos/metabolismo , RNA Mensageiro/biossíntese , Sarcoidose Pulmonar/metabolismo , Tretinoína/farmacologia , Lavagem Broncoalveolar , Células Cultivadas , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-13/farmacologia , Oxirredutases Intramoleculares/genética , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Prostaglandina H2/metabolismo , Prostaglandina-E Sintases , Sarcoidose Pulmonar/genética , Sarcoidose Pulmonar/patologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Regulação para Cima
9.
Curr Cancer Drug Targets ; 10(8): 922-31, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20879979

RESUMO

The efficacy of cancer-immunotherapy with complement-activating monoclonal antibodies is limited by over-expression of one or more membrane-bound complement regulatory proteins (mCRPs: CD46, CD55, CD59) on the surface of neoplastic cells. In this study we designed small interfering RNAs (siRNAs) for posttranscriptional gene knock down of CD46, CD55 and CD59 aiming at to sensitize tumor cells to complement attack and thereby to better exploit complement for tumor cell destruction. Tumor cell lines of different origin, such as Du145 (prostate), BT474 (breast) and K562 (erythroleukemia) were selected for the study. FACS-analysis demonstrated that siRNA anti-CD46(301) reduced CD46 protein expression up to 80%, siRNA anti-CD55(255) diminished CD55 protein expression up to 49%, and CD59 protein expression was inhibited up to 82% by siRNA anti-CD59(1339). Time course experiments revealed a long-lasting silencing effect with >50% complement regulator inhibition up to day 13. Upon mCRP knock down, complement-dependent cytotoxicity (CDC) was augmented by 20-30% for CD46, by up to 24% for CD55 and by up to 55% for CD59. The combined inhibition of all three inhibitors further enhanced CDC by up to 66%. Dependent on the cell line, CD46 and CD55 downregulation increased significantly C3 ospsonization, which is known to support cell-mediated defense mechanisms. mCRP blocking antibodies were only partly able to further augment the tumor cells' susceptibility to complement lysis. Thus, siRNA-induced inhibition of complement regulator expression clearly sensitizes malignant cells to complement attack and, if specifically targeted to the tumor, appears suited as adjuvant to improve antibody-based cancer immunotherapy.


Assuntos
Antígenos CD55/metabolismo , Antígenos CD59/metabolismo , Ativação do Complemento/genética , Proteína Cofatora de Membrana/metabolismo , Neoplasias/genética , Neoplasias/imunologia , RNA Interferente Pequeno/genética , Antígenos CD55/química , Antígenos CD55/genética , Antígenos CD59/química , Antígenos CD59/genética , Ativação do Complemento/imunologia , Citometria de Fluxo , Humanos , Proteína Cofatora de Membrana/antagonistas & inibidores , Proteína Cofatora de Membrana/genética , Neoplasias/patologia , RNA Mensageiro/genética , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
10.
J Immunol ; 180(10): 6553-65, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18453574

RESUMO

Alternatively activated (M2) macrophages regulate steady state-, cancer-, and inflammation-related tissue remodeling. They are induced by Th2-cytokines and glucocorticoids (GC). The responsiveness of mature macrophages to TGF-beta, a cytokine involved in inflammation, cancer, and atherosclerosis, is currently controversial. Recently, we demonstrated that IL-17 receptor B is up-regulated in human monocyte-derived macrophages differentiated in the presence of Th2 cytokines IL-4 and TGF-beta1. In this study, we show that mature human macrophages differentiated in the presence of IL-4, and dexamethasone (M2(IL-4/GC)) but not M2(IL-4) responds to TGF-beta1 which induced a gene expression program comprising 111 genes including transcriptional/signaling regulators (ID3 and RGS1), immune modulators (ALOX5AP and IL-17 receptor B) and atherosclerosis-related genes (ALOX5AP, ORL1, APOC1, APOC2, and APOE). Analysis of molecular mechanism underlying GC/TGF-beta cooperation revealed that surface expression of TGF-betaRII was high in M2(GC) and M2(IL-4/GC), but absent from M2(IL-4), whereas the expression of TGF-betaRI/II mRNA, TGF-betaRII total protein, and surface expression of TGF-betaRIII were unchanged. GC dexamethasone was essential for increased surface expression of functional TGF-betaRII because its effect was observed also in combination with IL-13, M-CSF, and GM-CSF. Prolonged Smad2-mediated signaling observed in TGF-beta1-treated M2(IL-4/GC) was due to insufficient activity of negative feedback mechanism what can be explained by up-regulation of SIRT1, a negative regulator of Smad7, and the retention of TGF-betaRII complex on the cell surface. In summary, mature human M2 macrophages made permissive to TGF-beta by GC-induced surface expression of TGF-betaRII activate in response to TGF-beta1, a multistep gene expression program featuring traits of macrophages found within an atherosclerotic lesion.


Assuntos
Expressão Gênica , Glucocorticoides/metabolismo , Ativação de Macrófagos/genética , Macrófagos/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Fator de Crescimento Transformador beta/metabolismo , Diferenciação Celular/imunologia , Membrana Celular/metabolismo , Citometria de Fluxo , Perfilação da Expressão Gênica , Humanos , Interleucina-4/metabolismo , Ativação de Macrófagos/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/análise , Receptores de Interleucina/biossíntese , Receptores de Interleucina-17 , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
J Immunol ; 180(5): 3028-37, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18292525

RESUMO

Alternatively activated (M2) macrophages regulate immune responses and tissue remodelling. In many tissues including placenta, M2 express stabilin-1, a multidomain protein that exerts a dual role as a scavenger receptor for acetylated low density lipoprotein (acLDL) and SPARC (secreted protein acidic and rich in cysteine) and as an intracellular cargo carrier for SI-CLP. Using yeast two-hybrid screening, we identified the developmental hormone placental lactogen (PL) as a novel ligand of stabilin-1. In Chinese hamster ovary-stabilin-1 cells and M2, FACS and confocal microscopy demonstrated that stabilin-1 mediates internalization and endosomal sorting of PL. In M2 macrophages, PL was partially degraded in lysosomes; part of PL escaped degradation and was delivered to novel PL+ storage vesicles lacking endosomal/lysosomal markers. During formation, PL+ vesicles underwent transient interaction with the trans-Golgi network (TGN). Upon placement of PL-loaded M2 into PL-free medium, PL was secreted into the supernatant. Leupeptin, an inhibitor of lysosomal hydrolases, reduced PL degradation, enhanced sorting of PL into the TGN/storage vesicle pathway and increased PL secretion. Thus, processing of PL in M2 macrophages occurs either by the classical lysosomal pathway or by a novel TGN-associated trans-secretory pathway. Macrophages isolated from human placental villi efficiently endocytosed PL-FITC and transported it to the storage vesicles. Our data show that extracellular PL levels are determined by uptake, degradation, storage, and release in M2. During pregnancy PL concentration reaches 10 microg/ml in maternal circulation and stays below 0.5 microg/ml in fetal circulation. We propose that stabilin-1-positive macrophages determine the difference in PL levels between maternal and fetal circulation.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Endocitose/imunologia , Espaço Extracelular/metabolismo , Ativação de Macrófagos , Macrófagos/metabolismo , Lactogênio Placentário/metabolismo , Receptores de Retorno de Linfócitos/fisiologia , Animais , Células CHO , Moléculas de Adesão Celular Neuronais/sangue , Moléculas de Adesão Celular Neuronais/genética , Células Cultivadas , Cricetinae , Cricetulus , Endocitose/genética , Endossomos/imunologia , Endossomos/metabolismo , Espaço Extracelular/imunologia , Feminino , Humanos , Ligantes , Ativação de Macrófagos/genética , Ativação de Macrófagos/imunologia , Macrófagos/classificação , Troca Materno-Fetal/imunologia , Circulação Placentária/imunologia , Lactogênio Placentário/biossíntese , Lactogênio Placentário/sangue , Gravidez , Proteínas da Gravidez/sangue , Proteínas da Gravidez/genética , Proteínas da Gravidez/fisiologia , Receptores de Retorno de Linfócitos/sangue , Receptores de Retorno de Linfócitos/genética , Transfecção
12.
Biomark Insights ; 3: 39-44, 2008 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-19578492

RESUMO

YKL-39 is a Glyco_18 domain containing chitinase-like protein which is currently recognized as a biomarker for the activation of chondrocytes and the progress of the osteoarthritis in human. YKL-39 was identified as an abundantly secreted protein in primary culture of human articular chondrocytes. Two biological activities of YKL-39 might contribute to the disease progression. One is the induction of autoimmune response and second is the participation in tissue remodeling. Other mammalian chitinase-like proteins including chitotriosidase, SI-CLP, YKL-40 and YM1 are expressed by macrophages in various pathological conditions. In contrast, YKL-39 was never reported to be produced by macrophages. We used in vitro model of human monocyte-derived macrophage differentiation to analyse regulation of YKL-39 expression. Expression of YKL-39 was examined by real-time RT-PCR. CD14+ MACS sorted human monocytes differentiated for 6 days under different stimulations including IFNgamma, IL-4, dexamethasone and TGF-beta. We found that both IL-4 and TGF-beta have weak stimulatory effect on YKL-39 expression in all donors tested (3.2 +/- 1.7 fold, p = 0.006 and 6.3 +/- 3.1 fold, p = 0.014 respectively). However the combination of IL-4 and TGF-beta had strong stimulatory effect on the expression of YKL-39 in all analysed individual macrophage cultures (34 +/- 36 fold, p = 0.05). IFN-gamma did not show statistically significant effect of YKL-39 mRNA expression. Presence of dexamethasone almost completely abolished the stimulatory effects of IL-4 and TGF-beta. In summary, we show here for the first time, that human cells of monocyte origin are able to produce YKL-39. Maturation of monocyte derived macrophages in the presence of Th2 cytokine IL-4 and TGF-beta leads to the strong activation of YKL-39 expression. Thus elevated levels of YKL-39 observed during chronic inflammations can not be attributed solely to the activity of chondrocytes. In perspective, YKL-39 might serve as a useful biomarker to detect macrophage-specific response in pathologies like tumour, atherosclerosis and Alzheimer disease.

13.
Immunobiology ; 210(2-4): 161-73, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16164023

RESUMO

Stabilin-1 is a type 1 transmembrane receptor specifically expressed by tissue macrophages and sinusoidal endothelial cells. We recently demonstrated that stabilin-1 is involved in the endocytic/recycling pathway and shuttles between the endosomal system and the trans-Golgi network (TGN) in human macrophages. In the present study, we designed a model cell system to study the mechanisms of stabilin-1-mediated endocytosis. Using CHO-K1 cells stably transfected with stabilin-1, we demonstrated that acetylated low-density lipoprotein (acLDL) induces recruitment of stabilin-1 into the endocytic pathway. Stabilin-1 mediates internalisation of acLDL and its delivery to early endosomes, and it is translocated together with its ligand to the late endosomal compartment. Treatment with wortmannin, a specific inhibitor of phosphatidylinositide 3-kinase (PI3K), did not block stabilin-1 mediated internalisation of acLDL as well as its trafficking to early endosomes, whereas it induced enlargement of stabilin-1/acLDL positive endosomes as well as partial dissociation of EEA1 from these structures. The major effect of wortmannin was the abrogation of stabilin-1/acLDL trafficking into the late endocytic pathway. In stabilin-1 positive human type 2 macrophages, wortmannin treatment resulted in formation of both enlarged and small stabilin-1 positive endosomes. This effect, however, was significantly weaker in macrophages as compared to CHO-stabilin-1 cells. Our data indicate that PI3K activity is required for the transfer of stabilin-1 and its ligand acLDL from early to late endosomal compartments.


Assuntos
Endocitose/fisiologia , Endossomos/metabolismo , Lipoproteínas LDL/metabolismo , Macrófagos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Androstadienos/farmacologia , Animais , Transporte Biológico/fisiologia , Western Blotting , Células CHO , Cricetinae , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Citometria de Fluxo , Humanos , Macrófagos/efeitos dos fármacos , Microscopia Confocal , Inibidores de Proteínas Quinases/farmacologia , Transfecção , Wortmanina , Rede trans-Golgi/metabolismo
14.
J Leukoc Biol ; 76(6): 1151-61, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15345724

RESUMO

Stabilin-1 and stabilin-2 constitute a novel family of fasciclin domain-containing hyaluronan receptor homologues recently described by us. Whereas stabilin-1 is expressed in sinusoidal endothelial cells and in macrophages in vivo, stabilin-2 is absent from the latter. In the present study, we analyzed the subcellular distribution of stabilin-1 in primary human macrophages. Using flow cytometry, expression of stabilin-1 was demonstrated on the surface of interleukin-4/dexamethasone-stimulated macrophages (MPhi2). By immunofluorescence and confocal microscopy, we established that stabilin-1 is preferentially localized in early endosome antigen-1-positive early/sorting endosomes and in recycling endosomes identified by transferrin endocytosis. Association of stabilin-1 was infrequently seen with p62 lck ligand-positive late endosomes and with CD63-positive lysosomes but not in lysosome-associated membrane protein-1-positive lysosomes. Stabilin-1 was also found in the trans-Golgi network (TGN) but not in Golgi stack structures. Glutathione S-transferase pull-down assay revealed that the cytoplasmic tail of stabilin-1 but not stabilin-2 binds to recently discovered Golgi-localized, gamma-ear-containing, adenosine 5'-diphosphate-ribosylation factor-binding (GGA) adaptors GGA1, GGA2, and GGA3 long, mediating traffic between Golgi and endosomal/lysosomal compartments. Stabilin-1 did not bind to GGA3 short, which lacks a part of the Vps27p/Hrs/STAM domain. Deletion of DDSLL and LL amino acid motifs resulted in decreased binding of stabilin-1 with GGAs. A small portion of stabilin-1 colocalized with GGA2 and GGA3 in the TGN in MPhi2. Treatment with brefeldin A resulted in accumulation of stabilin-1 in the TGN. Our results suggest that stabilin-1 is involved in the GGA-mediated sorting processes at the interface of the biosynthetic and endosomal pathways; similarly to other GGA-interacting proteins, stabilin-1 may thus function in endocytic and secretory processes of human macrophages.


Assuntos
Fatores de Ribosilação do ADP/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Endossomos/metabolismo , Macrófagos/metabolismo , Rede trans-Golgi/imunologia , Fatores de Ribosilação do ADP/imunologia , Proteínas Adaptadoras de Transporte Vesicular/imunologia , Motivos de Aminoácidos/imunologia , Antígenos CD/imunologia , Brefeldina A/farmacologia , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular Neuronais/imunologia , Compartimento Celular/efeitos dos fármacos , Compartimento Celular/imunologia , Células Cultivadas , Dexametasona/farmacologia , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Endossomos/efeitos dos fármacos , Endossomos/imunologia , Citometria de Fluxo , Humanos , Interleucina-4/farmacologia , Proteínas de Membrana Lisossomal , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Glicoproteínas de Membrana/imunologia , Glicoproteínas de Membrana/metabolismo , Glicoproteínas da Membrana de Plaquetas/imunologia , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/imunologia , Receptores Imunológicos/imunologia , Receptores de Retorno de Linfócitos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Tetraspanina 30 , Transferrina/metabolismo , Rede trans-Golgi/efeitos dos fármacos , Rede trans-Golgi/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA