Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 19(6): e0304985, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38843278

RESUMO

Signal regulatory protein alpha (SIRPα) is an immune inhibitory receptor on myeloid cells including macrophages and dendritic cells, which binds to CD47, a ubiquitous self-associated molecule. SIRPα-CD47 interaction is exploited by cancer cells to suppress anti-tumor activity of myeloid cells, therefore emerging as a novel immune checkpoint for cancer immunotherapy. In blood cancer, several SIRPα-CD47 blockers have shown encouraging monotherapy activity. However, the anti-tumor activity of SIRPα-CD47 blockers in solid tumors seems limited, suggesting the need for combination therapies to fully exploit the myeloid immune checkpoint in solid tumors. Here we tested whether combination of SIRPα-CD47 blocker with antibody-drug conjugate bearing a topoisomerase I inhibitor DXd (DXd-ADC) would enhance anti-tumor activity in solid tumors. To this end, DS-1103a, a newly developed anti-human SIRPα antibody (Ab), was assessed for the potential combination benefit with datopotamab deruxtecan (Dato-DXd) and trastuzumab deruxtecan (T-DXd), DXd-ADCs targeting human trophoblast cell-surface antigen 2 and human epidermal growth factor receptor 2, respectively. DS-1103a inhibited SIRPα-CD47 interaction and enhanced antibody-dependent cellular phagocytosis of Dato-DXd and T-DXd against human cancer cells. In a whole cancer cell vaccination model, vaccination with DXd-treated cancer cells led to activation of tumor-specific T cells when combined with an anti-mouse SIRPα (anti-mSIRPα) Ab, implying the benefit of combining DXd-ADCs with anti-SIRPα Ab on anti-tumor immunity. Furthermore, in syngeneic mouse models, both Dato-DXd and T-DXd combination with anti-mSIRPα Ab showed stronger anti-tumor activity over the monotherapies. Taken together, this study provides a preclinical rationale of novel therapies for solid tumors combining SIRPα-CD47 blockers with DXd-ADCs.


Assuntos
Antígenos de Diferenciação , Antígeno CD47 , Imunoconjugados , Receptores Imunológicos , Antígeno CD47/antagonistas & inibidores , Antígeno CD47/imunologia , Animais , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/metabolismo , Receptores Imunológicos/imunologia , Humanos , Camundongos , Imunoconjugados/farmacologia , Antígenos de Diferenciação/imunologia , Linhagem Celular Tumoral , Feminino , Trastuzumab/farmacologia , Inibidores da Topoisomerase I/farmacologia , Imunoterapia/métodos , Camundongos Endogâmicos BALB C
2.
Sci Rep ; 14(1): 5135, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429446

RESUMO

G-protein-coupled receptor class 5 member D (GPRC5D) is detected in malignant plasma cells in approximately 90% of patients diagnosed with multiple myeloma (MM). Here, we constructed BsAb5003, a novel humanized bispecific monoclonal antibody targeting CD3 and GPRC5D, and evaluated its therapeutic impact on MM. BsAb5003 induced specific cytotoxicity of GPRC5D-positive MM cells with concomitant T cell activation and cytokine release. The efficacy of BsAb5003 was associated with GPRC5D expression levels in MM cell lines. Flow cytometry analysis of bone marrow mononuclear cells (BMMNCs) from 49 MM patients revealed that GPRC5D was expressed in a wide population of MM patients, including heavily treated and high-risk patients. In ex vivo assays using BMMNCs, BsAb5003 induced potent efficacy against CD138 + MM cells in both newly diagnosed and relapsed/refractory patient samples in a GPRC5D expression-dependent manner. BsAb5003 significantly enhanced T cell activation and cytokine production in combination with immunomodulatory drugs (IMiDs) against MM cell lines. BsAb5003 also demonstrated significant inhibition of in vivo tumor growth by recruiting T cells. Taken together, these results suggest that T cell-redirecting bispecific antibody targeting GPRC5D as monotherapy and combination therapy with IMiDs could be a highly potent and effective treatment approach for a wide population of MM patients.


Assuntos
Anticorpos Biespecíficos , Mieloma Múltiplo , Humanos , Anticorpos Biespecíficos/uso terapêutico , Citocinas/metabolismo , Agentes de Imunomodulação , Mieloma Múltiplo/imunologia , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Receptores Acoplados a Proteínas G , Linfócitos T
3.
J Oncol ; 2022: 3552793, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36385956

RESUMO

CD147 is an immunoglobulin-like receptor that is highly expressed in various cancers and involved in the growth, metastasis, and activation of inflammatory pathways via interactions with various functional molecules, such as integrins, CD44, and monocarboxylate transporters. Through screening of CD147-targeting antibodies with antitumor efficacy, we discovered a novel rat monoclonal antibody #147D. This humanized IgG4-formatted antibody, h4#147D, showed potent antitumor efficacy in xenograft mouse models harboring the human PDAC cell line MIA PaCa-2, HCC cell line Hep G2, and CML cell line KU812, which featured low sensitivity to the corresponding standard-of-care drugs (gemcitabine, sorafenib, and imatinib, respectively). An analysis of tumor cells derived from MIA PaCa-2 xenograft mice treated with h4#147D revealed that cell surface expression of CD147 and its binding partners, including CD44 and integrin α3ß1/α6ß1, was significantly reduced by h4#147D. Inhibition of focal adhesion kinase (FAK), activation of multiple stress responsible signal proteins such as c-JunN-terminal kinase (JNK) and mitogen-activated protein kinase p38 (p38MAPK), and expression of SMAD4, as well as activation of caspase-3 were obviously observed in the tumor cells, suggesting that h4#147D induced tumor shrinkage by inducing multiple stress responsible signals. These results suggest that the anti-CD147 antibody h4#147D offers promise as a new antibody drug candidate.

4.
Mol Cancer Ther ; 18(10): 1832-1843, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31350344

RESUMO

The FGFR4/FGF19 signaling axis is overactivated in 20% of liver tumors and currently represents a promising targetable signaling mechanism in this cancer type. However, blocking FGFR4 or FGF19 has proven challenging due to its physiological role in suppressing bile acid synthesis which leads to increased toxic bile acid plasma levels upon FGFR4 inhibition. An FGFR4-targeting antibody, U3-1784, was generated in order to investigate its suitability as a cancer treatment without major side effects.U3-1784 is a high-affinity fully human antibody that was obtained by phage display technology and specifically binds to FGFR4. The antibody inhibits cell signaling by competing with various FGFs for their FGFR4 binding site thereby inhibiting receptor activation and downstream signaling via FRS2 and Erk. The inhibitory effect on tumor growth was investigated in 10 different liver cancer models in vivo The antibody specifically slowed tumor growth of models overexpressing FGF19 by up to 90% whereas tumor growth of models not expressing FGF19 was unaffected. In cynomolgus monkeys, intravenous injection of U3-1784 caused elevated serum bile acid and liver enzyme levels indicating potential liver damage. These effects could be completely prevented by the concomitant oral treatment with the bile acid sequestrant colestyramine, which binds and eliminates bile acids in the gut. These results offer a new biomarker-driven treatment modality in liver cancer without toxicity and they suggest a general strategy for avoiding adverse events with FGFR4 inhibitors.


Assuntos
Anticorpos Monoclonais/toxicidade , Anticorpos Monoclonais/uso terapêutico , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/imunologia , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Resina de Colestiramina/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Íleo/efeitos dos fármacos , Íleo/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Células NIH 3T3 , Transdução de Sinais/efeitos dos fármacos , Sorafenibe/farmacologia
5.
Theranostics ; 6(12): 2225-2234, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27924159

RESUMO

Background: DS-8273a, an anti-human death receptor 5 (DR5) agonistic antibody, has cytotoxic activity against human cancer cells and induces apoptosis after specific binding to DR5. DS-8273a is currently being used in clinical Phase I trials. This study evaluated the molecular imaging of DR5 expression in vivo in mouse tumor models using SPECT/CT and PET/MRI, as a tool for drug development and trial design. Methods: DS-8273a was radiolabeled with indium-111 and zirconium-89. Radiochemical purity, immunoreactivity, antigen binding affinity and serum stability were assessed in vitro. In vivo biodistribution and pharmacokinetic studies were performed, including SPECT/CT and PET/MR imaging. A dose-escalation study using a PET/MR imaging quantitative analysis was also performed to determine DR5 receptor saturability in a mouse model. Results:111In-CHX-A″-DTPA-DS-8273a and 89Zr-Df-Bz-NCS-DS-8273a showed high immunoreactivity (100%), high serum stability, and bound to DR5 expressing cells with high affinity (Ka, 1.02-1.22 × 1010 M-1). The number of antibodies bound per cell was 32,000. In vivo biodistribution studies showed high and specific uptake of 111In-CHX-A″-DTPA-DS-8273a and 89Zr-Df-Bz-NCS-DS-8273a in DR5 expressing COLO205 xenografts, with no specific uptake in normal tissues or in DR5-negative CT26 xenografts. DR5 receptor saturation was observed in vivo by biodistribution studies and quantitative PET/MRI analysis. Conclusion:89Zr-Df-Bz-NCS-DS-8273a is a potential novel PET imaging reagent for human bioimaging trials, and can be used for effective dose assessment and patient response evaluation in clinical trials.


Assuntos
Adenocarcinoma/diagnóstico por imagem , Adenocarcinoma/terapia , Anticorpos/administração & dosagem , Radioisótopos/administração & dosagem , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/antagonistas & inibidores , Nanomedicina Teranóstica/métodos , Zircônio/administração & dosagem , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Xenoenxertos , Humanos , Índio/administração & dosagem , Índio/farmacocinética , Imageamento por Ressonância Magnética , Camundongos Endogâmicos BALB C , Tomografia por Emissão de Pósitrons , Radioisótopos/farmacocinética , Radioterapia/métodos , Tomografia Computadorizada de Emissão de Fóton Único , Resultado do Tratamento , Zircônio/farmacocinética
6.
Faraday Discuss ; 180: 479-93, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25893229

RESUMO

Conducting polymers (CPs), including polypyrrole, have attracted attention for their potential in the protection of metals against corrosion; however, CP coatings have the limitation of poor adhesion to metal substrates. In this study, a composite coating, comprising a self-organized porous anodic oxide layer and a polypyrrole layer, has been developed on iron. Because of electropolymerization in the pores of the anodic oxide layer, the composite coating showed improved adhesion to the substrate along with prolonged corrosion protection in a NaCl aqueous corrosive environment. The anodic oxide layers are formed in a fluoride-containing organic electrolyte and contain a large amount of fluoride species. The removal of these fluoride species from the oxide layer and the metal/oxide interface region is crucial for improving the corrosion protection.

7.
Colloids Surf B Biointerfaces ; 64(1): 16-21, 2008 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-18282692

RESUMO

Dialkyl disulfide-linked naphthoquinone, (NQ-Cn-S)2, and anthraquinone, (AQ-Cn-S)2, derivatives with different spacer alkyl chains (Cn: n=2, 6, 12) were synthesized and these quinone derivatives were self-assembled on a gold electrode. The formation of self-assembled monolayers (SAMs) of these derivatives on a gold electrode was confirmed by infrared reflection-absorption spectroscopy (IR-RAS). Electron transfer between the derivatives and the gold electrode was studied by cyclic voltammetry. On the cyclic voltammogram a reversible redox reaction between quinone (Q) and hydroquinone (QH2) was clearly observed under an aqueous condition. The formal potentials for NQ and AQ derivatives were -0.48 and -0.58 V, respectively, that did not depend on the spacer length. The oxidation and reduction peak currents were strongly dependent on the spacer alkyl chain length. The redox behavior of quinone derivatives depended on the pH condition of the buffer solution. The pH dependence was in agreement with a theoretical value of E 1/2 (mV)=E'-59pH for 2H+/2e(-) process in the pH range 3-11. In the range higher than pH 11, the value was estimated with E 1/2 (mV)=E'-30pH , which may correspond to H+/2e(-) process. The tunneling barrier coefficients (beta) for NQ and AQ SAMs were determined to be 0.12 and 0.73 per methylene group (CH2), respectively. Comparison of the structures and the alkyl chain length of quinones derivatives on these electron transfers on the electrode is made.


Assuntos
Antraquinonas/química , Dissulfetos/química , Ouro , Naftoquinonas/química , Antraquinonas/síntese química , Benzoquinonas/química , Dissulfetos/síntese química , Eletrodos , Transporte de Elétrons , Concentração de Íons de Hidrogênio , Naftoquinonas/síntese química , Prótons
8.
Colloids Surf B Biointerfaces ; 61(1): 106-12, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-17681456

RESUMO

Phospholipid-linked naphthoquinones separated by spacer methylene groups (C(n)), PE-C(n)-NQ (n=0, 5, 11), were synthesized to investigate the quinone-mediated electron transfers on a glassy carbon (GC) electrode covered with phospholipids membrane. The PE-C(n)-NQ could be incorporated in lipid bilayer composed of phosphatidylcholine and exhibited characteristic absorption spectral change corresponding to their redox state, quinone/hydroquinone. The cyclic voltammogram of PE-C(n)-NQ-containing lipid bilayer modified on a GC electrode indicated a set of waves corresponding to the consecutive two-electron and two-proton transfer reduction of the quinone moiety. The peak currents of PE-C(n)-NQ as a function of temperature showed a sharp break point in the current-temperature behavior, reflecting the gel-fluid phase transition. The shape of the cyclic voltammograms changed with the pH of the buffer solution. Below pH 6 the first step of the reduction of quinone was a monoprotonation of quinone, whereas above pH 10 the first step of the oxidation was a monodeprotonation of hydroquinone. This indicates that reaction sequences of quinone/hydroquinone were different with the change of the pH. These results showed that the PE-C(n)-NQ exhibited electron transfer associated with proton transfer in the lipid membranes, depending on the diffusivity of the redox species in the membrane and pH. Interestingly, less effect of the number of methylene of the spacer group on the peak currents was observed. Comparison of manganese porphyrin-mediated electron transfer that depends on the spacer methylene lengths [M. Nango, T. Hikita, T. Nakano, T. Yamada, M. Nagata, Y. Kurono, T. Ohtsuka, Langmuir 14 (1998) 407] is made.


Assuntos
Transporte de Elétrons , Bicamadas Lipídicas/química , Naftoquinonas/química , Eletrodos , Concentração de Íons de Hidrogênio , Lipídeos de Membrana , Naftoquinonas/síntese química
9.
J Clin Invest ; 117(10): 2812-24, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17885681

RESUMO

Marked sarcomere disorganization is a well-documented characteristic of cardiomyocytes in the failing human myocardium. Myosin regulatory light chain 2, ventricular/cardiac muscle isoform (MLC2v), which is involved in the development of human cardiomyopathy, is an important structural protein that affects physiologic cardiac sarcomere formation and heart development. Integrated cDNA expression analysis of failing human myocardia uncovered a novel protein kinase, cardiac-specific myosin light chain kinase (cardiac-MLCK), which acts on MLC2v. Expression levels of cardiac-MLCK were well correlated with the pulmonary arterial pressure of patients with heart failure. In cultured cardiomyocytes, knockdown of cardiac-MLCK by specific siRNAs decreased MLC2v phosphorylation and impaired epinephrine-induced activation of sarcomere reassembly. To further clarify the physiologic roles of cardiac-MLCK in vivo, we cloned the zebrafish ortholog z-cardiac-MLCK. Knockdown of z-cardiac-MLCK expression using morpholino antisense oligonucleotides resulted in dilated cardiac ventricles and immature sarcomere structures. These results suggest a significant role for cardiac-MLCK in cardiogenesis.


Assuntos
Baixo Débito Cardíaco/enzimologia , Coração/embriologia , Miocárdio/ultraestrutura , Quinase de Cadeia Leve de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/fisiologia , Organogênese , Sarcômeros/metabolismo , Proteínas de Peixe-Zebra/fisiologia , Adulto , Idoso , Sequência de Aminoácidos , Animais , Miosinas Cardíacas/metabolismo , Baixo Débito Cardíaco/genética , Baixo Débito Cardíaco/patologia , Células Cultivadas , Clonagem Molecular , Embrião não Mamífero/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Miocárdio/enzimologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/genética , Análise de Sequência com Séries de Oligonucleotídeos , Oligorribonucleotídeos Antissenso/farmacologia , Organogênese/genética , Ratos , Regulação para Cima , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/antagonistas & inibidores , Proteínas de Peixe-Zebra/genética
10.
Circ J ; 68(8): 747-50, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15277733

RESUMO

BACKGROUND: Elevated concentrations of cardiac troponin T and heart-type fatty acid-binding protein (H-FABP) identify patients with chronic heart failure (CHF) and ongoing myocardial damage (OMD) who are at increased risk for future cardiac events. Cardiomyocyte necrosis and/or apoptosis via activated tumor necrosis factor (TNF) and the Fas/Fas ligand (FasL) system may be related to the development of OMD. METHODS AND RESULTS: The serum concentrations of H-FABP, a sensitive marker of membrane damage of cardiomyocytes, soluble Fas (sFas) and TNF-alpha were measured in 38 patients with CHF. The concentrations of H-FABP, TNF-alpha and s-Fas in patients with New York Heart Association (NYHA) III + IV were all significantly higher than in those patients in NYHA II (H-FABP; III + IV 9.3+/-5.9 vs II 5.1+/-1.8 ng/ml, p=0.003, TNF-alpha; III + IV 10.5+/-3.8 vs II 8.0+/-2.7 pg/ml, p=0.02, sFas; III + IV 3.36+/-1.37 vs II 2.58 +/-0.84 ng/ml, p=0.03). Increased concentrations of H-FABP significantly correlated with the concentrations of TNF- alpha (r=0.57, p=0.0001) and sFas (r=0.69, p<0.0001), independent of renal function. CONCLUSION: OMD detected by H-FABP, a marker of membrane damage, is related to activated TNF and the Fas/FasL system, which suggests a pathophysiological role of cardiomyocyte necrosis and/or apoptosis in patients with worsening heart failure.


Assuntos
Insuficiência Cardíaca/patologia , Glicoproteínas de Membrana/sangue , Miocárdio/patologia , Fator de Necrose Tumoral alfa/análise , Receptor fas/sangue , Idoso , Biomarcadores/sangue , Proteínas de Transporte/sangue , Proteína Ligante Fas , Proteínas de Ligação a Ácido Graxo , Feminino , Insuficiência Cardíaca/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Análise de Regressão
11.
J Biol Chem ; 279(36): 37832-41, 2004 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-15231837

RESUMO

The 2-5A system is one of the major pathways for antiviral and antitumor functions that can be induced by interferons (IFNs). The 2-5A system is modulated by 5'-triphosphorylated, 2',5'-phosphodiester-linked oligoadenylates (2-5A), which are synthesized by 2',5'-oligoadenylate synthetases (2',5'-OASs), inactivated by 5'-phosphatase and completely degraded by 2'-phosphodiesterase (2'-PDE). Generated 2-5A activates 2-5A-dependent endoribonuclease, RNase L, which induces RNA degradation in cells and finally apoptosis. Although 2',5'-OASs and RNase L have been molecularly cloned and studied well, the identification of 2'-PDE has remained elusive. Here, we describe the first identification of 2'-PDE, the third key enzyme of the 2-5A system. We found a putative 2'-PDE band on SDS-PAGE by successive six-step chromatographies from ammonium sulfate precipitates of bovine liver and identified a partial amino acid sequence of the human 2'-PDE by mass spectrometry. Based on the full-length sequence of the human 2'-PDE obtained by in silico expressed sequence tag assembly, the gene was cloned by reverse transcription-PCR. The recombinant human 2'-PDE expressed in mammalian cells certainly cleaved the 2',5'-phosphodiester bond of 2-5A trimer and 2-5A analogs. Because no sequences with high homology to this human 2'-PDE were found, the human 2'-PDE was considered to be a unique enzyme without isoform. Suppression of 2'-PDE by a small interfering RNA and a 2'-PDE inhibitor resulted in significant reduction of viral replication, whereas overexpression of 2'-PDE protected cells from IFN-induced antiproliferative activity. These observations identify 2'-PDE as a key regulator of the 2-5A system and as a potential novel target for antiviral and antitumor treatments.


Assuntos
Nucleotídeos de Adenina/metabolismo , Exorribonucleases/metabolismo , Oligorribonucleotídeos/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Bovinos , Primers do DNA , Exorribonucleases/antagonistas & inibidores , Exorribonucleases/química , Células HeLa , Humanos , Fígado/enzimologia , Dados de Sequência Molecular , Inibidores de Fosfodiesterase/farmacologia , Homologia de Sequência de Aminoácidos , Replicação Viral/efeitos dos fármacos
12.
J Immunol ; 171(2): 1061-9, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12847280

RESUMO

TRAIL has been proposed as an anti-inflammatory cytokine in animal models of rheumatoid arthritis (RA). Using two agonistic mAbs specific for TRAIL-R1 (DR4) and TRAIL-R2 (DR5), we examined the expression and function of these death receptors in RA synovial fibroblast cells. The synovial tissues and primary synovial fibroblast cells isolated from patients with RA, but not those isolated from patients with osteoarthritis, selectively expressed high levels of cell surface DR5 and were highly susceptible to anti-DR5 Ab (TRA-8)-mediated apoptosis. In contrast, RA synoviocytes did not show increased expression of TRAIL-R1 (DR4), nor was there any difference in expression of Fas between RA and osteoarthritis synovial cells. In vitro TRA-8 induced apoptosis of RA synovial cells and inhibited production of matrix metalloproteinases induced by pro-inflammatory cytokines. In vivo TRA-8 effectively inhibited hypercellularity of a SV40-transformed RA synovial cell line and completely prevented bone erosion and cartilage destruction induced by these cells. These results indicate that increased DR5 expression and susceptibility to DR5-mediated apoptosis are characteristic of the proliferating synovial cells in RA. As highly proliferative transformed-appearing RA synovial cells play a crucial role in bone erosion and cartilage destruction in RA, the specific targeting of DR5 on RA synovial cells with an agonistic anti-DR5 Ab may be a potential therapy for RA.


Assuntos
Apoptose/imunologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Fibroblastos/imunologia , Fibroblastos/patologia , Receptores do Fator de Necrose Tumoral/fisiologia , Membrana Sinovial/imunologia , Membrana Sinovial/patologia , Adulto , Idoso , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Artrite Reumatoide/prevenção & controle , Linhagem Celular , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibroblastos/transplante , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/biossíntese , Receptores do Fator de Necrose Tumoral/imunologia , Membrana Sinovial/metabolismo , Membrana Sinovial/transplante , Ativação Transcricional/imunologia , Transplante Heterólogo/imunologia , Transplante Heterólogo/patologia , Regulação para Cima/imunologia
13.
Oncogene ; 22(13): 2034-44, 2003 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-12673208

RESUMO

Using two agonistic monoclonal antibodies specific for each death receptor of TRAIL, 2E12 (anti-human DR4) and TRA-8 (anti-human DR5), we examined the signal transduction of the death receptors in combination with or without chemotherapy agents such as Adriamycin (doxorubicin hydrochloride) and Cisplatin. Our results demonstrated that chemotherapy agents were able to enhance apoptosis-inducing activity of these antibodies against several different types of tumor cell lines through enhanced caspase activation. The combination of the antibodies and chemotherapy agents led to a synergistical activation of the JNK/p38 MAP kinase, which was mediated by MKK4. The combination also caused an increased release of cytochrome c and Smac/DIABLO from mitochondria in parallel with the profound loss of mitochondrial membrane potential. These results suggest that the enhanced activation of the JNK/p38 kinase and the mitochondrial apoptosis pathways play a crucial role in synergistic induction of the death receptor-mediated apoptosis by chemotherapy agents. Thus, the simultaneous targeting of cell surface death receptors with agonistic antibodies and the intracellular JNK/p38 and the mitochondrial death pathways with chemotherapy agents would enhance the efficacy and selectivity of both agents in cancer therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Doxorrubicina/farmacologia , MAP Quinase Quinase 4 , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Paclitaxel/farmacologia , Receptores do Fator de Necrose Tumoral/agonistas , Acetilcisteína/farmacologia , Adenocarcinoma/patologia , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Apoptose/fisiologia , Proteínas Reguladoras de Apoptose , Neoplasias da Mama/patologia , Proteínas de Transporte/análise , Caspases/fisiologia , Grupo dos Citocromos c/análise , Fragmentação do DNA/efeitos dos fármacos , Sinergismo Farmacológico , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Membranas Intracelulares/efeitos dos fármacos , Membranas Intracelulares/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Quinases JNK Ativadas por Mitógeno , Potenciais da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/fisiologia , Proteínas Mitocondriais/análise , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/fisiologia , Células Tumorais Cultivadas/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno
14.
J Exp Med ; 197(8): 1029-35, 2003 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-12707301

RESUMO

Osteoclasts are multinucleated cells that resorb bones, and are derived from hematopoietic cells of the monocyte/macrophage lineage. The receptor activator of NF-kappaB ligand (RANKL, also called ODF/TRANCE/OPGL) stimulates both osteoclast differentiation from osteoclast progenitors and activation of mature osteoclasts. To identify genes responsible for osteoclast differentiation, we used a molecular indexing technique. Here, we report a clone of one of these genes whose transcription is induced by soluble RANKL (sRANKL) in both the RAW264.7 cells of the mouse macrophage cell line and the mouse primary bone marrow cells. The predicted protein was found to be a mouse homologue of Jun dimerization protein 2 (JDP2), a member of the AP-1 family of transcription factors, containing a basic region-leucine zipper motif. Transient transfection experiments revealed that overexpression of JDP2 leads to activation of both tartrate-resistant acid phosphatase (TRAP) and cathepsin K gene promoters in RAW264.7 cells. Infection of mouse primary bone marrow cells with retroviruses expressing JDP2-facilitated sRANKL-mediated formation of TRAP-positive multinuclear osteoclasts. Importantly, antisense oligonucleotide to JDP2 strongly suppressed sRANKL-induced osteoclast formation of RAW264.7 cells. Our findings suggest that JDP2 may play an important role in the RANK-mediated signal transduction system, especially in osteoclast differentiation.


Assuntos
Proteínas de Transporte/metabolismo , Diferenciação Celular/fisiologia , Glicoproteínas de Membrana/metabolismo , Osteoclastos/fisiologia , Proteínas Repressoras/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Células Cultivadas , Sistema de Sinalização das MAP Quinases/fisiologia , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Osteoclastos/citologia , Regiões Promotoras Genéticas , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B
15.
Antisense Nucleic Acid Drug Dev ; 12(5): 301-9, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12477280

RESUMO

The highly specific posttranscriptional silencing of gene expression induced by double-stranded RNA (dsRNA) is known as RNA interference (RNAi) and has been demonstrated in plants, nematodes, Drosophila, and protozoa, as well as in mammalian cells. The suppression of expression of specific genes by chemically synthesized 21-nucleotide (21-nt) RNA duplexes has been achieved in various lines of mammalian cells, and this technique might prove to be a valuable tool in efforts to analyze biologic functions of genes in mammalian cells. In order to investigate the utility of potential modifications that can be introduced into small interfering RNAs (siRNAs) and also to study their functional anatomy, we synthesized different types of siRNA targeted to mRNA of Jun dimerization protein 2 (JDP2). Our detailed analysis demonstrated that siRNAs with only one mismatch, relative to the target, on the antisense strand had reduced RNAi effect, whereas the corresponding mutation on the sense strand did not interfere with the RNAi. Moreover, one 2-hydroxyethylphosphate (hp) substitution at the 3'-end of the antisense strand but not of the sense strand also prevented RNAi, whereas a related modification at the 3'-end of either strand, using 2'-O,4'-C-ethylene thymidine (eT), which is a component of ethylene-bridge nucleic acids (ENA), completely abolished RNAi. These results support the hypothesis that the two strands have different functions in RNAi in cultured mammalian cells and indicate that their chemical modification of siRNAs at the 3'-end of the sense strand exclusively is possible, without loss of RNAi activity, depending on the type of modification. Because modification at the 3'-end of the antisense strand by hp or eT abolished the RNAi effect, it appears possible that the 3'-end is recognized by the RNA-induced silencing complex (RISC).


Assuntos
Pareamento Incorreto de Bases , Expressão Gênica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Animais , Sequência de Bases , Western Blotting , Linhagem Celular , Primers do DNA , Inativação Gênica , Camundongos , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , Proteínas Repressoras/genética
16.
J Biol Chem ; 277(32): 29294-303, 2002 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-12034736

RESUMO

Bisindolylmaleimide VIII (Bis VIII) has been previously shown to enhance Fas-mediated apoptosis through a protein kinase C-independent mechanism. In the present study, we examined the effect of Bis VIII on apoptosis induced by DR5 (TRAIL-R2), using an agonistic anti-human DR5 monoclonal antibody, TRA-8. Our results demonstrated that Bis VIII was able to enhance the apoptosis-inducing activity of TRA-8 both in vitro and in vivo. The combination of TRA-8 and Bis VIII led to a synergistic and sustained activation of the c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase, which was mediated by MAPK kinase 4 and was caspase-8-dependent. The mitochondrial pathway is involved in the synergistic induction of apoptosis by Bis VIII and TRA-8. Bis VIII alone induced the loss of mitochondrial membrane potential in a caspase-independent fashion without subsequent release of cytochrome c. However, in the presence of Bis VIII, TRA-8 induced more profound loss of mitochondrial membrane potential and release of cytochrome c. These results suggest that the enhanced and persistent activation of the JNK/p38 and the decreased mitochondrial membrane potential play a crucial role in synergistic induction of the death receptor-mediated apoptosis by Bis VIII. The unique ability of Bis VIII to enhance DR5-mediated apoptosis signal transduction discloses a potential utility of this compound in combination with anti-DR5 antibody in cancer therapy.


Assuntos
Apoptose , Indóis/farmacologia , MAP Quinase Quinase 4 , Maleimidas/farmacologia , Mitocôndrias/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais , Caspase 8 , Caspase 9 , Caspases/metabolismo , Grupo dos Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Células Jurkat , Microscopia de Fluorescência , Proteína Quinase 8 Ativada por Mitógeno , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Fatores de Tempo , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...