Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
1.
Front Immunol ; 15: 1418061, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38903499

RESUMO

Extracellular vesicles (EVs), characterized by low immunogenicity, high biocompatibility and targeting specificity along with excellent blood-brain barrier permeability, are increasingly recognized as promising drug delivery vehicles for treating a variety of diseases, such as cancer, inflammation and viral infection. However, recent findings demonstrate that the intracellular delivery efficiency of EVs fall short of expectations due to phagocytic clearance mediated by the host mononuclear phagocyte system through Fcγ receptors, complement receptors as well as non-opsonic phagocytic receptors. In this text, we investigate a range of bacterial virulence proteins that antagonize host phagocytic machinery, aiming to explore their potential in engineering EVs to counteract phagocytosis. Special emphasis is placed on IdeS secreted by Group A Streptococcus and ImpA secreted by Pseudomonas aeruginosa, as they not only counteract phagocytosis but also bind to highly upregulated surface biomarkers αVß3 on cancer cells or cleave the tumor growth and metastasis-promoting factor CD44, respectively. This suggests that bacterial anti-phagocytic proteins, after decorated onto EVs using pre-loading or post-loading strategies, can not only improve EV-based drug delivery efficiency by evading host phagocytosis and thus achieve better therapeutic outcomes but also further enable an innovative synergistic EV-based cancer therapy approach by integrating both phagocytosis antagonism and cancer targeting or deactivation.


Assuntos
Vesículas Extracelulares , Fagocitose , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/metabolismo , Fagocitose/imunologia , Humanos , Animais , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/metabolismo , Integrina alfaVbeta3/metabolismo , Integrina alfaVbeta3/imunologia , Receptores de Hialuronatos/metabolismo , Receptores de Hialuronatos/imunologia , Pseudomonas aeruginosa/imunologia
2.
Angew Chem Int Ed Engl ; 61(10): e202113649, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34994999

RESUMO

Precise and effective manipulation of protein functions still faces tremendous challenges. Herein we report a programmable peptide molecule, consisted of targeting and self-assembly modules, that enables specific and highly efficient assembly governed by targeting receptor proteins. Upon binding to the cell membrane receptor, peptide conformation is somewhat stabilized along with decreased self-assembly activation energy, promoting peptide-protein complex oligomerization. We first design a GNNQQNY-RGD peptide (G7-RGD) to recognize integrin αV ß3 receptor for proof-of-concept study. In the presence of αV ß3 protein, the critical assembly concentration of free G7-RGD decreases from 525 to 33 µM and the resultant G7-RGD cluster drives integrin receptor oligomerization. Finally, a bispecific assembling peptide antiCD3-G7-RGD is rationally designed for cancer immunotherapy, which validates CD3 oligomerization and concomitant T cell activation, leading to T cell-mediated cancer cell cytolysis.


Assuntos
Imunoterapia , Integrina alfaVbeta3/análise , Neoplasias/terapia , Peptídeos/química , Humanos , Integrina alfaVbeta3/imunologia , Neoplasias/imunologia , Peptídeos/imunologia
3.
J Biol Chem ; 296: 100399, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33571526

RESUMO

CD40L is a member of the TNF superfamily that participates in immune cell activation. It binds to and signals through several integrins, including αvß3 and α5ß1, which bind to the trimeric interface of CD40L. We previously showed that several integrin ligands can bind to the allosteric site (site 2), which is distinct from the classical ligand-binding site (site 1), raising the question of if CD40L activates integrins. In our explorations of this question, we determined that integrin α4ß1, which is prevalently expressed on the same CD4+ T cells as CD40L, is another receptor for CD40L. Soluble (s)CD40L activated soluble integrins αvß3, α5ß1, and α4ß1 in cell-free conditions, indicating that this activation does not require inside-out signaling. Moreover, sCD40L activated cell-surface integrins in CHO cells that do not express CD40. To learn more about the mechanism of binding, we determined that sCD40L bound to a cyclic peptide from site 2. Docking simulations predicted that the residues of CD40L that bind to site 2 are located outside of the CD40L trimer interface, at a site where four HIGM1 (hyper-IgM syndrome type 1) mutations are clustered. We tested the effect of these mutations, finding that the K143T and G144E mutants were the most defective in integrin activation, providing support that this region interacts with site 2. We propose that allosteric integrin activation by CD40L also plays a role in CD40L signaling, and defective site 2 binding may be related to the impaired CD40L signaling functions of these HIGM1 mutants.


Assuntos
Ligante de CD40/metabolismo , Integrina alfa4beta1/metabolismo , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Receptores de Superfície Celular/química , Linfócitos T/metabolismo , Sítio Alostérico , Animais , Ligante de CD40/imunologia , Linhagem Celular , Cricetinae , Humanos , Integrina alfa4beta1/imunologia , Integrina alfa5beta1/imunologia , Integrina alfaVbeta3/imunologia , Simulação de Acoplamento Molecular , Ligação Proteica , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Linfócitos T/imunologia
4.
Mol Pharm ; 17(10): 3740-3747, 2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32845640

RESUMO

Cyclic RGD (cRGD) peptide-conjugated boronated albumin was developed to direct toward integrin αvß3, which overexpresses on many cancer cells. A stepwise conjugation of c[RGDfK(Mal)] and maleimide-conjugated closo-dodecaborate (MID) to bovine serum albumin (BSA) afforded cRGD-MID-BSA, which was noncytotoxic toward both U87MG and A549 cells. As compared with l-BPA, selective antitumor activity of cRGD-MID-BSA toward U87MG cells overexpressing integrin αvß3 was identified after thermal neutron irradiation. In vivo fluorescence live imaging of Cy5-conjugated cRGD-MID-BSA and MID-BSA revealed that both cRGD-MID-BSA and MID-BSA similarly reached the maximum accumulation during 8-12 h after injection. The selective accumulation and retention of Cy5-cRGD-MID-BSA was more pronounced than Cy5-MID-BSA after 24 h. An in vivo boron neutron capture therapy (BNCT) study revealed that the cRGD peptide ligand combination enhanced accumulation of MID-BSA into tumor cells in U87MG xenograft models. The significant tumor growth suppression was observed in U87MG xenograft models at a dose of 7.5 mg [10B]/kg after neutron irradiation.


Assuntos
Terapia por Captura de Nêutron de Boro/métodos , Boro/administração & dosagem , Portadores de Fármacos/química , Integrina alfaVbeta3/metabolismo , Isótopos/administração & dosagem , Neoplasias/radioterapia , Animais , Boro/química , Compostos de Boro/administração & dosagem , Compostos de Boro/química , Linhagem Celular Tumoral , Feminino , Humanos , Integrina alfaVbeta3/imunologia , Microscopia Intravital , Isótopos/química , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/imunologia , Neoplasias/patologia , Peptídeos Cíclicos/química , Soroalbumina Bovina/química , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Nat Commun ; 11(1): 1242, 2020 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-32144270

RESUMO

Expression of the matricellular protein CCN1 (CYR61) is associated with inflammation and is required for successful wound repair. Here, we show that CCN1 binds bacterial pathogen-associated molecular patterns including peptidoglycans of Gram-positive bacteria and lipopolysaccharides of Gram-negative bacteria. CCN1 opsonizes methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa and accelerates their removal by phagocytosis and increased production of bactericidal reactive oxygen species in macrophages through the engagement of integrin αvß3. Mice with myeloid-specific Ccn1 deletion and knock-in mice expressing CCN1 unable to bind αvß3 are more susceptible to infection by S. aureus or P. aeruginosa, resulting in increased mortality and organ colonization. Furthermore, CCN1 binds directly to TLR2 and TLR4 to activate MyD88-dependent signaling, cytokine expression and neutrophil mobilization. CCN1 is therefore a pattern recognition receptor that opsonizes bacteria for clearance and functions as a damage-associated molecular pattern to activate inflammatory responses, activities that contribute to wound healing and tissue repair.


Assuntos
Proteína Rica em Cisteína 61/metabolismo , Proteínas Opsonizantes/metabolismo , Infecções por Pseudomonas/imunologia , Infecções Estafilocócicas/imunologia , Receptores Toll-Like/metabolismo , Animais , Proteína Rica em Cisteína 61/genética , Proteína Rica em Cisteína 61/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Técnicas de Introdução de Genes , Técnicas de Silenciamento de Genes , Humanos , Integrina alfaVbeta3/imunologia , Integrina alfaVbeta3/metabolismo , Masculino , Staphylococcus aureus Resistente à Meticilina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Opsonizantes/genética , Moléculas com Motivos Associados a Patógenos/metabolismo , Fagocitose/imunologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/imunologia , Células Sf9 , Transdução de Sinais/imunologia , Infecções Estafilocócicas/microbiologia , Receptores Toll-Like/imunologia
6.
Cancer Res ; 79(19): 5048-5059, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31416839

RESUMO

Tumor-associated macrophages (TAM) are highly expressed within the tumor microenvironment of a wide range of cancers, where they exert a protumor phenotype by promoting tumor cell growth and suppressing antitumor immune function. Here, we show that TAM accumulation in human and mouse tumors correlates with tumor cell expression of integrin αvß3, a known driver of epithelial cancer progression and drug resistance. A monoclonal antibody targeting αvß3 (LM609) exploited the coenrichment of αvß3 and TAMs to not only eradicate highly aggressive drug-resistant human lung and pancreas cancers in mice, but also to prevent the emergence of circulating tumor cells. Importantly, this antitumor activity in mice was eliminated following macrophage depletion. Although LM609 had no direct effect on tumor cell viability, it engaged macrophages but not natural killer (NK) cells to induce antibody-dependent cellular cytotoxicity (ADCC) of αvß3-expressing tumor cells despite their expression of the CD47 "don't eat me" signal. In contrast to strategies designed to eliminate TAMs, these findings suggest that anti-αvß3 represents a promising immunotherapeutic approach to redirect TAMs to serve as tumor killers for late-stage or drug-resistant cancers. SIGNIFICANCE: Therapeutic antibodies are commonly engineered to optimize engagement of NK cells as effectors. In contrast, LM609 targets αvß3 to suppress tumor progression and enhance drug sensitivity by exploiting TAMs to trigger ADCC.


Assuntos
Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Integrina alfaVbeta3/imunologia , Macrófagos/imunologia , Neoplasias Epiteliais e Glandulares/imunologia , Animais , Antineoplásicos/farmacologia , Progressão da Doença , Humanos , Camundongos , Neoplasias Epiteliais e Glandulares/patologia , Fagocitose/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
7.
Anal Chem ; 91(17): 11045-11054, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31361124

RESUMO

Early precise diagnosis of cancers is crucial to realize more effective therapeutic interventions with minimal toxic effects. Cancer phenotypes may also alter greatly among patients and within individuals over the therapeutic process. The identification and characterization of specific biomarkers expressed on tumor cells are in high demand for diagnosis and treatment, but they are still a challenge. Herein, we designed three new bioorthogonal surface-enhanced Raman scattering (SERS) nanoprobes and successfully applied the cocktail of them for MDA-MB-231 and MCF-7 breast cancer multiplex phenotype detection. The SERS nanoprobes containing Raman reporters with diynl, azide, or cyano moieties demonstrated apparent Raman shift peaks in 2205, 2120, and 2230 cm-1, respectively, in the biologically Raman-silent region. Three target ligands, including oligonucleotide aptamer (AS1411), arginine-glycine-aspatic acid (RGD) peptide, and homing cell adhesion molecule antibody (anti-CD44), were separately conjugated to the nanoprobes for active recognition capability. The cocktail of the nanoprobes manifested minimal cytotoxicity and simultaneously multiplex phenotype imaging of MDA-MB-231 and MCF-7 cells. Quantitative measurement of cellular uptake by inductively coupled plasma mass spectrometry (ICPMS) verified that MDA-MB-231 cells harbored a much higher expression level of CD44 receptor than MCF-7 cells. For in vivo SERS detection, Raman shift peaks of 2120, 2205, and 2230 cm-1 in the micro-tumor were clearly observed, representing the existence of three specific biomarkers of nucleolin, integrin αvß3, and CD44 reporter, which could be used for early cancer phenotype identification. The biodistribution results indicated that target ligand modified nanoprobes exhibited much more accumulation in tumors than those nanoprobes without target ligands. The multicolor cocktail of bioorthogonal SERS nanoprobes offers an attractive and insightful strategy for early cancer multiplex phenotype targeting and diagnosis in vivo that is noninvasive and has low cross-talk, unique spectral-molecular signature, high sensitivity, and negligible background interference.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/diagnóstico por imagem , Imunoconjugados/química , Sondas Moleculares/química , Nanopartículas/química , Análise Espectral Raman/métodos , Animais , Anticorpos/química , Aptâmeros de Nucleotídeos/química , Biomarcadores Tumorais/imunologia , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Feminino , Expressão Gênica , Xenoenxertos , Humanos , Receptores de Hialuronatos/antagonistas & inibidores , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/imunologia , Imunoconjugados/administração & dosagem , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/imunologia , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sondas Moleculares/administração & dosagem , Nanopartículas/administração & dosagem , Oligopeptídeos/química , Fosfoproteínas/genética , Fosfoproteínas/imunologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/imunologia , Espectrofotometria Atômica , Nucleolina
8.
Nat Commun ; 10(1): 1482, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30931941

RESUMO

Integrins are components of cell-matrix adhesions, and function as scaffolds for various signal transduction pathways. So far no lipid ligand for integrin has been reported. Here we show that a lipid, oxysterol 25-hydroxycholesterol (25HC), directly binds to α5ß1 and αvß3 integrins to activate integrin-focal adhesion kinase (FAK) signaling. Treatment of macrophages and epithelial cells with 25HC results in an increase in activated αvß3 integrin in podosome and focal adhesion matrix adhesion sites. Moreover, activation of pattern recognition receptor on macrophages induces secretion of 25HC, triggering integrin signaling and the production of proinflammatory cytokines such as TNF and IL-6. Thus, the lipid molecule 25HC is a physiologically relevant activator of integrins and is involved in positively regulating proinflammatory responses. Our data suggest that extracellular 25HC links innate immune inflammatory response with integrin signaling.


Assuntos
Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Hidroxicolesteróis/metabolismo , Imunidade Inata/imunologia , Integrina alfa5beta1/imunologia , Integrina alfaVbeta3/imunologia , Macrófagos/imunologia , Animais , Adesões Focais , Inflamação , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Interleucina-6/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/imunologia
9.
Adv Healthc Mater ; 8(4): e1801234, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30537061

RESUMO

Macrophages are a predominant immune cell population that drive inflammatory responses and exhibit transitions in phenotype and function during tissue remodeling in disease and repair. Thus, engineering an immunomodulatory biomaterial has significant implications for resolving inflammation. Here, a biomimetic and photoresponsive hyaluronan (HA) hydrogel nanocomposite with tunable 3D extracellular matrix (ECM) adhesion sites for dynamic macrophage immunomodulation is engineered. Photodegradative alkoxylphenacyl-based polycarbonate (APP) nanocomposites are exploited to permit user-controlled Arg-Gly-Asp (RGD) adhesive peptide release and conjugation to a HA-based ECM for real-time integrin activation of macrophages encapsulated in 3D HA-APP nanocomposite hydrogels. It is demonstrated that photocontrolled 3D ECM-RGD peptide conjugation can activate αvß3 integrin of macrophages, and periodic αvß3 integrin activation can enhance anti-inflammatory M2 macrophage polarization. Altogether, an emerging use of biomimetic, photoresponsive, and bioactive HA-APP nanocomposite hydrogel is highlighted to command 3D cell-ECM interactions for modulating macrophage polarization, which may shed light on cell-ECM interactions in innate immunity and inspire new biomaterial-based immunomodulatory therapies.


Assuntos
Ácido Hialurônico , Hidrogéis , Imunomodulação , Macrófagos/imunologia , Nanocompostos/química , Raios Ultravioleta , Animais , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Hidrogéis/química , Hidrogéis/farmacologia , Imunomodulação/efeitos dos fármacos , Imunomodulação/efeitos da radiação , Integrina alfaVbeta3/imunologia , Camundongos , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Células RAW 264.7
10.
J Thromb Haemost ; 16(9): 1843-1856, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29953749

RESUMO

Essentials The pathogenesis of immune thrombocytopenia (ITP) has not been fully clarified. We analyzed the role of anti-αvß3 autoantibody in the pathogenesis of ITP in patients. Anti-αvß3 autoantibody impeded megakaryocyte migration and adhesion to the vascular niche. Anti-αv ß3 autoantibody potentially contributes to the pathogenesis of refractory ITP. SUMMARY: Background The pathogenesis of immune thrombocytopenia (ITP) has not been fully clarified. Anti-αvß3 integrin autoantibody is detected in chronic ITP patients, but its contribution to ITP is still unclear. Objectives To clarify the potential role of anti-αvß3 integrin autoantibody in chronic ITP and the related mechanism. Methods Relationship between levels of anti-αvß3 autoantibody and platelets in chronic ITP patients was evaluated. The influence of anti-αvß3 antibody on megakaryocyte (MK) survival, differentiation, migration and adhesion was assessed, and the associated signal pathways were investigated. Platelet recovery and MKs' distribution were observed in an ITP mouse model pretreated with different antibodies. Result In this study, we showed that the anti-αvß3 autoantibody usually coexists with anti-αIIbß3 autoantibody in chronic ITP patients, and patients with both autoantibodies have lower platelets. In in vitro studies, we showed that the anti-αvß3 antibody had no significant effect on the survival and proliferation of MKs, whereas it decreased formations of proplatelet significantly. Anti-αvß3 antibody impeded stromal cell derived facor-1 alpha (SDF-1α)- mediated migration and inhibited the phosphorylation of protein kinase B. Anti-αvß3 antibody significantly inhibited MKs' adhesion to endothelial cells and Fibrogen. The phosphorylation of focal adhesion kinase and proto-oncogene tyrosine-protein kinase Src induced by adhesion was inhibited when MKs were pretreated with anti-αvß3 antibody. In in vivo studies, we showed that injection with anti-αv antibody delayed platelet recovery in a mouse model of ITP. Conclusions These findings demonstrate that the autoantibody against integrin αv ß3 may aggravate thrombocytopenia in ITP patients by impeding MK migration and adhesion to the vascular niche, which provides new insights into the pathogenesis of ITP.


Assuntos
Autoanticorpos/imunologia , Autoantígenos/imunologia , Integrina alfaVbeta3/imunologia , Megacariócitos/imunologia , Púrpura Trombocitopênica Idiopática/imunologia , Adolescente , Adulto , Idoso , Animais , Adesão Celular , Movimento Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Células Endoteliais/metabolismo , Feminino , Sangue Fetal/citologia , Humanos , Masculino , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Fosforilação , Contagem de Plaquetas , Glicoproteína IIb da Membrana de Plaquetas/imunologia , Proteínas Quinases/metabolismo , Processamento de Proteína Pós-Traducional , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Púrpura Trombocitopênica Idiopática/sangue , Células Estromais/metabolismo , Trombopoese , Adulto Jovem
11.
Front Immunol ; 9: 3118, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30687322

RESUMO

Intestinal macrophages are highly mobile cells with extraordinary plasticity and actively contribute to cytokine-mediated epithelial cell damage. The mechanisms triggering macrophage polarization into a proinflammatory phenotype are unknown. Here, we report that during inflammation macrophages enhance its intercellular adhesion properties in order to acquire a M1-phenotype. Using in vitro and in vivo models we demonstrate that intercellular adhesion is mediated by integrin-αVß3 and relies in the presence of the unconventional class I myosin 1F (Myo1F). Intercellular adhesion mediated by αVß3 stimulates M1-like phenotype in macrophages through hyperactivation of STAT1 and STAT3 downstream of ILK/Akt/mTOR signaling. Inhibition of integrin-αVß3, Akt/mTOR, or lack of Myo1F attenuated the commitment of macrophages into a pro-inflammatory phenotype. In a model of colitis, Myo1F deficiency strongly reduces the secretion of proinflammatory cytokines, decreases epithelial damage, ameliorates disease activity, and enhances tissue repair. Together our findings uncover an unknown role for Myo1F as part of the machinery that regulates intercellular adhesion and polarization in macrophages.


Assuntos
Colite Ulcerativa/imunologia , Integrina alfaVbeta3/metabolismo , Ativação de Macrófagos , Macrófagos/imunologia , Miosina Tipo I/metabolismo , Animais , Linhagem Celular Tumoral , Colite Ulcerativa/induzido quimicamente , Citoesqueleto/imunologia , Citoesqueleto/metabolismo , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Humanos , Integrina alfaVbeta3/imunologia , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miosina Tipo I/genética , Miosina Tipo I/imunologia , Cultura Primária de Células , Células RAW 264.7
12.
Structure ; 25(11): 1732-1739.e5, 2017 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-29033288

RESUMO

The LM609 antibody specifically recognizes αVß3 integrin and inhibits angiogenesis, bone resorption, and viral infections in an arginine-glycine-aspartate-independent manner. LM609 entered phase II clinical trials for the treatment of several cancers and was also used for αVß3-targeted radioimmunotherapy. To elucidate the mechanisms of recognition and inhibition of αVß3 integrin, we solved the structure of the LM609 antigen-binding fragment by X-ray crystallography and determined its binding affinity for αVß3. Using single-particle electron microscopy, we show that LM609 binds at the interface between the ß-propeller domain of the αV chain and the ßI domain of the ß3 chain, near the RGD-binding site, of all observed integrin conformational states. Integrating these data with fluorescence size-exclusion chromatography, we demonstrate that LM609 sterically hinders access of large ligands to the RGD-binding pocket, without obstructing it. This work provides a structural framework to expedite future efforts utilizing LM609 as a diagnostic or therapeutic tool.


Assuntos
Anticorpos Monoclonais/química , Antígenos/química , Fragmentos Fab das Imunoglobulinas/química , Integrina alfaVbeta3/química , Oligopeptídeos/química , Motivos de Aminoácidos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/imunologia , Inibidores da Angiogênese/metabolismo , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/imunologia , Antígenos/genética , Antígenos/imunologia , Antivirais/química , Antivirais/imunologia , Antivirais/metabolismo , Sítios de Ligação , Conservadores da Densidade Óssea/química , Conservadores da Densidade Óssea/imunologia , Conservadores da Densidade Óssea/metabolismo , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/imunologia , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/imunologia , Ligantes , Modelos Moleculares , Oligopeptídeos/genética , Oligopeptídeos/imunologia , Ligação Proteica , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
13.
Atherosclerosis ; 258: 40-50, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28189040

RESUMO

BACKGROUND AND AIMS: Diabetes is a major risk factor for the development of atherosclerosis. Hyperglycemia stimulates vascular smooth muscle cells (VSMC) to secrete ligands that bind to the αVß3 integrin, a receptor that regulates VSMC proliferation and migration. This study determined whether an antibody that had previously been shown to block αVß3 activation and to inhibit VSMC proliferation and migration in vitro, inhibited the development of atherosclerosis in diabetic pigs. METHODS: Twenty diabetic pigs were maintained on a high fat diet for 22 weeks. Ten received injections of anti-ß3 F(ab)2 and ten received control F(ab)2 for 18 weeks. RESULTS: The active antibody group showed reduction of atherosclerosis of 91 ± 9% in the left main, 71 ± 11%, in left anterior descending, 80 ± 10.2% in circumflex, and 76 ± 25% in right coronary artery, (p < 0.01 compared to lesions areas from corresponding control treated arteries). There were significant reductions in both cell number and extracellular matrix. Histologic analysis showed neointimal hyperplasia with macrophage infiltration, calcifications and cholesterol clefts. Antibody treatment significantly reduced number of macrophages contained within lesions, suggesting that this change contributed to the decrease in lesion cellularity. Analysis of the biochemical changes within the femoral arteries that received the active antibody showed a 46 ± 12% (p < 0.05) reduction in the tyrosine phosphorylation of the ß3 subunit of αVß3 and a 40 ± 14% (p < 0.05) reduction in MAP kinase activation. CONCLUSIONS: Blocking ligand binding to the αVß3 integrin inhibits its activation and attenuates increased VSMC proliferation that is induced by chronic hyperglycemia. These changes result in significant decreases in atherosclerotic lesion size in the coronary arteries. The results suggest that this approach may have efficacy in treating the proliferative phase of atherosclerosis in patients with diabetes.


Assuntos
Doença da Artéria Coronariana/prevenção & controle , Diabetes Mellitus Experimental/tratamento farmacológico , Angiopatias Diabéticas/prevenção & controle , Fragmentos Fab das Imunoglobulinas/farmacologia , Integrina alfaVbeta3/antagonistas & inibidores , Músculo Liso Vascular/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Doença da Artéria Coronariana/etiologia , Doença da Artéria Coronariana/metabolismo , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/metabolismo , Angiopatias Diabéticas/etiologia , Angiopatias Diabéticas/metabolismo , Artéria Femoral/efeitos dos fármacos , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Injeções Subcutâneas , Integrina alfaVbeta3/imunologia , Integrina alfaVbeta3/metabolismo , Ligantes , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Neointima , Fosforilação , Placa Aterosclerótica , Ligação Proteica , Sus scrofa
14.
Biopolymers ; 108(1)2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27603160

RESUMO

Peptide-conjugated polysaccharide matrices using bioactive laminin-derived peptides are useful biomaterials for tissue and cell engineering. Here, we demonstrate an easy handling preparation method for peptide-polysaccharide matrices using polyion complex with both alginate and chitosan. First, aldehyde-alginate was synthesized by oxidization of alginate using NaIO4 , and then, reacted with Cys-peptides. Next, the peptide-alginate solution was added to a chitosan-coated plate, and the peptide-polyion complex matrices (peptide-PCMs) were prepared. The peptide-PCMs using an integrin αvß3-binding peptide (A99a: ALRGDN, mouse laminin α1 chain 1145-1150) and an integrin α2ß1-binding peptide (EF1XmR: RLQLQEGRLHFXFD, X = Nle, mouse laminin α1 chain 2751-2763) showed strong cell attachment activity in a dose-dependent manner. When we examined the effect of various spacers on the biological activity of A99a-PCM, hydrophobic and long spacers enhanced the cell attachment activity. Further, the A99a-PCM with the spacers strongly promoted neurite outgrowth. The polyion complex method is an easy way to obtain insolubilized matrix and is widely applicable for various polysaccharides. The peptide-PCM is useful as a biomaterial for cell and tissue engineering.


Assuntos
Alginatos/química , Quitosana/química , Peptídeos/química , Aldeídos/química , Sequência de Aminoácidos , Animais , Anticorpos/química , Anticorpos/imunologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Ácido Edético/química , Heparina/química , Humanos , Integrina alfa1beta1/química , Integrina alfa1beta1/imunologia , Integrina alfaVbeta3/química , Integrina alfaVbeta3/imunologia , Laminina/química , Camundongos , Microscopia de Fluorescência , Neuritos/metabolismo , Oxirredução , Peptídeos/síntese química , Peptídeos/metabolismo , Peptídeos/farmacologia
15.
Biochem J ; 474(4): 589-596, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27993971

RESUMO

Tetraspanins play important roles in normal (e.g. cell adhesion, motility, activation, and proliferation) and pathological conditions (e.g. metastasis and viral infection). Tetraspanins interact with integrins and regulate integrin functions, but the specifics of tetraspanin-integrin interactions are unclear. Using co-immunoprecipitation with integrins as a sole method to detect interaction between integrins and full-length tetraspanins, it has been proposed that the variable region (helices D and E) of the extracellular-2 (EC2) domain of tetraspanins laterally associates with a non-ligand-binding site of integrins. We describe that, using adhesion assays, the EC2 domain of CD81, CD9, and CD151 bound to integrin αvß3, and this binding was suppressed by cRGDfV, a specific inhibitor of αvß3, and antibody 7E3, which is mapped to the ligand-binding site of ß3. We also present evidence that the specificity loop of ß3 directly bound to the EC2 domains. This suggests that the EC2 domains specifically bind to the classical ligand-binding site of αvß3. αvß3 was a more effective receptor for the EC2 domains than the previously known tetraspanin receptors α3ß1, α4ß1, and α6ß1. Docking simulation predicted that the helices A and B of CD81 EC2 bind to the RGD-binding site of αvß3. Substituting Lys residues at positions 116 and 144/148 of CD81 EC2 in the predicted integrin-binding interface reduced the binding of CD81 EC2 to αvß3, consistent with the docking model. These findings suggest that, in contrast with previous models, the ligand-binding site of integrin αvß3, a new tetraspanin receptor, binds to the constant region (helices A and B) of the EC2 domain.


Assuntos
Integrina alfaVbeta3/química , Tetraspanina 24/química , Tetraspanina 28/química , Tetraspanina 29/química , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/química , Sítios de Ligação , Células CHO , Clonagem Molecular , Cricetulus , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/imunologia , Cinética , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Tetraspanina 24/genética , Tetraspanina 24/imunologia , Tetraspanina 28/genética , Tetraspanina 28/imunologia , Tetraspanina 29/genética , Tetraspanina 29/imunologia
16.
J Neuroinflammation ; 13(1): 227, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27586239

RESUMO

BACKGROUND: The pro-inflammatory cytokine, tumor necrosis factor-α (TNF-α), is expressed in ischemic tissue and is known to modulate angiogenesis; however, the role of the two distinct TNF-α receptors, TNFR1 and TNFR2, in mediating angiogenic signaling after cerebral ischemic stroke is relatively unknown. METHODS: C57BL6 mice were subject to 90 min of ischemia by temporary occlusion of the middle cerebral artery (MCAO) and given daily intra-cerebroventricular injections of antibodies against TNFR1, TNFR2 or control IgG (doses of 10, 50, and 100 ng/day) for 4 days following 90 min MCAO. Vascular remodeling and α5ß1 and αVß3 integrin expression were then examined in the brains of these mice after 4, 7, and 14 days post-ischemia. In parallel in vitro studies, flow cytometry was used to determine the influence of TNF-α on proliferation and integrin expression of human brain microvascular endothelial cells (HBMECs). RESULTS: The post-ischemic cerebral angiogenic response was inhibited by antibodies against TNFR1 but not TNFR2, and this correlated with reduced endothelial proliferation and decreased α5ß1 and αVß3 integrin expression after 4 and 7 days post-ischemia. Consistent with these findings, in vitro studies showed that TNF-α induced endothelial proliferation and upregulation of α5ß1 and αVß3 integrins was abrogated by anti-TNFR1 but not anti-TNFR2 antibodies in cultured HBMECs. In addition, blocking antibodies to α5ß1 and αVß3 integrins significantly inhibited TNF-α-induced HBMEC proliferation. CONCLUSIONS: Our results suggest that TNFR1-mediated signaling plays a critical role in triggering angiogenic integrins and subsequent angiogenic responses following cerebral ischemia. These novel findings could form a platform for future therapeutic strategies aimed at stimulating angiogenesis following cerebral ischemia.


Assuntos
Anticorpos/farmacologia , Integrina alfa5beta1/metabolismo , Integrina alfaVbeta3/metabolismo , Neovascularização Patológica/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Encéfalo/citologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Infarto da Artéria Cerebral Média/complicações , Integrina alfa5beta1/imunologia , Integrina alfaVbeta3/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/etiologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
17.
PLoS One ; 11(7): e0158592, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27391603

RESUMO

OBJECTIVE: Hepatocellular carcinoma (HCC), mostly derived from hepatitis or cirrhosisis, is one of the most common types of liver cancer. T-cell mediated immune response elicited by CD/TK double suicide gene has shown a substantial antitumor effect in HCC. Integrin αVß3 over expresssion has been suggested to regulate the biology behavior of HCC. In this study, we investigated the strategy of incorporating CD/TK double suicide gene and anti-αVß3 integrin monoclonal antibodies into cationic microbubbles (CMBsαvß3), and evaluated its killing effect in HCC cells. METHODS: To improve the transfection efficiency of targeted CD/TK double suicide gene, we adopted cationic microbubbles (CMBs), a cationic delivery agent with enhanced DNA-carrying capacity. The ultrasound and high speed shearing method was used to prepare the non-targeting cationic microbubbles (CMBs). Using the biotin-avidin bridge method, αVß3 integrin antibody was conjugated to CMBs, and CMBsαvß3 was generated to specifically target to HepG2 cells. The morphology and physicochemical properties of the CMBsαvß3 was detected by optical microscope and zeta detector. The conjugation of plasmid and the antibody in CMBsαvß3 were examined by immunofluorescent microscopy and flow cytometry. The binding capacities of CMBsαvß3 and CMBs to HCC HepG2 and normal L-02 cells were compared using rosette formation assay. To detect EGFP fluorescence and examine the transfection efficiencies of CMBsαvß3 and CMBs in HCC cells, fluorescence microscope and contrast-enhanced sonography were adopted. mRNA and protein level of CD/TK gene were detected by RT-PCR and Western blot, respectively. To evaluate the anti-tumor effect of CMBsαvß3, HCC cells with CMBsαvß3 were exposed to 5-flurocytosine / ganciclovir (5-FC/GCV). Then, cell cycle distribution after treatment were detected by PI staining and flow cytometry. Apoptotic cells death were detected by optical microscope and assessed by MTT assay and TUNEL-staining assay. RESULTS: CMBsαvß3 had a regular shape and good dispersion. Compared to CMBs, CMBsαvß3 had more stable concentrations of αVß3 ligand and pEGFP-KDRP-CD/TK, and CMBsαvß3 was much sticker to HepG2 HCC cells than normal liver L-02cells. Moreover, after exposed to anti-αVß3 monoclonal antibody, the adhesion of CMBsαvß3 to HepG2 cells and L-02 cells were significantly reduced. Also, CMBsαvß3 demonstrated a substantially higher efficiency in pEGFP-KDRP-CD/TK plasmid transfection in HepG2 cells than CMBs. In addition, CMBsαvß3 could significantly facilitate 5-FC/GCV-induced cell cycle arrest in S phase. Moreover, treatment of 5-FC/GCV combined with CMBsαvß3 resulted in a marked apoptotic cell death in HepG2 and SK-Herp-1 HCC cells. In vitro, treatment of 5-FC/GCV combined with CMBsαvß3 suppresed cell proliferation. In nude mice model, 5-FU + GCV combined with plasmid + CMBsαvß3were able to significantly suppress tumor volumes. CONCLUSION: Through biotin-avidin mediation system, CMBsαvß3 were successfully generated to specifically target HCC HepG2 cells. More importantly, CMBsαvß3 could significantly facilitate 5-FC/GCV-induced cell cycle arrest and apoptotic cell death in HepG2 cells. Our study demonstrated a potential strategy that could be translated clinically to improve liver tumor gene delivery.


Assuntos
Anticorpos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Citosina Desaminase/metabolismo , Integrina alfaVbeta3/imunologia , Neoplasias Hepáticas/tratamento farmacológico , Microbolhas , Timidina Quinase/metabolismo , Animais , Anticorpos/imunologia , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Citosina Desaminase/genética , Citometria de Fluxo , Células Hep G2 , Humanos , Immunoblotting , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fase S , Timidina Quinase/genética , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Leukoc Biol ; 100(5): 855-864, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27317750

RESUMO

ADAM23 is a member of the brain macrophage-derived chemokine family. Structural homology of ADAM proteins suggests their function as integrin receptors. Previous studies have linked ADAM23 as a dominant contributor to brain development and cancer metastasis. The present studies now show that ADAM23 expression on DCs partially governs antigen-presentation capacities to responder CD4+ T cells. With the use of RNAi approaches, knockdown of ADAM23 in murine BMDCs resulted in impaired T cell activation, proliferation, and cytokine production. Knockdown did not alter the maturation profile of DCs (i.e., costimulatory molecule expression or production of proinflammatory cytokines) but markedly impaired cognate T cell responses. There was a significant decrease in antigen-specific clonal expansion coupled with a global decrease in Th cytokine production. Impaired early activation and proliferation did not alter/skew the balance of Th polarization but significantly depressed total levels of IL-2, IFN-γ, IL-4, and IL-17 cytokine production in CD4+ T cells primed by ADAM23 knockdown versus control DCs. Finally, neutralizing antibodies targeting the α(v)ß(3) integrin receptors resulted in similar phenotypes of impaired CD4+ T cell responses. Taken together, these studies show a novel role of ADAM23 in governing DC antigen presentation to cognate CD4+ T cells.


Assuntos
Proteínas ADAM/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Citocinas/biossíntese , Células Dendríticas/imunologia , Integrina alfaVbeta3/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/genética , Animais , Anticorpos Neutralizantes/imunologia , Linfócitos T CD4-Positivos/imunologia , Divisão Celular/efeitos dos fármacos , Citocinas/genética , Técnicas de Silenciamento de Genes , Integrina alfaVbeta3/imunologia , Ativação Linfocitária , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Ovalbumina/imunologia , Fragmentos de Peptídeos/imunologia , Interferência de RNA , RNA Interferente Pequeno/genética
19.
Virology ; 492: 251-8, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27011223

RESUMO

Integrins can function as receptors for foot-and-mouth disease virus (FMDV) in epithelium. Horses are believed to be insusceptible to this disease, but the mechanism of resistance remains unclear. To detect whether FMDV can use integrin to attach to equine epithelial, we compared the utilities of αvß3 and αvß6 between bovine and equine kidney epithelial cells (KECs). Equine KECs showed almost equal efficiency to those of bovine. Further, the integrin αv, ß3, and ß6 subunits from bovine and equine were cloned and vectors were transfected into SW480 cells and COS-1 cells alone or together, and virus titers were used to determine the viral replication. In all cases, the virus reproduced successfully. Overall, FMDV can replicate in SW480 cells transfected with equine ß3/ß6 subunits and COS-1 cells transfected with equine αvß3/αvß6 integrins, but not in EKECs. These results indicated that failure of FMDV replication in EKECs was not attributed to integrin receptors.


Assuntos
Antígenos de Neoplasias/imunologia , Resistência à Doença , Células Epiteliais/imunologia , Vírus da Febre Aftosa/genética , Integrina alfaVbeta3/imunologia , Integrinas/imunologia , Receptores Virais/imunologia , Animais , Antígenos de Neoplasias/genética , Células COS , Bovinos , Chlorocebus aethiops , Clonagem Molecular , Cricetulus , Células Epiteliais/patologia , Células Epiteliais/virologia , Vírus da Febre Aftosa/crescimento & desenvolvimento , Expressão Gênica , Cavalos , Especificidade de Hospedeiro , Integrina alfaVbeta3/genética , Integrinas/genética , Rim/imunologia , Rim/patologia , Rim/virologia , Receptores Virais/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transgenes , Replicação Viral
20.
Oncogene ; 35(15): 1977-87, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-26165836

RESUMO

Ovarian carcinoma is the fifth common cause of cancer death in women, despite advanced therapeutic approaches. αvß3 integrin, a plasma membrane receptor, binds thyroid hormones (L-thyroxine, T4; 3,5,3'-triiodo-L-thyronine, T3) and is overexpressed in ovarian cancer. We have demonstrated selective binding of fluorescently labeled hormones to αvß3-positive ovarian cancer cells but not to integrin-negative cells. Physiologically relevant T3 (1 nM) and T4 (100 nM) concentrations in OVCAR-3 (high αvß3) and A2780 (low αvß3) cells promoted αv and ß3 transcription in association with basal integrin levels. This transcription was effectively blocked by RGD (Arg-Gly-Asp) peptide and neutralizing αvß3 antibodies, excluding T3-induced ß3 messenger RNA, suggesting subspecialization of T3 and T4 binding to the integrin receptor pocket. We have provided support for extracellular regulated kinase (ERK)-mediated transcriptional regulation of the αv monomer by T3 and of ß3 monomer by both hormones and documented a rapid (30-120 min) and dose-dependent (0.1-1000 nM) ERK activation. OVCAR-3 cells and αvß3-deficient HEK293 cells treated with αvß3 blockers confirmed the requirement for an intact thyroid hormone-integrin interaction in ERK activation. In addition, novel data indicated that T4, but not T3, controls integrin's outside-in signaling by phosphorylating tyrosine 759 in the ß3 subunit. Both hormones induced cell proliferation (cell counts), survival (Annexin-PI), viability (WST-1) and significantly reduced the expression of genes that inhibit cell cycle (p21, p16), promote mitochondrial apoptosis (Nix, PUMA) and tumor suppression (GDF-15, IGFBP-6), particularly in cells with high integrin expression. At last, we have confirmed that hypothyroid environment attenuated ovarian cancer growth using a novel experimental platform that exploited paired euthyroid and severe hypothyroid serum samples from human subjects. To conclude, our data define a critical role for thyroid hormones as potent αvß3-ligands, driving ovarian cancer cell proliferation and suggest that disruption of this axis may present a novel treatment strategy in this aggressive disease.


Assuntos
Integrina alfaVbeta3/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Ovarianas/metabolismo , Tiroxina/fisiologia , Tri-Iodotironina/fisiologia , Anticorpos Neutralizantes/farmacologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Meios de Cultura/farmacologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hipotireoidismo/sangue , Integrina alfaV/genética , Integrina alfaV/metabolismo , Integrina alfaVbeta3/biossíntese , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/imunologia , Integrina beta3/genética , Integrina beta3/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/patologia , Oligopeptídeos/farmacologia , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Fosforilação , Processamento de Proteína Pós-Traducional , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Tiroxina/sangue , Tiroxina/farmacologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/fisiologia , Tri-Iodotironina/sangue , Tri-Iodotironina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA