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










Base de dados
Intervalo de ano de publicação
1.
Mucosal Immunol ; 14(2): 366-376, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32814824

RESUMO

In addition to their well characterized role in mediating IgE-dependent allergic diseases, aberrant accumulation and activation of mast cells (MCs) is associated with many non-allergic inflammatory diseases, whereby their activation is likely triggered by non-IgE stimuli (e.g., IL-33). Siglec-8 is an inhibitory receptor expressed on MCs and eosinophils that has been shown to inhibit IgE-mediated MC responses and reduce allergic inflammation upon ligation with a monoclonal antibody (mAb). Herein, we evaluated the effects of an anti-Siglec-8 mAb (anti-S8) in non-allergic disease models of experimental cigarette-smoke-induced chronic obstructive pulmonary disease and bleomycin-induced lung injury in Siglec-8 transgenic mice. Therapeutic treatment with anti-S8 inhibited MC activation and reduced recruitment of immune cells, airway inflammation, and lung fibrosis. Similarly, using a model of MC-dependent, IL-33-induced inflammation, anti-S8 treatment suppressed neutrophil influx, and cytokine production through MC inhibition. Transcriptomic profiling of MCs further demonstrated anti-S8-mediated downregulation of MC signaling pathways induced by IL-33, including TNF signaling via NF-κB. Collectively, these findings demonstrate that ligating Siglec-8 with an antibody reduces non-allergic inflammation and inhibits IgE-independent MC activation, supporting the evaluation of an anti-Siglec-8 mAb as a therapeutic approach in both allergic and non-allergic inflammatory diseases in which MCs play a role.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos B/metabolismo , Lectinas/metabolismo , Mastócitos/imunologia , Pneumonia/imunologia , Doença Pulmonar Obstrutiva Crônica/metabolismo , Sistema Respiratório/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Antígenos CD/genética , Antígenos de Diferenciação de Linfócitos B/genética , Degranulação Celular , Fumar Cigarros , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Imunoglobulina E/metabolismo , Interleucina-33/metabolismo , Lectinas/genética , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Ativação de Neutrófilo , Transdução de Sinais
2.
Cells ; 10(1)2020 12 24.
Artigo em Inglês | MEDLINE | ID: mdl-33374255

RESUMO

Siglecs (sialic acid-binding immunoglobulin-like lectins) are single-pass cell surface receptors that have inhibitory activities on immune cells. Among these, Siglec-8 is a CD33-related family member selectively expressed on human mast cells and eosinophils, and at low levels on basophils. These cells can participate in inflammatory responses by releasing mediators that attract or activate other cells, contributing to the pathogenesis of allergic and non-allergic diseases. Since its discovery in 2000, initial in vitro studies have found that the engagement of Siglec-8 with a monoclonal antibody or with selective polyvalent sialoglycan ligands induced the cell death of eosinophils and inhibited mast cell degranulation. Anti-Siglec-8 antibody administration in vivo to humanized and transgenic mice selectively expressing Siglec-8 on mouse eosinophils and mast cells confirmed the in vitro findings, and identified additional anti-inflammatory effects. AK002 (lirentelimab) is a humanized non-fucosylated IgG1 antibody against Siglec-8 in clinical development for mast cell- and eosinophil-mediated diseases. AK002 administration has safely demonstrated the inhibition of mast cell activity and the depletion of eosinophils in several phase 1 and phase 2 trials. This article reviews the discovery and functions of Siglec-8, and strategies for its therapeutic targeting for the treatment of eosinophil- and mast cell-associated diseases.


Assuntos
Anticorpos Monoclonais Humanizados , Antígenos CD , Antígenos de Diferenciação de Linfócitos B , Eosinófilos/imunologia , Hipersensibilidade , Inflamação , Lectinas , Mastócitos/imunologia , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacologia , Antígenos CD/imunologia , Antígenos CD/fisiologia , Antígenos de Diferenciação de Linfócitos B/imunologia , Antígenos de Diferenciação de Linfócitos B/fisiologia , Ensaios Clínicos como Assunto , Eosinófilos/patologia , Humanos , Hipersensibilidade/tratamento farmacológico , Hipersensibilidade/imunologia , Inflamação/tratamento farmacológico , Inflamação/imunologia , Lectinas/imunologia , Lectinas/fisiologia , Mastócitos/patologia , Camundongos , Camundongos Transgênicos
3.
JCI Insight ; 4(19)2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31465299

RESUMO

Aberrant accumulation and activation of eosinophils and potentially mast cells (MCs) contribute to the pathogenesis of eosinophilic gastrointestinal diseases (EGIDs), including eosinophilic esophagitis (EoE), gastritis (EG), and gastroenteritis (EGE). Current treatment options, such as diet restriction and corticosteroids, have limited efficacy and are often inappropriate for chronic use. One promising new approach is to deplete eosinophils and inhibit MCs with a monoclonal antibody (mAb) against sialic acid-binding immunoglobulin-like lectin 8 (Siglec-8), an inhibitory receptor selectively expressed on MCs and eosinophils. Here, we characterize MCs and eosinophils from human EG and EoE biopsies using flow cytometry and evaluate the effects of an anti-Siglec-8 mAb using a potentially novel Siglec-8-transgenic mouse model in which EG/EGE was induced by ovalbumin sensitization and intragastric challenge. MCs and eosinophils were significantly increased and activated in human EG and EoE biopsies compared with healthy controls. Similar observations were made in EG/EGE mice. In Siglec-8-transgenic mice, anti-Siglec-8 mAb administration significantly reduced eosinophils and MCs in the stomach, small intestine, and mesenteric lymph nodes and decreased levels of inflammatory mediators. In summary, these findings suggest a role for both MCs and eosinophils in EGID pathogenesis and support the evaluation of anti-Siglec-8 as a therapeutic approach that targets both eosinophils and MCs.


Assuntos
Anticorpos Monoclonais/farmacologia , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos B/imunologia , Enterite/tratamento farmacológico , Eosinofilia/tratamento farmacológico , Eosinófilos/efeitos dos fármacos , Gastrite/tratamento farmacológico , Lectinas/efeitos dos fármacos , Mastócitos/imunologia , Animais , Modelos Animais de Doenças , Enterite/imunologia , Eosinofilia/imunologia , Esofagite Eosinofílica/tratamento farmacológico , Eosinófilos/imunologia , Feminino , Gastrite/imunologia , Gastroenterite , Humanos , Lectinas/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina
4.
Int Arch Allergy Immunol ; 180(2): 91-102, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31401630

RESUMO

INTRODUCTION: Pathologic accumulation and activation of mast cells and eosinophils are implicated in allergic and inflammatory diseases. Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is an inhibitory receptor selectively expressed on mast cells, eosinophils and, at a lower extent, basophils. When engaged with an antibody, Siglec-8 can induce apoptosis of activated eosinophils and inhibit mast cell activation. AK002 is a humanized, non-fucosylated IgG1 anti-Siglec-8 antibody undergoing clinical investigation for treatment of allergic, inflammatory, and proliferative diseases. Here we examine the human tissue selectivity of AK002 and evaluate the in vitro, ex vivo, and in vivo activity of AK002 on eosinophils and mast cells. METHODS: The affinity of AK002 for Siglec-8 and CD16 was determined by biolayer interferometry. Ex vivo activity of AK002 on human eosinophils from blood and dissociated human tissue was tested in apoptosis and antibody-dependent cell-mediated cytotoxicity (ADCC) assays. The in vivo activity of a murine precursor of AK002 (mAK002) was tested in a passive systemic anaphylaxis (PSA) humanized mouse model. RESULTS: AK002 bound selectively to mast cells, eosinophils and, at a lower level, to basophils in human blood and tissue and not to other cell types examined. AK002 induced apoptosis of interleukin-5-activated blood eosinophils and demonstrated potent ADCC activity against blood eosinophils in the presence of natural killer cells. AK002 also significantly reduced eosinophils in dissociated human lung tissue. Furthermore, mAK002 prevented PSA in humanized mice through mast cell inhibition. CONCLUSION: AK002 selectively evokes potent apoptotic and ADCC activity against eosinophils and prevents systemic anaphylaxis through mast cell inhibition.


Assuntos
Anafilaxia/prevenção & controle , Anticorpos Monoclonais Humanizados/imunologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos B/imunologia , Eosinófilos/imunologia , Lectinas/imunologia , Mastócitos/imunologia , Anafilaxia/imunologia , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Basófilos/imunologia , Humanos , Camundongos , Ácido N-Acetilneuramínico/imunologia , Receptores de IgG/imunologia
5.
J Allergy Clin Immunol ; 143(6): 2227-2237.e10, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30543818

RESUMO

BACKGROUND: Sialic acid-binding immunoglobulin-like lectin (Siglec) 8 is selectively expressed on eosinophils, mast cells, and basophils and, when engaged on eosinophils, can cause cell death. OBJECTIVE: We sought to characterize surface and soluble Siglec-8 (sSiglec-8) levels in normal donors (NDs) and eosinophilic donors (EOs) and assess the efficacy of anti-Siglec-8 antibodies in inducing eosinophil cell death in vitro. METHODS: Eosinophil expression of Siglec-8 was assessed by using flow cytometry and quantitative PCR. Serum sSiglec-8 levels were measured by means of ELISA. Induction of eosinophil death by IgG4 (chimeric 2E2 IgG4) and afucosylated IgG1 (chimeric 2E2 IgG1 [c2E2 IgG1]) anti-Siglec-8 antibodies was evaluated in vitro by using flow cytometry and in vivo in humanized mice. RESULTS: Siglec-8 was consistently expressed on eosinophils from NDs and EOs and did not correlate with absolute eosinophil count or disease activity. sSiglec-8 levels were measurable in sera from most donors unrelated to absolute eosinophil counts or Siglec-8 surface expression. c2E2 IgG1 and chimeric 2E2 IgG4 were equally effective at inducing cell death (Annexin-V positivity) of purified eosinophils from NDs and EOs after overnight IL-5 priming. In contrast, killing of purified eosinophils without IL-5 was only seen in EOs, and natural killer cell-mediated eosinophil killing was seen only with c2E2 IgG1. Finally, treatment of humanized mice with anti-Siglec antibody led to robust depletion of IL-5-induced eosinophilia in vivo. CONCLUSIONS: Siglec-8 is highly expressed on blood eosinophils from EOs and NDs and represents a potential therapeutic target for eosinophilic disorders. Enhanced killing of eosinophils in the presence of IL-5 might lead to increased efficacy in patients with IL-5-driven eosinophilia.


Assuntos
Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos B/metabolismo , Eosinofilia/imunologia , Eosinófilos/imunologia , Células Matadoras Naturais/imunologia , Lectinas/metabolismo , Animais , Anticorpos Bloqueadores/genética , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos de Diferenciação de Linfócitos B/genética , Antígenos de Diferenciação de Linfócitos B/imunologia , Morte Celular , Células Cultivadas , Citotoxicidade Imunológica , Eosinofilia/terapia , Humanos , Imunoglobulina G/genética , Interleucina-5/metabolismo , Lectinas/genética , Lectinas/imunologia , Contagem de Leucócitos , Camundongos , Camundongos SCID , Terapia de Alvo Molecular , Proteínas Recombinantes de Fusão/genética , Transcriptoma
6.
J Allergy Clin Immunol ; 138(3): 769-779, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27139822

RESUMO

BACKGROUND: Mast cells are a critical component of allergic responses in humans, and animal models that allow the in vivo investigation of their contribution to allergy and evaluation of new human-specific therapeutics are urgently needed. OBJECTIVE: To develop a new humanized mouse model that supports human mast cell engraftment and human IgE-dependent allergic responses. METHODS: This model is based on the NOD-scid IL2rg(null)SCF/GM-CSF/IL3 (NSG-SGM3) strain of mice engrafted with human thymus, liver, and hematopoietic stem cells (termed Bone marrow, Liver, Thymus [BLT]). RESULTS: Large numbers of human mast cells develop in NSG-SGM3 BLT mice and populate the immune system, peritoneal cavity, and peripheral tissues. The human mast cells in NSG-SGM3 BLT mice are phenotypically similar to primary human mast cells and express CD117, tryptase, and FcεRI. These mast cells undergo degranulation in an IgE-dependent and -independent manner, and can be readily cultured in vitro for additional studies. Intradermal priming of engrafted NSG-SGM3 mice with a chimeric IgE containing human constant regions resulted in the development of a robust passive cutaneous anaphylaxis response. Moreover, we describe the first report of a human mast cell antigen-dependent passive systemic anaphylaxis response in primed mice. CONCLUSIONS: NSG-SGM3 BLT mice provide a readily available source of human mast cells for investigation of mast cell biology and a preclinical model of passive cutaneous anaphylaxis and passive systemic anaphylaxis that can be used to investigate the pathogenesis of human allergic responses and to test new therapeutics before their advancement to the clinic.


Assuntos
Anafilaxia/imunologia , Modelos Animais de Doenças , Mastócitos/imunologia , Anafilaxia Cutânea Passiva/imunologia , Animais , Transplante de Células-Tronco Hematopoéticas , Humanos , Imunoglobulina E/imunologia , Transplante de Fígado , Camundongos , Timo/transplante
7.
Growth Factors ; 32(6): 223-35, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25413948

RESUMO

EphA3 is expressed in solid tumors and leukemias and is an attractive target for the therapy. We have generated a panel of Humaneered® antibodies to the ligand-binding domain using a Fab epitope-focused library that has the same specificity as monoclonal antibody mIIIA4. A high-affinity antibody was selected that competes with the mIIIA4 antibody for binding to EphA3 and has an improved affinity of ∼1 nM. In order to generate an antibody with potent cell-killing activity the variable regions were assembled with human IgG1k constant regions and expressed in a Chinese hamster ovary (CHO) cell line deficient in fucosyl transferase. Non-fucosylated antibodies have been reported to have enhanced binding affinity for the IgG receptor CD16a (FcγRIIIa). The affinity of the antibody for recombinant CD16a was enhanced approximately 10-fold. This resulted in enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) activity against EphA3-expressing leukemic cells, providing a potent antibody for the evaluation as a therapeutic agent.


Assuntos
Anticorpos Monoclonais/imunologia , Afinidade de Anticorpos , Citotoxicidade Celular Dependente de Anticorpos , Receptor EphA3/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/genética , Células CHO , Cricetinae , Cricetulus , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Macaca mulatta , Dados de Sequência Molecular , Receptores de IgG/imunologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
8.
J Allergy Clin Immunol ; 133(5): 1439-47, 1447.e1-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24530099

RESUMO

BACKGROUND: Although several novel agents are currently in clinical trials for eosinophilic disorders, none has demonstrated efficacy in reducing blood and tissue eosinophilia in all subjects. Additional approaches are clearly needed. OBJECTIVE: We sought to explore the potential of the human eosinophil surface receptor epidermal growth factor-like module containing mucin-like hormone receptor 1 (EMR1) as a therapeutic target for eosinophilic disorders. METHODS: EMR1 expression was assessed in blood and bone marrow specimens from eosinophilic and healthy subjects, cell lines, CD34(+) cells differentiated in vitro, and tissue biopsy specimens by using flow cytometry, quantitative PCR, and immunostaining. Eosinophil targeting by a novel, humanized, afucosylated anti-EMR1 IgG1 was evaluated in vitro by using a natural killer cell-mediated killing assay and in vivo in cynomolgus monkeys. RESULTS: Analysis of blood and bone marrow cells from healthy and eosinophilic donors and in vitro-differentiated CD34(+) cells confirmed restriction of human EMR1 surface and mRNA expression to mature eosinophils. Tissue eosinophils also expressed EMR1. Although EMR1 was highly expressed on eosinophils from all subjects, surface expression was negatively correlated with absolute eosinophil counts (r = -0.46, P < .001), and soluble plasma levels correlated positively with absolute eosinophil counts (r = 0.69, P < .001), suggesting modulation of EMR1 in vivo. Nevertheless, afucosylated anti-EMR1 mAb dramatically enhanced natural killer cell-mediated killing of eosinophils from healthy and eosinophilic donors and induced a rapid and sustained depletion of eosinophils in monkeys. CONCLUSION: EMR1 expression is restricted to mature blood and tissue eosinophils. Targeting of eosinophils with afucosylated anti-EMR1 antibody shows promise as a treatment for eosinophilic disorders.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Eosinofilia/tratamento farmacológico , Eosinófilos/imunologia , Regulação da Expressão Gênica/efeitos dos fármacos , Imunoglobulina G/farmacologia , Glicoproteínas de Membrana/imunologia , Mucinas/imunologia , Receptores Acoplados a Proteínas G/imunologia , Anticorpos Monoclonais Murinos/imunologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Proteínas de Ligação ao Cálcio , Eosinofilia/imunologia , Eosinofilia/patologia , Eosinófilos/patologia , Feminino , Humanos , Imunoglobulina G/imunologia , Células K562 , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Mucinas/antagonistas & inibidores , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Células U937
9.
Mol Biol Cell ; 20(15): 3552-60, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19477926

RESUMO

The Wnt coreceptor LRP6 is required for canonical Wnt signaling. To understand the molecular regulation of LRP6 function, we generated a series of monoclonal antibodies against the extra cellular domain (ECD) of LRP6 and selected a high-affinity mAb (mAb135) that recognizes cell surface expression of endogenous LRP6. mAb135 enhanced Wnt dependent TCF reporter activation and antagonized DKK1 dependent inhibition of Wnt3A signaling, suggesting a role in modulation of LRP6 function. Detailed analysis of LRP6 domain mutants identified Ser 243 in the first propeller domain of LRP6 as a critical residue for mAb135 binding, implicating this domain in regulating the sensitivity of LRP6 to DKK1. In agreement with this notion, mAb135 directly disrupted the interaction of DKK1 with recombinant ECD LRP6 and a truncated form of the LRP6 ECD containing only repeats 1 and 2. Finally, we found that mAb135 completely protected LRP6 from DKK1 dependent internalization. Together, these results identify the first propeller domain as a novel regulatory domain for DKK1 binding to LRP6 and show that mAb against the first propeller domain of LRP6 can be used to modulate this interaction.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Sítios de Ligação/genética , Sítios de Ligação/imunologia , Western Blotting , Linhagem Celular , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas Relacionadas a Receptor de LDL/genética , Proteínas Relacionadas a Receptor de LDL/imunologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Mutação , Ligação Proteica/efeitos dos fármacos , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt3 , Proteína Wnt3A
10.
Mol Biol Cell ; 19(6): 2588-96, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18400942

RESUMO

The R-Spondin (RSpo) family of secreted proteins is implicated in the activation of the Wnt signaling pathway. Despite the high structural homology between the four members, expression patterns and phenotypes in knockout mice have demonstrated striking differences. Here we dissected and compared the molecular and cellular function of all RSpo family members. Although all four RSpo proteins activate the canonical Wnt pathway, RSpo2 and 3 are more potent than RSpo1, whereas RSpo4 is relatively inactive. All RSpo members require Wnt ligands and LRP6 for activity and amplify signaling of Wnt3A, Wnt1, and Wnt7A, suggesting that RSpo proteins are general regulators of canonical Wnt signaling. Like RSpo1, RSpo2-4 antagonize DKK1 activity by interfering with DKK1 mediated LRP6 and Kremen association. Analysis of RSpo deletion mutants indicates that the cysteine-rich furin domains are sufficient and essential for the amplification of Wnt signaling and inhibition of DKK1, suggesting that Wnt amplification by RSpo proteins may be a direct consequence of DKK1 inhibition. Together, these findings indicate that RSpo proteins modulate the Wnt pathway by a common mechanism and suggest that coexpression with specific Wnt ligands and DKK1 may determine their biological specificity in vivo.


Assuntos
Transdução de Sinais , Trombospondinas/metabolismo , Proteínas Wnt/metabolismo , Animais , Linhagem Celular , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Relacionadas a Receptor de LDL/metabolismo , Ligantes , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Camundongos , Estrutura Terciária de Proteína , Receptores de Superfície Celular/metabolismo , Fatores de Transcrição TCF/metabolismo , Trombospondinas/química , beta Catenina/metabolismo
11.
Proc Natl Acad Sci U S A ; 104(37): 14700-5, 2007 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-17804805

RESUMO

The R-Spondin (RSpo) family of secreted proteins act as potent activators of the Wnt/beta-catenin signaling pathway. We have previously shown that RSpo proteins can induce proliferative effects on the gastrointestinal epithelium in mice. Here we provide a mechanism whereby RSpo1 regulates cellular responsiveness to Wnt ligands by modulating the cell-surface levels of the coreceptor LRP6. We show that RSpo1 activity critically depends on the presence of canonical Wnt ligands and LRP6. Although RSpo1 does not directly activate LRP6, it interferes with DKK1/Kremen-mediated internalization of LRP6 through an interaction with Kremen, resulting in increased LRP6 levels on the cell surface. Our results support a model in which RSpo1 relieves the inhibition DKK1 imposes on the Wnt pathway.


Assuntos
Proteínas Relacionadas a Receptor de LDL/antagonistas & inibidores , Transdução de Sinais , Trombospondinas/metabolismo , Proteínas Wnt/metabolismo , Animais , Linhagem Celular , Drosophila/citologia , Drosophila/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Rim/citologia , Proteínas Relacionadas a Receptor de LDL/metabolismo , Ligantes , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade , Luciferases/metabolismo , Proteínas de Membrana/metabolismo , Modelos Biológicos , Fosforilação , Testes de Precipitina , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/metabolismo , Trombospondinas/genética , Transfecção , beta Catenina/genética , beta Catenina/metabolismo
12.
Proc Natl Acad Sci U S A ; 104(29): 11933-8, 2007 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-17606906

RESUMO

Src kinase mediates growth factor signaling and causes oncogenic transformation, which includes dramatic changes in the actin cytoskeleton, cell shape, and motility. Cortactin was discovered as a substrate for Src. How phosphorylation of cortactin can enhance actin assembly is unknown. Here, using an actin assembly system reconstituted from purified components, we demonstrate for the first time a biochemical mechanism by which Src phosphorylation of cortactin affects actin assembly. The adaptor Nck is an important component of the system, linking phosphorylated cortactin with neuronal WASp (N-WASp) and WASp-interacting protein (WIP) to activate Arp2/3 complex.


Assuntos
Actinas/química , Actinas/metabolismo , Cortactina/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/metabolismo , Bovinos , Cortactina/química , Humanos , Proteínas Oncogênicas/metabolismo , Fosforilação , Ligação Proteica , Estrutura Quaternária de Proteína , Transporte Proteico , Suínos , Tirosina/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo
13.
Anal Biochem ; 364(1): 67-77, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17362870

RESUMO

A total of 22 individuals participated in this benchmark study to characterize the thermodynamics of small-molecule inhibitor-enzyme interactions using Biacore instruments. Participants were provided with reagents (the enzyme carbonic anhydrase II, which was immobilized onto the sensor surface, and four sulfonamide-based inhibitors) and were instructed to collect response data from 6 to 36 degrees C. van't Hoff enthalpies and entropies were calculated from the temperature dependence of the binding constants. The equilibrium dissociation and thermodynamic constants determined from the Biacore analysis matched the values determined using isothermal titration calorimetry. These results demonstrate that immobilization of the enzyme onto the sensor surface did not alter the thermodynamics of these interactions. This benchmark study also provides insights into the opportunities and challenges in carrying out thermodynamic studies using optical biosensors.


Assuntos
Técnicas Biossensoriais/instrumentação , Calorimetria/instrumentação , Calorimetria/normas , Inibidores da Anidrase Carbônica/classificação , Inibidores da Anidrase Carbônica/metabolismo , Sulfonamidas/antagonistas & inibidores , Benchmarking , Pesquisa Biomédica , Técnicas Biossensoriais/normas , Anidrase Carbônica II/química , Anidrase Carbônica II/metabolismo , Variações Dependentes do Observador , Ligação Proteica , Sulfonamidas/classificação , Ressonância de Plasmônio de Superfície/instrumentação , Ressonância de Plasmônio de Superfície/normas , Termodinâmica
14.
Biochemistry ; 46(11): 3494-502, 2007 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-17302440

RESUMO

The Wiskott-Aldrich syndrome protein (WASP) and neural WASP (N-WASP) are key players in regulating actin cytoskeleton via the Arp2/3 complex. It has been widely reported that the WASP proteins are activated by Rho family small GTPase Cdc42 and that Rac1 acts through SCAR/WAVE proteins. However, a systematic study of the specificity of different GTPases for different Arp2/3 activators has not been conducted. In this study, we have expressed, purified, and characterized completely soluble, highly active, and autoinhibited full-length human WASP and N-WASP from mammalian cells. We show a novel N-WASP activation by Rho family small GTPase Rac1. This GTPase exclusively stimulates N-WASP and has no effects on WASP. Rac1 is a significantly more potent N-WASP activator than Cdc42. In contrast, Cdc42 is a more effective activator of WASP than N-WASP. Lipid vesicles containing PIP2 significantly improve actin nucleation by the Arp2/3 complex and N-WASP in the presence of Rac1 or Cdc42. PIP2 vesicles have no effect on WASP activity alone. Moreover, the inhibition of WASP-stimulated actin nucleation in the presence of Cdc42 and PIP2 vesicles has been observed. We found that adaptor proteins Nck1 or Nck2 are the most potent WASP and N-WASP activators with distinct effects on the WASP family members. Our in vitro data demonstrates differential regulation of full-length WASP and N-WASP by cellular activators that highlights fundamental differences of response at the protein-protein level.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/fisiologia , Actinas/metabolismo , Proteínas Oncogênicas/fisiologia , Fosfatidilinositol 4,5-Difosfato/fisiologia , Proteína Neuronal da Síndrome de Wiskott-Aldrich/fisiologia , Proteína da Síndrome de Wiskott-Aldrich/fisiologia , Proteína cdc42 de Ligação ao GTP/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Humanos , Proteínas Recombinantes/biossíntese , Transdução de Sinais
15.
Mol Cell ; 13(6): 817-28, 2004 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-15053875

RESUMO

U8 snoRNP is required for accumulation of mature 5.8S and 28S rRNA in vertebrates. We are identifying proteins that bind U8 RNA with high specificity to understand how U8 functions in ribosome biogenesis. Here, we characterize a Xenopus 29 kDa protein (X29), which we previously showed binds U8 RNA with high affinity. X29 and putative homologs in other vertebrates contain a NUDIX domain found in MutT and other nucleotide diphosphatases. Recombinant X29 protein has diphosphatase activity that removes m(7)G and m(227)G caps from U8 and other RNAs in vitro; the putative 29 kDa human homolog also displays this decapping activity. X29 is primarily nucleolar in Xenopus tissue culture cells. We propose that X29 is a member of a conserved family of nuclear decapping proteins that function in regulating the level of U8 snoRNA and other nuclear RNAs with methylated caps.


Assuntos
Núcleo Celular/enzimologia , Endorribonucleases/metabolismo , RNA Nucleolar Pequeno/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus/metabolismo , Animais , Sequência de Bases , Sítios de Ligação , Nucléolo Celular/enzimologia , Células Cultivadas , Clonagem Molecular , Sequência Consenso , Dados de Sequência Molecular , Peso Molecular , Estrutura Terciária de Proteína , RNA Nucleolar Pequeno/química , RNA Nucleolar Pequeno/genética , Proteínas Recombinantes/metabolismo , Ribonucleoproteínas Nucleolares Pequenas/metabolismo , Xenopus/genética , Proteínas de Xenopus/química
16.
J Cell Sci ; 117(Pt 2): 177-87, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14657279

RESUMO

Targeted disruption of the focal adhesion kinase (FAK) gene in mice is lethal at embryonic day 8.5 (E8.5). Vascular defects in FAK-/- mice result from the inability of FAK-deficient endothelial cells to organize themselves into vascular network. We found that, although fibronectin (FN) levels were similar, its organization was less fibrillar in both FAK-/- endothelial cells and mesoderm of E8.5 FAK-/- embryos, as well as in mouse embryonic fibroblasts isolated from mutant embryos. FAK catalytic activity, proline-rich domains, and location in focal contacts were all required for proper allocation and patterning of FN matrix. Cells lacking FAK in focal adhesions fail to translocate supramolecular complexes of integrin-bound FN and focal adhesion proteins along actin filaments to form mature fibrillar adhesions. Taken together, our data suggest that proper FN allocation and organization are dependent on FAK-mediated remodeling of focal adhesions.


Assuntos
Fibronectinas/metabolismo , Adesões Focais/metabolismo , Proteínas Tirosina Quinases/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Camundongos , Camundongos Knockout , Fosforilação , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/fisiologia
17.
Mol Cell Biol ; 22(12): 4101-12, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12024024

RESUMO

U8 snoRNA plays a unique role in ribosome biogenesis: it is the only snoRNA essential for maturation of the large ribosomal subunit RNAs, 5.8S and 28S. To learn the mechanisms behind the in vivo role of U8 snoRNA, we have purified to near homogeneity and characterized a set of proteins responsible for the formation of a specific U8 RNA-binding complex. This 75-kDa complex is stable in the absence of added RNA and binds U8 with high specificity, requiring the conserved octamer sequence present in all U8 homologues. At least two proteins in this complex can be cross-linked directly to U8 RNA. We have identified the proteins as Xenopus homologues of the LSm (like Sm) proteins, which were previously reported to be involved in cytoplasmic degradation of mRNA and nuclear stabilization of U6 snRNA. We have identified LSm2, -3, -4, -6, -7, and -8 in our purified complex and found that this complex associates with U8 RNA in vivo. This purified complex can bind U6 snRNA in vitro but does not bind U3 or U14 snoRNA in vitro, demonstrating that the LSm complex specifically recognizes U8 RNA.


Assuntos
RNA Nuclear Pequeno/metabolismo , RNA Nucleolar Pequeno/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/metabolismo , Xenopus/genética , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células Cultivadas , Sequência Conservada , Reagentes de Ligações Cruzadas/química , Evolução Molecular , Feminino , Dados de Sequência Molecular , Acetiltransferase N-Terminal C , Oócitos , RNA Nuclear Pequeno/química , RNA Nucleolar Pequeno/química , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteína Nuclear Pequena U4-U6/imunologia , Ribonucleoproteína Nuclear Pequena U4-U6/isolamento & purificação , Ribonucleoproteínas Nucleares Pequenas , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...