Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Haematologica ; 105(12): 2813-2823, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33256380

RESUMO

The mechanisms of drug resistance in multiple myeloma are poorly understood. Here we show that CD47, an integrin-associated receptor, is significantly upregulated in drug resistant myeloma cells in comparison with parental cells, and that high expression of CD47 detected by immunohistochemistry is associated with shorter progression free and overall survivals in multiple myeloma patients. We show that miR-155 is expressed at low levels in drug resistant myeloma cells and is a direct regulator of CD47 through its 3'UTR. Furthermore, low miR-155 levels are associated with advanced stages of disease. MiR-155 overexpression suppressed CD47 expression on myeloma cell surface, leading to induction of phagocytosis of myeloma cells by macrophages and inhibition of tumor growth. MiR-155 overexpression also re-sensitized drug-resistant myeloma cells to bortezomib leading to cell death through targeting TNFAIP8, a negative mediator of apoptosis in vitro and in vivo. Thus, miR-155 mimics may serve as a promising new therapeutic modality by promoting phagocytosis and inducing apoptosis in patients with refractory/relapsed multiple myeloma.


Assuntos
MicroRNAs , Mieloma Múltiplo , Apoptose , Proteínas Reguladoras de Apoptose , Antígeno CD47/genética , Linhagem Celular Tumoral , Resistência a Medicamentos , Humanos , MicroRNAs/genética , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Fagocitose
2.
BMC Cancer ; 19(1): 975, 2019 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-31638931

RESUMO

BACKGROUND: Multiple myeloma (MM) cells gain protection against drugs through interaction with bone marrow stromal cells (BMSCs). This form of resistance largely accounts for resistance to therapy in MM patients which warrants further exploration to identify more potential therapeutic targets. METHODS: We performed miRNA/mRNA qPCR arrays and western blotting to analyze transcriptional and translational changes in MM cells co-cultured with BMSCs. Drug cytotoxicity and apoptosis in MMGFP-BMSC co-cultures were measured using fluorescence plate reader and flowcytometry, respectively. miRNA was overexpressed in MM cell lines using Lentiviral transduction, miRNA-3'UTR binding was examined using luciferase assay. RESULTS: We found that BMSCs downregulated miR-101-3p and upregulated survivin (BIRC5) in MM cells. Survivin was downregulated by miR-101-3p overexpression and found to be a direct target of miR-101-3p using 3'UTR luciferase assay. Overexpression of survivin increased viability of MM cells in the presence of anti-myeloma drugs, and miR-101-3p inhibition by anti-miR against miR-101-3p upregulated survivin. Furthermore, overexpression of miR-101-3p or silencing of survivin triggered apoptosis in MM cells and sensitized them to anti-myeloma drugs in the presence of BMSCs overcoming the stroma-induced drug resistance. CONCLUSIONS: Our study demonstrates that BMSC-induced resistance to drugs is associated with survivin upregulation which is a direct target of miR-101-3p. This study also identifies miR-101-3p-survivin interaction as a druggable target involved in stroma-mediated drug resistance in MM and suggests it for developing more efficient therapeutic strategies.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Expressão Ectópica do Gene/genética , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Survivina/genética , Regiões 3' não Traduzidas/genética , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Bortezomib/farmacologia , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , MicroRNAs/antagonistas & inibidores , Mieloma Múltiplo/patologia , Transfecção
3.
Mol Cancer Ther ; 21(7): 1125-1135, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35499387

RESUMO

Luxeptinib (CG-806) simultaneously targets FLT3 and select other kinase pathways operative in myeloid malignancies. We investigated the range of kinases it inhibits, its cytotoxicity landscape ex vivo with acute myeloid leukemia (AML) patient samples, and its efficacy in xenograft models. Luxeptinib inhibits wild-type (WT) and many of the clinically relevant mutant forms of FLT3 at low nanomolar concentrations. It is a more potent inhibitor of the activity of FLT3-internal tandem duplication, FLT3 kinase domain and gatekeeper mutants than against WT FLT3. Broad kinase screens disclosed that it also inhibits other kinases that can drive oncogenic signaling and rescue pathways, but spares kinases known to be associated with clinical toxicity. In vitro profiling of luxeptinib against 186 AML fresh patient samples demonstrated greater potency relative to other FLT3 inhibitors, including cases with mutations in FLT3, isocitrate dehydrogenase-1/2, ASXL1, NPM1, SRSF2, TP53, or RAS, and activity was documented in a xenograft AML model. Luxeptinib administered continuously orally every 12 hours at a dose that yielded a mean Cmin plasma concentration of 1.0 ± 0.3 µmol/L (SEM) demonstrated strong antitumor activity but no myelosuppression or evidence of tissue damage in mice or dogs in acute toxicology studies. On the basis of these studies, luxeptinib was advanced into a phase I trial for patients with AML and myelodysplastic/myeloproliferative neoplasms.


Assuntos
Leucemia Mieloide Aguda , Animais , Cães , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Camundongos , Mutação , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
4.
Biomark Res ; 9(1): 34, 2021 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-33958003

RESUMO

The myristoylated alanine-rich C-kinase substrate (MARCKS) protein has been at the crossroads of multiple signaling pathways that govern several critical operations in normal and malignant cellular physiology. Functioning as a target of protein kinase C, MARCKS shuttles between the phosphorylated cytosolic form and the unphosphorylated plasma membrane-bound states whilst regulating several molecular partners including, but not limited to calmodulin, actin, phosphatidylinositol-4,5-bisphosphate, and phosphoinositide-3-kinase. As a result of these interactions, MARCKS directly or indirectly modulates a host of cellular functions, primarily including cytoskeletal reorganization, membrane trafficking, cell secretion, inflammatory response, cell migration, and mitosis. Recent evidence indicates that dysregulated expression of MARCKS is associated with the development and progression of hematological cancers. While it is understood that MARCKS impacts the overall carcinogenesis as well as plays a part in determining the disease outcome in blood cancers, we are still at an early stage of interpreting the pathophysiological roles of MARCKS in neoplastic disease. The situation is further complicated by contradictory reports regarding the role of phosphorylated versus an unphosphorylated form of MARCKS as an oncogene versus tumor suppressor in blood cancers. In this review, we will investigate the current body of knowledge and evolving concepts of the physical properties, molecular network, functional attributes, and the likely pathogenic roles of MARCKS in hematological malignancies. Key emphasis will also be laid upon understanding the novel mechanisms by which MARCKS determines the overall disease prognosis by playing a vital role in the induction of therapeutic resistance. Additionally, we will highlight the importance of MARCKS as a valuable therapeutic target in blood cancers and will discuss the potential of existing strategies available to tackle MARCKS-driven blood cancers.

5.
J Hematol Oncol ; 13(1): 96, 2020 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-32677994

RESUMO

CD47, or integrin-associated protein, is a cell surface ligand expressed in low levels by nearly all cells of the body. It plays an integral role in various immune responses as well as autoimmunity, by sending a potent "don't eat me" signal to prevent phagocytosis. A growing body of evidence demonstrates that CD47 is overexpressed in various hematological malignancies and its interaction with SIRPα on the phagocytic cells prevents phagocytosis of cancer cells. Additionally, it is expressed by different cell types in the tumor microenvironment and is required for establishing tumor metastasis. Overexpression of CD47 is thus often associated with poor clinical outcomes. CD47 has emerged as a potential therapeutic target and is being investigated in various preclinical studies as well as clinical trials to prove its safety and efficacy in treating hematological neoplasms. This review focuses on different therapeutic mechanisms to target CD47, either alone or in combination with other cell surface markers, and its pivotal role in impairing tumor growth and metastatic spread of various types of hematological malignancies.


Assuntos
Antígeno CD47/fisiologia , Neoplasias Hematológicas/fisiopatologia , Terapia de Alvo Molecular , Proteínas de Neoplasias/fisiologia , Proteínas Angiogênicas/metabolismo , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Antígenos de Diferenciação/metabolismo , Antineoplásicos Imunológicos/uso terapêutico , Antígeno CD47/antagonistas & inibidores , Ensaios Clínicos como Assunto , Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Neoplasias Hematológicas/terapia , Humanos , Integrinas/metabolismo , Leucemia/metabolismo , Leucemia/fisiopatologia , Linfoma não Hodgkin/metabolismo , Linfoma não Hodgkin/fisiopatologia , Mimetismo Molecular , Células Mieloides/metabolismo , Metástase Neoplásica , Proteínas de Neoplasias/antagonistas & inibidores , Oligopeptídeos/uso terapêutico , Ligação Proteica , Domínios Proteicos , Mapeamento de Interação de Proteínas , Receptores Imunológicos/antagonistas & inibidores , Receptores Imunológicos/metabolismo , Transdução de Sinais/fisiologia
6.
Cancer Lett ; 480: 29-38, 2020 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-32220540

RESUMO

Overexpression of Myristoylated Alanine-Rich C Kinase Substrate (MARCKS) is implicated in drug resistance and progression of multiple myeloma (MM). The basis for MARCKS induction and impact on MM are not known. Here we show that microRNA-34a (miR-34a), regulates MARCKS translation and is under-expressed in drug-resistant MM cells, leading to increased MARCKS protein level. Over-expression of miR-34a reduces MARCKS expression and sensitizes resistant cells to anti-myeloma drugs. A MARCKS peptide inhibitor (MPS) exerts a dose dependent cytotoxic effect on drug-resistant MM cells with minimal cytotoxicity to normal hematopoietic cells. MPS synergizes with the proteasomal-inhibitor bortezomib to effectively kill drug-resistant MM cells both in vitro and in a xenograft model of MM. While MARCKS inhibition killed MM cells, it also enhanced a pro-survival autophagic pathway that sustained growth following MARCKS inhibition. In accordance, combined treatment with MARCKS antagonists, bortezomib and the autophagy inhibitor, chloroquine, significantly diminished tumor growth in drug-resistant MM cell lines as well as primary MM cells. This study uncovers a mechanism of drug resistance involving miR-34a-MARCKS autoregulatory loop and provides a framework for a potentially new therapeutic strategy to overcome drug resistance in multiple myeloma.


Assuntos
Autofagia/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Substrato Quinase C Rico em Alanina Miristoilada/antagonistas & inibidores , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Bortezomib/administração & dosagem , Bortezomib/farmacologia , Linhagem Celular Tumoral , Cloroquina/administração & dosagem , Cloroquina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos SCID , MicroRNAs/genética , Mieloma Múltiplo/patologia , Substrato Quinase C Rico em Alanina Miristoilada/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cytokine ; 46(1): 43-50, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19278866

RESUMO

The use of cytokines as adjuvants has been shown to be a promising approach for enhancing DNA vaccine induced-immune responses. In this report, we investigate the administration of cytokines to modulate both humoral and cell-mediated immune responses elicited by an HCV-core plasmid DNA vaccine in Balb/c mice. Our studies indicate that the HCV-core DNA vaccine has been able to induce both antibody and cellular immunity in a DNA prime-protein boost regimen. GM-CSF (granulocyte-monocyte colony stimulating factor) which is considered to be a cytokine displaying both Th1 and Th2 characteristics, and plays an important role in augmenting antibody and cell-mediated immunity was also administered. The induction of cellular immunity was not as striking as humoral immunity in this case. To obtain a stronger cellular response, IL-23, a Th1 cytokine belonging to the IL-12 family, was also included in the regimen. Spleen cell proliferation, IFN-gamma production from spleen cells and specific serum IgG2a, all demonstrate the enhancement of cell-mediated immunity without any observable suppressive effect on antibody and humoral immune responses. We also examined the timing of plasmid IL-23 administration on the phenotype of the resultant T cell responses in a 3 day interval, before and after plasmid GM-CSF administration. The results did not indicate any change in the Th1/Th2 balance as compared with simultaneous IL-23 administration.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Hepacivirus/metabolismo , Interleucina-23/metabolismo , Vacinas de DNA/metabolismo , Adjuvantes Imunológicos/metabolismo , Animais , Linhagem Celular , Citocinas/metabolismo , Feminino , Humanos , Sistema Imunitário , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Recombinantes/química , Células Th1/citologia , Células Th1/metabolismo
8.
J Hematol Oncol ; 11(1): 46, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29580288

RESUMO

Even with recent advances in therapy regimen, multiple myeloma patients commonly develop drug resistance and relapse. The relevance of targeting the PD-1/PD-L1 axis has been demonstrated in pre-clinical models. Monotherapy with PD-1 inhibitors produced disappointing results, but combinations with other drugs used in the treatment of multiple myeloma seemed promising, and clinical trials are ongoing. However, there have recently been concerns about the safety of PD-1 and PD-L1 inhibitors combined with immunomodulators in the treatment of multiple myeloma, and several trials have been suspended. There is therefore a need for alternative combinations of drugs or different approaches to target this pathway. Protein expression of PD-L1 on cancer cells, including in multiple myeloma, has been associated with intrinsic aggressive features independent of immune evasion mechanisms, thereby providing a rationale for the adoption of new strategies directly targeting PD-L1 protein expression. Drugs modulating the transcriptional and post-transcriptional regulation of PD-L1 could represent new therapeutic strategies for the treatment of multiple myeloma, help potentiate the action of other drugs or be combined to PD-1/PD-L1 inhibitors in order to avoid the potentially problematic combination with immunomodulators. This review will focus on the pathophysiology of PD-L1 expression in multiple myeloma and drugs that have been shown to modulate this expression.


Assuntos
Mieloma Múltiplo/tratamento farmacológico , Receptor de Morte Celular Programada 1/metabolismo , Humanos
9.
Biomark Res ; 6: 34, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555699

RESUMO

Enhancer of zeste homolog 2 (EZH2) is a histone methyltransferase that is of great interest in human cancer. It has been shown to have a dual nature, as it can act as a gene repressor or activator. Studies have highlighted the various roles of EZH2 in the pathophysiology of multiple myeloma (MM). It was also shown to have a role in the development of drug resistance in MM. There are several ongoing clinical trials of EZH2 inhibitors in haematological malignancies. Pre-clinical studies have provided a rationale for the therapeutic relevance of EZH2 inhibitors in MM. This paper reviews the evidence supporting the role of EZH2 in MM pathophysiology and drug resistance, with an emphasis on interactions between EZH2 and microRNAs, as well as the prognostic significance of EZH2 expression in MM. Furthermore, results from the pre-clinical studies of EZH2 inhibition in MM and currently available interim results from clinical trials of EZH2 inhibitors in haematological malignancies are presented. Preliminary data exploring anticipated mechanisms of resistance to EZH2 inhibitors are also reviewed. There is therefore strong evidence to support the relevance of targeting EZH2 for the treatment of MM.

10.
Leukemia ; 32(11): 2471-2482, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29743723

RESUMO

EZH2 is highly expressed in multiple myeloma (MM). However, the molecular mechanisms underlying EZH2 overexpression and its role in drug resistance of MM remain undefined. Here we show that EZH2 is upregulated in drug-resistant MM cells and its aberrant overexpression is associated with poor prognosis of MM patients. Overexpression of EZH2 in parental MM cells renders them resistant to anti-myeloma drugs and suppression of EZH2 displays the opposite effects. Using miRNA target scan algorithms, we identify miR-138 as a regulator of EZH2, which is conversely repressed by EZH2-induced H3K27 trimethylation in MM-resistant cell lines and primary tumor cells. Analysis of ChIP-seq dataset and H3K27me3 ChIP reveals that RBPMS is a direct and functionally relevant target of EZH2. RBPMS silencing confers resistance to MM cells and restoration of RBPMS by miR-138 overexpression re-sensitizes the resistant cells to drug. Importantly, in vivo delivery of miR-138 mimics or pharmacological inhibitor of EZH2 in combination with a proteasome inhibitor, bortezomib, induces significant regression of tumors in xenograft model. This study establishes EZH2/miR-138 axis as a potential therapeutic target for MM.


Assuntos
Regulação para Baixo/genética , Resistência a Medicamentos/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , MicroRNAs/genética , Mieloma Múltiplo/genética , Proteínas de Ligação a RNA/genética , Bortezomib/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Humanos , Mieloma Múltiplo/tratamento farmacológico , Inibidores de Proteassoma/farmacologia
11.
J Hematol Oncol ; 10(1): 121, 2017 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-28623912

RESUMO

Despite administration of novel therapies, multiple myeloma (MM) remains incurable with resistance to drugs leading to relapse in most patients. Thus, it is critical to understand the detailed mechanisms underlying the drug resistance of MM and develop more effective therapeutic strategies. Genetic abnormalities are well known to play a central role in MM pathogenesis and therapy resistance; however, epigenetic aberrations mainly affecting the patterns of DNA methylation/histone modifications of genes (especially tumor suppressors) and miRNAs have also been shown to be involved. Importantly, while epigenetic silencing of miRNAs in MM is well documented, some epigenetic markers are known to be direct targets of miRNAs particularly the recently described "epimiRNAs". Drugs targeting epigenetic modifiers (e.g., HDACs, EZH2) can sensitize MM-resistant cells to anti-myeloma drugs and reversibility of epigenetic changes makes these drugs promising therapeutic agents. Therefore, combination of miRNA mimics with inhibitors of epigenetic modifiers would be a more potent therapeutic strategy in MM patients in relapse or refractory to treatments. In this review, we will discuss the findings of recent investigations on epigenetics/miRNA regulatory axis in development of drug resistance in MM and highlight possible approaches for therapeutic applications of such interaction.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Epigênese Genética/efeitos dos fármacos , MicroRNAs/genética , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Animais , Antineoplásicos/uso terapêutico , Metilação de DNA/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular/métodos
12.
J Hematol Oncol ; 10(1): 169, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29073933

RESUMO

The molecular mechanisms underlying dysregulated wild type (wt) p53 in multiple myeloma (MM) have been subjects of intense investigation for years. Indeed, correlation of rarely occurring TP53 gene mutations or deletions with adverse clinical outcomes in MM patients is strongly established, while in majority of cases wtp53 seems to be non-functional or dysregulated bearing a high clinical impact. Interestingly, findings from recent investigations show that micro-RNAs (miRNAs) may contribute to suppression of wtp53 in MM, as they are now known to function as key regulatory elements in the p53 network. This area is shedding new light on understanding the biologic effects of dysregulated p53 in MM pathogenesis especially drug resistance. miRNAs such as miR-125b (oncomiR) or miR-34a (tumor suppressor-miR) can be negative or positive regulators of wtp53 function, respectively, with specific effects on MM cell viability. On the other hand, our knowledge of miRNA interaction with mutant (mt) p53 in MM, which is rather related to disease progression and resistance to therapy, is limited which demands in-depth exploration. Here, we will put forward the current knowledge on miRNA-p53 interaction in MM and its role in MM pathogenesis including drug resistance. We will also highlight the pre-clinical approaches for therapeutic application of miRNAs targeting p53 pathway.


Assuntos
MicroRNAs/metabolismo , Mieloma Múltiplo/genética , Proteína Supressora de Tumor p53/metabolismo , Humanos , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia
13.
Hybridoma (Larchmt) ; 30(3): 229-38, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21707357

RESUMO

According to World Health Organization (WHO), cancer is a leading cause of death worldwide, accounting for 7.4 million deaths (around 13% of all deaths) in 2004. Monoclonal/recombinant antibodies, which specifically target clinical biomarkers of disease, have increasingly been applied as powerful tools in cancer imaging and therapy, a fact that is highlighted by some nine FDA-approved monoclonal antibodies (MAbs) or their immunoconjugates (as of December 2008) for use in cancer treatment. In this study, five monoclonal antibodies (MAbs) were generated and characterized against carcinoembryonic antigen (CEA), which is widely used clinically as both a blood and tissue tumor marker of epithelial malignancy. Variable domains (VH and VL) of one the stable MAbs with highest affinity were PCR-amplified and assembled as single-chain antibody fragment (scFv). Following the cloning and expression of scFv antibody fragments in Escherichia coli, the functional binding and specificity of the recombinant antibody were confirmed by ELISA. To develop a direct in vitro detection of CEA-positive cancer cells, scFv DNA was genetically fused to enhanced green fluorescent protein (EGFP) gene and expressed in bacteria. The chimeric fluorescent protein is able to specifically detect CEA-positive cell lines; no cross-reactivity was observed with a negative control cell line. This strategy will likely allow the establishment of a rapid, single-step detection assay of CEA, which is considered to be one of the best predictors of malignancy among all other tumor markers.


Assuntos
Biomarcadores Tumorais/metabolismo , Antígeno Carcinoembrionário/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Imunoterapia/métodos , Neoplasias/diagnóstico , Neoplasias/terapia , Anticorpos de Cadeia Única , Sequência de Aminoácidos , Animais , Sequência de Bases , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/isolamento & purificação , Antígeno Carcinoembrionário/imunologia , Antígeno Carcinoembrionário/isolamento & purificação , Clonagem Molecular , DNA Complementar/biossíntese , Ensaio de Imunoadsorção Enzimática , Escherichia coli , Proteínas de Fluorescência Verde/imunologia , Humanos , Hibridomas/imunologia , Hibridomas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Análise de Sequência de DNA , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/metabolismo , Espectrometria de Fluorescência
14.
J Biotechnol ; 144(4): 245-52, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19770007

RESUMO

Three-dimensional structure and alignment analyses of 3'-5' exonuclease domain of DNA polymerase I from thermophilic Geobacillus sp. MKK show that the key catalytic amino acids in 3'-5' exonuclease domain are changed and the enzyme looses the activity. In order to render the activity, a catalytic module is constructed in the active site using site-directed mutagenesis. Seven mutant clones of the enzyme are generated containing: M1 (V319D, E325L), M2 (A376D), M3 (D425F), M4 (InsY446, K450D), M12 (V319D, E325L, A376D), M123 (V319D, E325L, A376D, D425F), and M1234 (V319D, E325L, A376D, D425F, InsY446, K450D). In addition, a chimera MkkEc polymerase is constructed by exchanging 3'-5' exonuclease domain of the MKK polymerase (residues 301-466) with the same domain of homologous Escherichia coli polymerase (residues 327-519). For the first time, all essential amino acids for the 3'-5' exonuclease activity are introduced in one mutant. As a result, among all mutants, only M1234 and MkkEc mutants show significant 3'-5' exonuclease activity. Moreover, M1234 mutant was kept most of its polymerase activity while the activity of MkkEc mutants is decreased dramatically compared to the wild type enzyme.


Assuntos
Domínio Catalítico/genética , DNA Polimerase I/metabolismo , Exodesoxirribonucleases , Geobacillus/enzimologia , Mutagênese Sítio-Dirigida , Clonagem Molecular , DNA Polimerase I/genética , Ativação Enzimática , Escherichia coli/genética , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Geobacillus/genética , Geobacillus/isolamento & purificação , Fontes Termais/microbiologia , Irã (Geográfico) , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade
15.
Hybridoma (Larchmt) ; 28(5): 305-13, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19857111

RESUMO

Beet necrotic yellow vein virus (BNYVV) infects sugar beet plants worldwide and is responsible for the rhizomania disease and severe economic losses. Disease severity and lack of naturally occurring resistant plants make it very difficult to control the virus, both from epidemiological and economic standpoints. Therefore, early detection is vital to impose hygiene restrictions and prevent further spread of the virus in the field. Immunoassays are one of the most popular methodologies for the primary identification of plant pathogens including BNYVV since they are robust, sensitive, fast, and inexpensive. In this study, the major coat protein (CP21) of BNYVV was cloned and expressed in Escherichia coli. Thereafter, mice were immunized with purified CP21 and a phage antibody library was constructed from their PCR-amplified immunoglobulin repertoire. Following filamentous phage rescue of the library and four rounds of panning against recombinant CP21 antigen, several specific single chain Fv fragments were isolated and characterized. This approach may pave the way to develop novel immunoassays for a rapid detection of viral infection. Moreover, it will likely provide essential tools to establish antibody-mediated resistant transgenic technology in sugar beet plants.


Assuntos
Bacteriófagos/imunologia , Proteínas do Capsídeo/imunologia , Fragmentos de Imunoglobulinas/imunologia , Vírus de Plantas/imunologia , Vírus de RNA/imunologia , Sequência de Aminoácidos , Animais , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Ensaio de Imunoadsorção Enzimática , Fragmentos de Imunoglobulinas/química , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Plantas Geneticamente Modificadas , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA