Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 9(2): 1641-1655, 2018 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-29416720

RESUMO

Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (EGFR-TKIs) were demonstrated to provide survival benefit in patients with non-small cell lung cancer (NSCLC) harboring activating mutations of EGFR; however, emergence of acquired resistance to EGFR-TKIs has been shown to cause poor outcome. To overcome the TKI resistance, drugs with different mode of action are required. We previously reported that M-COPA (2-methylcoprophilinamide), a Golgi disruptor, suppressed the growth of gastric cancers overexpressing receptor tyrosine kinases (RTKs) such as hepatocyte growth factor receptor (MET) via downregulating their cell surface expression. In this study, we examined the antitumor effect of M-COPA on NSCLC cells with TKI resistance. As a result, M-COPA effectively downregulated cell surface EGFR and its downstream signals, and finally exerted in vivo antitumor effect in NSCLC cells harboring secondary (T790M/del19) and tertiary (C797S/T790M/del19) mutated EGFR, which exhibit acquired resistance to first- and third generation EGFR-TKIs, respectively. M-COPA also downregulated MET expression potentially involved in the acquired resistance to EGFR-TKIs via bypassing the EGFR pathway blockade. These results provide the first evidence that targeting the Golgi apparatus might be a promising therapeutic strategy to overcome the vicious cycle of TKI resistance in EGFR-mutated NSCLC cells via downregulating cell surface RTK expression.

2.
Biochemistry ; 56(38): 5125-5133, 2017 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-28858527

RESUMO

Arf GTPases and their guanine nucleotide exchange factors (ArfGEFs) are major regulators of membrane traffic and organelle structure in cells. They are associated with a variety of diseases and are thus attractive therapeutic targets for inhibition by small molecules. Several inhibitors of unrelated chemical structures have been discovered, which have shown their potential in dissecting molecular pathways and blocking disease-related functions. However, their specificity across the ArfGEF family has remained elusive. Importantly, inhibitory responses in the context of membranes, which are critical determinants of Arf and ArfGEF cellular functions, have not been investigated. Here, we compare the efficiency and specificity of four structurally distinct ArfGEF inhibitors, Brefeldin A, SecinH3, M-COPA, and NAV-2729, toward six ArfGEFs (human ARNO, EFA6, BIG1, and BRAG2 and Legionella and Rickettsia RalF). Inhibition was assessed by fluorescence kinetics using pure proteins, and its modulation by membranes was determined with lipidated GTPases in the presence of liposomes. Our analysis shows that despite the intra-ArfGEF family resemblance, each inhibitor has a specific inhibitory profile. Notably, M-COPA is a potent pan-ArfGEF inhibitor, and NAV-2729 inhibits all GEFs, the strongest effects being against BRAG2 and Arf1. Furthermore, the presence of the membrane-binding domain in Legionella RalF reveals a strong inhibitory effect of BFA that is not measured on its GEF domain alone. This study demonstrates the value of family-wide assays with incorporation of membranes, and it should enable accurate dissection of Arf pathways by these inhibitors to best guide their use and development as therapeutic agents.


Assuntos
Brefeldina A/farmacologia , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Naftóis/farmacologia , Pirazóis/farmacologia , Piridinas/farmacologia , Pirimidinonas/farmacologia , Triazóis/farmacologia , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/antagonistas & inibidores , Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/metabolismo , Membrana Celular , Clorobenzenos , Fluorescência , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Lipossomos/química , Soluções
3.
Genes Cells ; 21(8): 901-6, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27302278

RESUMO

Shiga toxin (Stx) is a main virulence factor of Stx-producing Escherichia coli (STEC) that contributes to diarrhea and hemorrhagic colitis and occasionally to fatal systemic complications. Therefore, the development of an antidote to neutralize Stx toxicity is urgently needed. After internalization into cells, Stx is transferred to the Golgi apparatus via a retrograde vesicular transport system. We report here that 2-methylcoprophilinamide (M-COPA), a compound that induces disassembly of the Golgi apparatus by inactivating ADP-ribosylation factor 1 (Arf1), suppresses Stx-induced apoptosis. M-COPA inhibited transport of Stx from the plasma membrane to the Golgi apparatus and suppressed degradation of anti-apoptotic proteins and the activation of caspases. These findings suggest that inhibition of Stx retrograde transport by M-COPA could be a novel approach to suppress Stx toxicity.


Assuntos
Fator 1 de Ribosilação do ADP/genética , Alcenos/farmacologia , Antídotos/farmacologia , Naftóis/administração & dosagem , Piridinas/administração & dosagem , Toxina Shiga/antagonistas & inibidores , Escherichia coli Shiga Toxigênica/efeitos dos fármacos , Fator 1 de Ribosilação do ADP/antagonistas & inibidores , Alcenos/química , Antídotos/química , Apoptose/efeitos dos fármacos , Apoptose/genética , Colite/tratamento farmacológico , Colite/microbiologia , Diarreia/tratamento farmacológico , Diarreia/microbiologia , Complexo de Golgi/efeitos dos fármacos , Humanos , Toxina Shiga/toxicidade , Escherichia coli Shiga Toxigênica/patogenicidade
4.
Cancer Res ; 76(13): 3895-903, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27197184

RESUMO

The Golgi apparatus is responsible for transporting, processing, and sorting numerous proteins in the cell, including cell surface-expressed receptor tyrosine kinases (RTK). The small-molecule compound M-COPA [2-methylcoprophilinamide (AMF-26)] disrupts the Golgi apparatus by inhibiting the activation of Arf1, resulting in suppression of tumor growth. Here, we report an evaluation of M-COPA activity against RTK-addicted cancers, focusing specifically on human gastric cancer (GC) cells with or without MET amplification. As expected, the MET-addicted cell line MKN45 exhibited a better response to M-COPA than cell lines without MET amplification. Upon M-COPA treatment, cell surface expression of MET was downregulated with a concurrent accumulation of its precursor form. M-COPA also reduced levels of the phosphorylated form of MET along with the downstream signaling molecules Akt and S6. Similar results were obtained in additional GC cell lines with amplification of MET or the FGF receptor FGFR2 MKN45 murine xenograft experiments demonstrated the antitumor activity of M-COPA in vivo Taken together, our results offer an initial preclinical proof of concept for the use of M-COPA as a candidate treatment option for MET-addicted GC, with broader implications for targeting the Golgi apparatus as a novel cancer therapeutic approach. Cancer Res; 76(13); 3895-903. ©2016 AACR.


Assuntos
Complexo de Golgi/efeitos dos fármacos , Naftóis/farmacologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Piridinas/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Neoplasias Gástricas/patologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Estudos de Casos e Controles , Proliferação de Células/efeitos dos fármacos , Feminino , Seguimentos , Complexo de Golgi/metabolismo , Complexo de Golgi/patologia , Humanos , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Estadiamento de Neoplasias , Fosforilação/efeitos dos fármacos , Prognóstico , Proteínas Proto-Oncogênicas c-met/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biochem Pharmacol ; 86(9): 1272-84, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23973528

RESUMO

Tamoxifen is an anticancer agent widely used for treatment of estrogen receptor (ERα)-positive breast cancer. We previously developed a novel synthesis of tamoxifen and its derivatives, named Ridaifens (RIDs). Some of them, including RID-SB8, exhibited a stronger anticancer activity than tamoxifen in ERα-positive MCF-7 cells while having lost the affinity for ERα, suggesting an ERα-independent anticancer mode of action. In this study, we investigated the underlying mechanism by which RID-SB8 exerts anticancer activity. As expected, anticancer activity of RID-SB8 was not influenced upon knockdown of ERα expression in MCF-7 cells. RID-SB8 exerted similar anticancer effects on thirteen ERα-negative cancer cell lines including human gliosarcoma SF539 cells. In SF539 cells, RID-SB8 triggered loss of mitochondrial membrane potential (ΔΨ(m)) and progression of apoptosis accompanied by activation of caspases and translocation of apoptosis-inducing factor (AIF) to the nucleus. Furthermore, it induced reactive oxygen species (ROS), and a ROS scavenger, N-acetylcysteine (NAC), canceled loss of ΔΨ(m) and progression of apoptosis triggered by RID-SB8. Using fifteen human cancer cell lines, we demonstrated a significant correlation between RID-SB8 concentration required for ROS production and that required for cytotoxic effect across these cell lines, but such correlation was not observed for tamoxifen. Finally, the selective induction of ROS and cytotoxic effect on cancer cells by RID-SB8 were confirmed. From these results, we concluded that RID-SB8 exerts an anticancer effect via a mode of action distinct from tamoxifen, and that RID-SB8 could become a promising anticancer lead compound which selectively induces ROS formation and apoptosis in cancer cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Tamoxifeno/análogos & derivados , Acetilcisteína/farmacologia , Antineoplásicos/síntese química , Fator de Indução de Apoptose/genética , Fator de Indução de Apoptose/metabolismo , Caspases/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Sequestradores de Radicais Livres/farmacologia , Técnicas de Silenciamento de Genes , Gliossarcoma/tratamento farmacológico , Gliossarcoma/genética , Gliossarcoma/metabolismo , Gliossarcoma/patologia , Humanos , Células MCF-7/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/farmacologia
6.
J Med Chem ; 56(1): 150-9, 2013 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-23214926

RESUMO

An effective method for the total synthesis of 1 (AMF-26), a potentially promising new anticancer drug that disrupts the Golgi system by inhibiting the ADP-ribosylation factor 1 (Arf1) activation, has been developed for the first time. The construction of the chiral linear precursor (a key to the synthesis) was achieved by the asymmetric aldol reaction followed by the computer-assisted predictive stereoselective intramolecular Diels-Alder reaction. The global antitumor activity of the totally synthetic 1 against a variety of human cancer cells was assessed using a panel of 39 human cancer cell lines (JFCR39), and it was shown that the synthetic 1 strongly inhibited the growth of several cancer cell lines at concentrations of less than 0.04 µM. Biological assays of novel derivatives, 26 and 31, which have different side-chains at the C-4 positions in the Δ¹,²-octalin backbone, disclosed the importance of the suitable structure of the side-chain containing conjugated multidouble bonds.


Assuntos
Fator 1 de Ribosilação do ADP/metabolismo , Antineoplásicos/síntese química , Complexo de Golgi/efeitos dos fármacos , Naftóis/síntese química , Piridinas/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Complexo de Golgi/metabolismo , Humanos , Estrutura Molecular , Naftóis/química , Naftóis/farmacologia , Piridinas/química , Piridinas/farmacologia , Estereoisomerismo , Relação Estrutura-Atividade , Termodinâmica
7.
J Biol Chem ; 287(6): 3885-97, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22158626

RESUMO

ADP-ribosylation factor 1 (Arf1) plays a major role in mediating vesicular transport. Brefeldin A (BFA), a known inhibitor of the Arf1-guanine nucleotide exchange factor (GEF) interaction, is highly cytotoxic. Therefore, interaction of Arf1 with ArfGEF is an attractive target for cancer treatment. However, BFA and its derivatives have not progressed beyond the pre-clinical stage of drug development because of their poor bioavailability. Here, we aimed to identify novel inhibitors of the Arf1-ArfGEF interaction that display potent antitumor activity in vivo but with a chemical structure distinct from that of BFA. We exploited a panel of 39 cell lines (termed JFCR39) coupled with a drug sensitivity data base and COMPARE algorithm, resulting in the identification of a possible novel Arf1-ArfGEF inhibitor AMF-26, which differed structurally from BFA. By using a pulldown assay with GGA3-conjugated beads, we demonstrated that AMF-26 inhibited Arf1 activation. Subsequently, AMF-26 induced Golgi disruption, apoptosis, and cell growth inhibition. Computer modeling/molecular dynamics (MD) simulation suggested that AMF-26 bound to the contact surface of the Arf1-Sec7 domain where BFA bound. AMF-26 affected membrane traffic, including the cis-Golgi and trans-Golgi networks, and the endosomal systems. Furthermore, using AMF-26 and its derivatives, we demonstrated that there was a significant correlation between cell growth inhibition and Golgi disruption. In addition, orally administrated AMF-26 (83 mg/kg of body weight; 5 days) induced complete regression of human breast cancer BSY-1 xenografts in vivo, suggesting that AMF-26 is a novel anticancer drug candidate that inhibits the Golgi system, targeting Arf1 activation.


Assuntos
Fator 1 de Ribosilação do ADP/antagonistas & inibidores , Algoritmos , Simulação por Computador , Inibidores Enzimáticos/farmacologia , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Rede trans-Golgi/enzimologia , Fator 1 de Ribosilação do ADP/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Bases de Dados Factuais , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática/efeitos dos fármacos , Humanos
8.
Cancer Res ; 71(20): 6419-27, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21868757

RESUMO

Although the importance of glycans in malignant cell behavior is well documented, the potential involvement of endogenous lectins as modifiers of progression and metastasis in the tumor microenvironment has not been explored. In this study, we show that loss of the hepatic asialoglycoprotein receptor (ASGPR) in mice severely reduces the frequency of spontaneous lung metastasis after intrahepatic implantation of murine Lewis lung carcinoma (3LL) cells. Conversely, in vitro treatment with recombinant ASGPR increased the invasive and metastatic capacity of 3LL cells before intrahepatic implantation. ASGPR treatment in vitro increased the expression and production of matrix metalloproteinase-9 through activation of the epidermal growth factor receptor-extracellular signal-regulated kinase (EGFR-ERK) pathway. Our findings identify ASGPR as a novel important factor that responds to endogenous lectins in the tumor microenvironment to promote cancer metastasis by activating the EGFR-ERK pathway through interactions with counter-receptors on cancer cells.


Assuntos
Receptor de Asialoglicoproteína/metabolismo , Carcinoma Pulmonar de Lewis/secundário , Receptores ErbB/metabolismo , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases , Animais , Carcinoma Pulmonar de Lewis/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Camundongos Endogâmicos C57BL
9.
Glycoconj J ; 27(2): 267-76, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20077002

RESUMO

O-glycosylation of mucin is initiated by the attachment of N-acetyl-D-galactosamine (GalNAc) to serine or threonine residues in mucin core polypeptides by UDPGalNAc:polypeptide N-acetylgalactosaminyltransferases (ppGalNAc-Ts). It is not well understood how GalNAc attachment is regulated by multiple ppGalNAc-Ts in each cell. In the present study, the expression levels of murine ppGalNAc-Ts (mGalNAc-Ts), T1, T2, T3, T4, T6, and T7 were compared between mouse colon carcinoma colon 38 cells and variant SL4 cells, selected for their metastatic potentials, by using the competitive RT-PCR method. The expression levels of mGalNAc-T1, T2, and T7 were slightly higher in the SL4 cells than in the colon 38 cells, whereas the expression level of mGalNAc-T3 in the SL4 cells was 1.5% of that in the colon 38 cells. Products of enzymatic incorporations of GalNAc residues into FITCPTTTPITTTTK peptide by the use of microsome fractions of these cells as the enzyme source were separated and characterized for the number of attached GalNAc residues and their positions. The maximum number of attached GalNAc residues was 6 and 4 when the microsome fractions of the colon 38 cells and SL4 cells were used, respectively. When the microsome fractions of the colon 38 cells were treated with a polyclonal antibody raised against mGalNAc-T3, the maximum number of incorporated GalNAc residues was 4. These results strongly suggest that mGalNAc-T3 in colon 38 cells is involved in additional transfer of GalNAc residues to this peptide.


Assuntos
Neoplasias do Colo/enzimologia , Neoplasias Hepáticas/secundário , N-Acetilgalactosaminiltransferases/metabolismo , Acetilgalactosamina/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/farmacologia , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicopeptídeos/química , Glicopeptídeos/metabolismo , Glicosilação/efeitos dos fármacos , Humanos , Lectinas/metabolismo , Camundongos , Microssomos/efeitos dos fármacos , Microssomos/enzimologia , Dados de Sequência Molecular , Mucinas/metabolismo , N-Acetilgalactosaminiltransferases/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Especificidade por Substrato/efeitos dos fármacos , Polipeptídeo N-Acetilgalactosaminiltransferase
10.
Oncol Res ; 17(10): 437-45, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19725223

RESUMO

Twenty-three human colorectal carcinoma cell lines were examined for the binding of recombinant hepatic asialoglycoprotein receptor 1 (ASGR1), which is known to be exclusively expressed on hepatic parenchymal cells. The effects of the binding were assessed by adhesion to and proliferation on immobilized recombinant ASGR1. Recombinant ASGR1 bound strongly to six cell lines and moderately to 15 cell lines out of 23 lines tested, as shown by flow cytometric analysis. The first six cell lines (group A) also exhibited strong adherence to immobilized ASGR1, whereas 11 of the 15 cell lines of the second group (group B) showed significant adhesion with smaller enhancement by ASGR1 than the cell lines in group A. With a representative cell line (DLD-1 cells categorized in group B), a significant portion of the adhesion was inhibited by preincubation of ASGR1 with asialofetuin, a competitive inhibitor of the carbohydrate recognition by ASGR1. The growth rates of 13 cell lines (two of group A and 11 of group B) were significantly accelerated when they were cultured on immobilized recombinant ASGR1. The results indicate that ASGR is a potential organ-specific microenvironmental factor for colorectal carcinoma growth and metastasis formation in livers.


Assuntos
Receptor de Asialoglicoproteína/metabolismo , Proliferação de Células , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Assialoglicoproteínas/farmacologia , Adesão Celular , Fetuínas , Citometria de Fluxo , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Tumorais Cultivadas , alfa-Fetoproteínas/farmacologia
11.
Clin Exp Metastasis ; 22(6): 513-21, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16320114

RESUMO

Highly metastatic variants of mouse colon 38 colon carcinoma cells were established by repeated selection in vivo for liver metastasis and designated as SL4 cells. The SL4 cells formed colonies in the liver of 100% of syngenic mice when injected intrasplenically, while the incidence of liver metastasis was 27% of mice injected with parental cells. The weight of livers, which is an indicator of experimental hepatic metastasis formation, was significantly higher after intrasplenic injection and subsequent splenoctomy with SL4 cells than colon 38 cells. The incidence of hepatic metastasis after intracecal injection of SL4 cells was significantly higher than that of colon 38 cells. The SL4 cells were tested in vitro for their properties. Differences were not detected in the motility and invasive behavior between colon 38 cells and SL4 cells. SL4 cells showed a higher proliferation rate than colon 38 cells under adherent conditions. SL4 cells maintained a capacity to proliferate under non-adherent conditions whereas parental cells did not. SL4 cells should be a useful tool to study the mechanism of hepatic metastasis of colon carcinoma cells and to develop methods to prevent hepatic metastasis.


Assuntos
Carcinoma/secundário , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Neoplasias Hepáticas/secundário , Camundongos , Animais , Feminino , Camundongos Endogâmicos C57BL
12.
J Biol Chem ; 277(32): 28892-901, 2002 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-12016228

RESUMO

A novel mouse macrophage galactose-type C-type lectin 2 (mMGL2) was identified by BLAST analysis of expressed sequence tags. The sequence of mMGL2 is highly homologous to the mMGL, which should now be called mMGL1. The open reading frame of mMGL2 contains a sequence corresponding to a type II transmembrane protein with 332 amino acids having a single extracellular C-type lectin domain. The 3'-untranslated region included long terminal repeats of mouse early transposon. The Mgl2 gene was cloned from a 129/SvJ mouse genomic library and sequenced. The gene spans 7,136 base pairs and consists of 10 exons, which is similar to the genomic organization of mMGL1. The reverse transcriptase-PCR analysis indicates that mMGL2 is expressed in cell lines and normal mouse tissues in a macrophage-restricted manner, also very similar to that of mMGL1. The mMGL2 mRNA was also detected in mMGL1-positive cells, which were sorted from thioglycollate-induced peritoneal cells with a mMGL1-specific monoclonal antibody, LOM-8.7. The soluble recombinant proteins of mMGL2 exhibited carbohydrate specificity for alpha- and beta-GalNAc-conjugated soluble polyacrylamides, whereas mMGL1 preferentially bound Lewis X-conjugated soluble polyacrylamides in solid phase assays. These two lectins may function cooperatively as recognition and endocytic molecules on macrophages and related cells.


Assuntos
Lectinas Tipo C , Lectinas/biossíntese , Lectinas/genética , Lectinas/metabolismo , Macrófagos/metabolismo , Proteínas de Membrana , Regiões 3' não Traduzidas , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/metabolismo , Assialoglicoproteínas , Sequência de Bases , Metabolismo dos Carboidratos , Linhagem Celular , Clonagem Molecular , Citoplasma/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Endocitose , Éxons , Feminino , Concentração de Íons de Hidrogênio , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Filogenia , Ligação Proteica , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA