Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(38): 13870-5, 2014 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-25189770

RESUMO

Metastatic spread is the leading cause of cancer mortality. Breast cancer (BCa) metastatic recurrence can happen years after removal of the primary tumor. Here we show that Ubc13, an E2 enzyme that catalyzes K63-linked protein polyubiquitination, is largely dispensable for primary mammary tumor growth but is required for metastatic spread and lung colonization by BCa cells. Loss of Ubc13 inhibited BCa growth and survival only at metastatic sites. Ubc13 was dispensable for transforming growth factor ß (TGFß)-induced SMAD activation but was required for activation of non-SMAD signaling via TGFß-activating kinase 1 (TAK1) and p38, whose activity controls expression of numerous metastasis promoting genes. p38 activation restored metastatic activity to Ubc13-deficient cells, and its pharmacological inhibition attenuated BCa metastasis in mice, suggesting it is a therapeutic option for metastatic BCa.


Assuntos
Neoplasias da Mama/enzimologia , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Enzimas de Conjugação de Ubiquitina/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
2.
Virus Genes ; 45(3): 508-17, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22864547

RESUMO

Jaagsiekte sheep retrovirus (JSRV) is the causative agent of a transmissible lung cancer in sheep. A unique feature is that JSRV envelope protein is also the oncogene for this virus. Previous studies have identified the cytoplasmic tail (CT) of the envelope transmembrane (TM) protein as critical for transformation although other regions of Env have also been implicated. In this study, the roles of other Env regions in transformation were investigated. Chimeras between JSRV Env and the Env of a related non-oncogenic endogenous retrovirus (enJSRV, 5F16) were used. A chimera containing the membrane-spanning region (MSR) of enJSRV inserted into JSRV Env showed substantially reduced transformation, indicating that the MSR plays a role in transformation. Transformation by this chimera was highly dependent on both Ras/Raf/MEK/MAPK and PI3K/Akt/mTOR signaling. A chimera containing the two amino acids in the TM ectodomain that distinguish JSRV and enJSRV showed modestly reduced transformation. Chimeras in the SU protein indicated that the amino terminal region of SU contributes to transformation, while the C-terminal part is not important. To test if Env trimerization is important for transformation, we mutated a leucine-rich sequence in the putative trimerization domain in the ectodomain of TM (Tri-M). This mutant could not transform cells and it did not oligomerize. However, Tri-M could complement a non-transforming mutant CT mutant (Y590F) so oligomerization is not necessary for at least some aspects of transformation. These experiments provide new insight into the regions and residues of JSRV Env protein necessary for oncogenic transformation.


Assuntos
Transformação Celular Viral , Retrovirus Jaagsiekte de Ovinos/genética , Adenomatose Pulmonar Ovina/virologia , Proteínas do Envelope Viral/metabolismo , Animais , Eletroforese em Gel de Poliacrilamida , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células HEK293 , Humanos , Retrovirus Jaagsiekte de Ovinos/metabolismo , Leucina/genética , Leucina/metabolismo , Camundongos , Mutagênese Sítio-Dirigida , Células NIH 3T3 , Plasmídeos/genética , Plasmídeos/metabolismo , Multimerização Proteica , Estrutura Terciária de Proteína , Adenomatose Pulmonar Ovina/patologia , Ensaio de Radioimunoprecipitação , Ovinos/virologia , Relação Estrutura-Atividade , Proteínas do Envelope Viral/genética
3.
Sci Rep ; 10(1): 2688, 2020 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-32060361

RESUMO

B-cell acute lymphoblastic leukemia (B-ALL) accounts for nearly one fifth of all childhood cancers and current challenges in B-ALL treatment include resistance, relapse and late-onset side effects due to the chemotherapy. To overcome these hurdles, novel therapies need to be investigated. One promising target is Polo-like kinase 1 (Plk1), a key regulator of the cell cycle. In this study, the Plk family expression is investigated in primary peripheral blood and bone marrow mononuclear cells from ten pediatric B-ALL patients. For the first time, short interfering RiboNucleic Neutrals (siRNNs) that enter cells without a transfection reagent are used to target Plk1 mRNA in primary cells from pediatric B-ALL patients. Our results show that the expression of Plk1 and Plk4 is significantly higher in pediatric B-ALL patients compared to healthy donors. Moreover, treatment of primary peripheral blood and bone marrow mononuclear cells from pediatric B-ALL patients, cultured ex vivo, with Plk1-targeting siRNNs results in cleavage of Plk1 mRNA. Importantly, the Plk1 knockdown is specific and does not affect other Plk members in contrast to many small molecule Plk1 inhibitors. Thus, Plk1 is a potential therapeutic target in pediatric B-ALL and selective targeting of Plk1 can be achieved by the use of siRNNs.


Assuntos
Proteínas de Ciclo Celular/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Adolescente , Apoptose/efeitos dos fármacos , Linfócitos B/efeitos dos fármacos , Proteínas de Ciclo Celular/antagonistas & inibidores , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Criança , Pré-Escolar , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lactente , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , RNA Mensageiro , Quinase 1 Polo-Like
4.
Nucleic Acid Ther ; 27(5): 260-271, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28933656

RESUMO

Small double-stranded, left-handed hairpin (LHP) RNAs containing a 5'-guide-loop-passenger-3' structure induce RNAi responses by a poorly understood mechanism. To explore LHPs, we synthesized fully 2'-modified LHP RNAs targeting multiple genes and found all to induce robust RNAi responses. Deletion of the loop and nucleotides at the 5'-end of the equivalent passenger strand resulted in a smaller LHP that still induced strong RNAi responses. Surprisingly, progressive deletion of up to 10 nucleotides from the 3'-end of the guide strand resulted in a 32mer LHP capable of inducing robust RNAi responses. However, further guide strand deletion inhibited LHP activity, thereby defining the minimal length guide targeting length to 13 nucleotides. To dissect LHP processing, we examined LHP species that coimmunoprecipitated with Argonaute 2 (Ago2), the catalytic core of RNA-induced silencing complex, and found that the Ago2-associated processed LHP species was of a length that correlated with Ago2 cleavage of the passenger strand. Placement of a blocking 2'-OMe blocking modification at the LHP predicted Ago2 cleavage site resulted in an intact LHP loaded into Ago2 and no RNAi response. Taken together, these data argue that in the absence of a substantial loop, this novel class of small LHP RNAs enters the RNAi pathway by a Dicer-independent mechanism that involves Ago2 cleavage and results in an extended guide strand. This work establishes LHPs as an alternative RNAi trigger that can be produced from a single synthesis for potential use as an RNAi therapeutic.


Assuntos
Proteínas Argonautas/metabolismo , Interferência de RNA/efeitos dos fármacos , RNA de Cadeia Dupla/uso terapêutico , RNA Interferente Pequeno/uso terapêutico , Complexo de Inativação Induzido por RNA/metabolismo , Terapêutica com RNAi/métodos , Proteínas Argonautas/genética , Linhagem Celular Tumoral , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Humanos , RNA de Cadeia Dupla/síntese química , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , RNA Interferente Pequeno/síntese química , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Complexo de Inativação Induzido por RNA/genética , Ribonuclease III/genética , Ribonuclease III/metabolismo , Deleção de Sequência
5.
J Control Release ; 261: 199-206, 2017 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-28684168

RESUMO

Epidemiological studies of childhood leukemia survivors reveal an alarmingly high incidence of chronic health disabilities after treatment, therefore, more specific therapies need to be developed. Polo-like kinase 1 (Plk1) is a key player in mitosis and a target for drug development as it is upregulated in multiple cancer types. Small molecules targeting Plk1 are mainly ATP-competitors and, therefore, are known to elicit side effects due to lack of specificity. RNA interference (RNAi) is known for its high catalytic activity and target selectivity; however, the biggest barrier for its introduction into clinical use is its delivery. RNAi prodrugs are modified, self-delivering short interfering Ribonucleic Neutrals (siRNNs), cleaved by cytoplasmic enzymes into short interfering Ribonucleic Acids (siRNAs) once inside cells. In this study we aimed to investigate the potential of siRNNs as therapeutic tools in T-acute lymphoblastic leukemia (T-ALL) using T-ALL cell lines and patient-derived samples. We demonstrate for the first time that RNAi prodrugs (siRNNs) targeting Plk1, can enter pediatric T-ALL patient cells without a transfection reagent and induce Plk1 knockdown on both protein and mRNA levels resulting in G2/M-arrest and apoptosis. We also show that siRNNs targeting Plk1 generate less toxicity in normal cells compared to the small molecule Plk1 inhibitor, BI6727, suggesting a potentially good therapeutic index.


Assuntos
Proteínas de Ciclo Celular/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/terapia , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , Apoptose/genética , Linhagem Celular Tumoral , Criança , Sistemas de Liberação de Medicamentos , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Técnicas de Silenciamento de Genes , Inativação Gênica , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Pró-Fármacos , Pteridinas/farmacologia , Pteridinas/toxicidade , RNA Mensageiro/genética , RNA Interferente Pequeno/toxicidade , Quinase 1 Polo-Like
6.
Methods Mol Biol ; 1364: 1-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26472437

RESUMO

Due to their high potency (EC50 ~1 pM) and exquisite target selectivity for all expressed mRNAs, small interfering RNA (siRNA)-induced RNAi responses hold significant promise as a therapeutic modality. However, the size and high negative charge of siRNAs render them unable to enter cells without assistance from a delivery agent. Most current methods of siRNA delivery rely on encasing siRNA molecules in large nanoparticles or cationic liposomes. However, these approaches suffer from a number of problems, including a poor diffusion coefficient, cytotoxicity, and poor pharmacokinetics. To address the problem of siRNA in vivo delivery, we developed monomeric neutral RNAi prodrugs, termed siRibonucleic neutrals (siRNNs), that directly neutralize the phosphate backbone negative charge by synthesis with bioreversible phosphotriester groups that are enzymatically cleaved off in the cytoplasm of cells. Here we describe the synthesis and purification of siRNN conjugates that induce in vivo target gene knockdown following systemic delivery into mice.


Assuntos
Oligonucleotídeos/química , Pró-Fármacos/química , Pró-Fármacos/síntese química , RNA Interferente Pequeno/química , Acetilgalactosamina/química , Animais , Técnicas de Química Sintética , Técnicas de Silenciamento de Genes , Camundongos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
7.
Sci Rep ; 6: 32301, 2016 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-27604151

RESUMO

Bioactive macromolecular peptides and oligonucleotides have significant therapeutic potential. However, due to their size, they have no ability to enter the cytoplasm of cells. Peptide/Protein transduction domains (PTDs), also called cell-penetrating peptides (CPPs), can promote uptake of macromolecules via endocytosis. However, overcoming the rate-limiting step of endosomal escape into the cytoplasm remains a major challenge. Hydrophobic amino acid R groups are known to play a vital role in viral escape from endosomes. Here we utilize a real-time, quantitative live cell split-GFP fluorescence complementation phenotypic assay to systematically analyze and optimize a series of synthetic endosomal escape domains (EEDs). By conjugating EEDs to a TAT-PTD/CPP spilt-GFP peptide complementation assay, we were able to quantitatively measure endosomal escape into the cytoplasm of live cells via restoration of GFP fluorescence by intracellular molecular complementation. We found that EEDs containing two aromatic indole rings or one indole ring and two aromatic phenyl groups at a fixed distance of six polyethylene glycol (PEG) units from the TAT-PTD-cargo significantly enhanced cytoplasmic delivery in the absence of cytotoxicity. EEDs address the critical rate-limiting step of endosomal escape in delivery of macromolecular biologic peptide, protein and siRNA therapeutics into cells.


Assuntos
Produtos Biológicos/metabolismo , Peptídeos Penetradores de Células/metabolismo , Endossomos/metabolismo , Substâncias Macromoleculares/metabolismo , Sítios de Ligação/genética , Produtos Biológicos/administração & dosagem , Linhagem Celular , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/genética , Sistemas de Liberação de Medicamentos/métodos , Endocitose , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Células MCF-7 , Substâncias Macromoleculares/administração & dosagem , Microscopia de Fluorescência , Peptídeos/administração & dosagem , Peptídeos/genética , Peptídeos/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Imagem com Lapso de Tempo/métodos
8.
Nat Biotechnol ; 32(12): 1256-61, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25402614

RESUMO

RNA interference (RNAi) has great potential to treat human disease. However, in vivo delivery of short interfering RNAs (siRNAs), which are negatively charged double-stranded RNA macromolecules, remains a major hurdle. Current siRNA delivery has begun to move away from large lipid and synthetic nanoparticles to more defined molecular conjugates. Here we address this issue by synthesis of short interfering ribonucleic neutrals (siRNNs) whose phosphate backbone contains neutral phosphotriester groups, allowing for delivery into cells. Once inside cells, siRNNs are converted by cytoplasmic thioesterases into native, charged phosphodiester-backbone siRNAs, which induce robust RNAi responses. siRNNs have favorable drug-like properties, including high synthetic yields, serum stability and absence of innate immune responses. Unlike siRNAs, siRNNs avidly bind serum albumin to positively influence pharmacokinetic properties. Systemic delivery of siRNNs conjugated to a hepatocyte-specific targeting domain induced extended dose-dependent in vivo RNAi responses in mice. We believe that siRNNs represent a technology that will open new avenues for development of RNAi therapeutics.


Assuntos
Sistemas de Liberação de Medicamentos , Pró-Fármacos/uso terapêutico , RNA Interferente Pequeno/uso terapêutico , Animais , Humanos , Camundongos , Nanopartículas/química , Nanopartículas/uso terapêutico , Pró-Fármacos/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/genética , Albumina Sérica/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA