Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cancer Discov ; 14(4): 610-614, 2024 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-38571419

RESUMO

SUMMARY: Cancer is traditionally perceived through a genetic lens, with therapeutic strategies targeting oncogenic driver mutations. We advocate an overarching framework recognizing tumors as comprising driver, passenger, and trailer cell states: Tailoring therapies to simultaneously target driver genetics and cell states may enhance effectiveness and durability. SIGNIFICANCE: We redefine cancer progression by introducing a model that categorizes tumor cells into "driver," "passenger," and "trailer" phenotypes, expanding the focus on genetic aberrations to cellular behavior. This approach offers a roadmap to guide refining therapeutic strategies for more precise and durable cancer treatments that address tumor heterogeneity and plasticity.


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/patologia
2.
Nat Commun ; 15(1): 897, 2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38316783

RESUMO

Descriptive data are rapidly expanding in biomedical research. Instead, functional validation methods with sufficient complexity remain underdeveloped. Transcriptional reporters allow experimental characterization and manipulation of developmental and disease cell states, but their design lacks flexibility. Here, we report logical design of synthetic cis-regulatory DNA (LSD), a computational framework leveraging phenotypic biomarkers and trans-regulatory networks as input to design reporters marking the activity of selected cellular states and pathways. LSD uses bulk or single-cell biomarkers and a reference genome or custom cis-regulatory DNA datasets with user-defined boundary regions. By benchmarking validated reporters, we integrate LSD with a computational ranking of phenotypic specificity of putative cis-regulatory DNA. Experimentally, LSD-designed reporters targeting a wide range of cell states are functional without minimal promoters. Applied to broadly expressed genes from human and mouse tissues, LSD generates functional housekeeper-like sLCRs compatible with size constraints of AAV vectors for gene therapy applications. A mesenchymal glioblastoma reporter designed by LSD outperforms previously validated ones and canonical cell surface markers. In genome-scale CRISPRa screens, LSD facilitates the discovery of known and novel bona fide cell-state drivers. Thus, LSD captures core principles of cis-regulation and is broadly applicable to studying complex cell states and mechanisms of transcriptional regulation.


Assuntos
DNA , Regulação da Expressão Gênica , Animais , Humanos , Camundongos , Regiões Promotoras Genéticas/genética , Expressão Gênica , Biomarcadores
3.
Sci Adv ; 9(25): eadf4975, 2023 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-37343108

RESUMO

Epithelial immune responses govern tissue homeostasis and offer drug targets against maladaptation. Here, we report a framework to generate drug discovery-ready reporters of cellular responses to viral infection. We reverse-engineered epithelial cell responses to SARS-CoV-2, the viral agent fueling the ongoing COVID-19 pandemic, and designed synthetic transcriptional reporters whose molecular logic comprises interferon-α/ß/γ and NF-κB pathways. Such regulatory potential reflected single-cell data from experimental models to severe COVID-19 patient epithelial cells infected by SARS-CoV-2. SARS-CoV-2, type I interferons, and RIG-I drive reporter activation. Live-cell image-based phenotypic drug screens identified JAK inhibitors and DNA damage inducers as antagonistic modulators of epithelial cell response to interferons, RIG-I stimulation, and SARS-CoV-2. Synergistic or antagonistic modulation of the reporter by drugs underscored their mechanism of action and convergence on endogenous transcriptional programs. Our study describes a tool for dissecting antiviral responses to infection and sterile cues and rapidly discovering rational drug combinations for emerging viruses of concern.


Assuntos
COVID-19 , Interferon Tipo I , Humanos , SARS-CoV-2 , Pandemias , Células Epiteliais
4.
Elife ; 102021 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-34075878

RESUMO

High spliceosome activity is a dependency for cancer cells, making them more vulnerable to perturbation of the splicing machinery compared to normal cells. To identify splicing factors important for prostate cancer (PCa) fitness, we performed pooled shRNA screens in vitro and in vivo. Our screens identified heterogeneous nuclear ribonucleoprotein M (HNRNPM) as a regulator of PCa cell growth. RNA- and eCLIP-sequencing identified HNRNPM binding to transcripts of key homeostatic genes. HNRNPM binding to its targets prevents aberrant exon inclusion and backsplicing events. In both linear and circular mis-spliced transcripts, HNRNPM preferentially binds to GU-rich elements in long flanking proximal introns. Mimicry of HNRNPM-dependent linear-splicing events using splice-switching-antisense-oligonucleotides was sufficient to inhibit PCa cell growth. This suggests that PCa dependence on HNRNPM is likely a result of mis-splicing of key homeostatic coding and non-coding genes. Our results have further been confirmed in other solid tumors. Taken together, our data reveal a role for HNRNPM in supporting cancer cell fitness. Inhibition of HNRNPM activity is therefore a potential therapeutic strategy in suppressing growth of PCa and other solid tumors.


Assuntos
Adenocarcinoma/metabolismo , Proliferação de Células , Ribonucleoproteínas Nucleares Heterogêneas Grupo M/metabolismo , Neoplasias da Próstata/metabolismo , Splicing de RNA , RNA Circular/biossíntese , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Ribonucleoproteínas Nucleares Heterogêneas Grupo M/genética , Humanos , Masculino , Camundongos SCID , Células PC-3 , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , RNA Circular/genética , Carga Tumoral , Células Tumorais Cultivadas
5.
Sci Adv ; 7(9)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33627422

RESUMO

Epithelial-mesenchymal transition (EMT) is a developmental process hijacked by cancer cells to modulate proliferation, migration, and stress response. Whereas kinase signaling is believed to be an EMT driver, the molecular mechanisms underlying epithelial-mesenchymal interconversion are incompletely understood. Here, we show that the impact of chromatin regulators on EMT interconversion is broader than that of kinases. By combining pharmacological modulation of EMT, synthetic genetic tracing, and CRISPR interference screens, we uncovered a minority of kinases and several chromatin remodelers, writers, and readers governing homeostatic EMT in lung cancer cells. Loss of ARID1A, DOT1L, BRD2, and ZMYND8 had nondeterministic and sometimes opposite consequences on epithelial-mesenchymal interconversion. Together with RNAPII and AP-1, these antagonistic gatekeepers control chromatin of active enhancers, including pan-cancer-EMT signature genes enabling supraclassification of anatomically diverse tumors. Thus, our data uncover general principles underlying transcriptional control of cancer cell plasticity and offer a platform to systematically explore chromatin regulators in tumor-state-specific therapy.

6.
Cancer Discov ; 11(3): 754-777, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33361384

RESUMO

Glioblastoma is a lethal brain tumor that exhibits heterogeneity and resistance to therapy. Our understanding of tumor homeostasis is limited by a lack of genetic tools to selectively identify tumor states and fate transitions. Here, we use glioblastoma subtype signatures to construct synthetic genetic tracing cassettes and investigate tumor heterogeneity at cellular and molecular levels, in vitro and in vivo. Through synthetic locus control regions, we demonstrate that proneural glioblastoma is a hardwired identity, whereas mesenchymal glioblastoma is an adaptive and metastable cell state driven by proinflammatory and differentiation cues and DNA damage, but not hypoxia. Importantly, we discovered that innate immune cells divert glioblastoma cells to a proneural-to-mesenchymal transition that confers therapeutic resistance. Our synthetic genetic tracing methodology is simple, scalable, and widely applicable to study homeostasis in development and diseases. In glioblastoma, the method causally links distinct (micro)environmental, genetic, and pharmacologic perturbations and mesenchymal commitment. SIGNIFICANCE: Glioblastoma is heterogeneous and incurable. Here, we designed synthetic reporters to reflect the transcriptional output of tumor cell states and signaling pathways' activity. This method is generally applicable to study homeostasis in normal tissues and diseases. In glioblastoma, synthetic genetic tracing causally connects cellular and molecular heterogeneity to therapeutic responses.This article is highlighted in the In This Issue feature, p. 521.


Assuntos
Comunicação Celular , Regulação Neoplásica da Expressão Gênica , Glioblastoma/etiologia , Glioblastoma/patologia , Imunidade Inata , Biomarcadores Tumorais , Comunicação Celular/genética , Suscetibilidade a Doenças , Glioblastoma/metabolismo , Humanos , Imunidade Inata/genética , Gradação de Tumores , Estadiamento de Neoplasias , Transcriptoma , Microambiente Tumoral
7.
Cell Rep ; 33(13): 108533, 2020 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-33378683

RESUMO

Altering ubiquitination by disruption of deubiquitinating enzymes (DUBs) affects hematopoietic stem cell (HSC) maintenance. However, comprehensive knowledge of DUB function during hematopoiesis in vivo is lacking. Here, we systematically inactivate DUBs in mouse hematopoietic progenitors using in vivo small hairpin RNA (shRNA) screens. We find that multiple DUBs may be individually required for hematopoiesis and identify ubiquitin-specific protease 15 (USP15) as essential for HSC maintenance in vitro and in transplantations and Usp15 knockout (KO) mice in vivo. USP15 is highly expressed in human hematopoietic tissues and leukemias. USP15 depletion in murine progenitors and leukemia cells impairs in vitro expansion and increases genotoxic stress. In leukemia cells, USP15 interacts with and stabilizes FUS (fused in sarcoma), a known DNA repair factor, directly linking USP15 to the DNA damage response (DDR). Our study underscores the importance of DUBs in preserving normal hematopoiesis and uncovers USP15 as a critical DUB in safeguarding genome integrity in HSCs and leukemia cells.


Assuntos
Enzimas Desubiquitinantes/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Leucemia/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Proteases Específicas de Ubiquitina/fisiologia , Animais , Linhagem Celular , Proliferação de Células , Dano ao DNA , Reparo do DNA , Hematopoese , Células-Tronco Hematopoéticas/enzimologia , Humanos , Células K562 , Leucemia/enzimologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ubiquitinação
8.
J Exp Med ; 215(12): 3115-3135, 2018 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-30487290

RESUMO

Kras-driven non-small-cell lung cancers (NSCLCs) are a leading cause of death with limited therapeutic options. Many NSCLCs exhibit high levels of Ezh2, the enzymatic subunit of polycomb repressive complex 2 (PRC2). We tested Ezh2 inhibitors as single agents or before chemotherapy in mice with orthotopic Kras-driven NSCLC grafts, which homogeneously express Ezh2. These tumors display sensitivity to EZH2 inhibition by GSK126 but also amplify an inflammatory program involving signaling through NF-κB and genes residing in PRC2-regulated chromatin. During this process, tumor cells overcome GSK126 antiproliferative effects. We identified oncogenes that may mediate progression through an in vivo RNAi screen aimed at targets of PRC2/NF-κB. An in vitro compound screening linked GSK126-driven inflammation and therapeutic vulnerability in human cells to regulation of RNA synthesis and proteostasis. Interestingly, GSK126-treated NSCLCs in vivo also showed an enhanced response to a combination of nimesulide and bortezomib. Thus, Ezh2 inhibition may restrict cell proliferation and promote defined adaptive responses. Targeting these responses potentially improves outcomes in Kras-driven NSCLCs.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proliferação de Células , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Células A549 , Animais , Bortezomib/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Indóis/farmacologia , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Proto-Oncogênicas p21(ras)/genética , Piridonas/farmacologia , Sulfonamidas/farmacologia
9.
Mol Cell Oncol ; 3(3): e1152345, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27314096

RESUMO

The Polycomb transcriptional repressors regulate normal tissue homeostasis and their function is often hijacked during oncogenesis. We recently uncovered the Polycomb repressive complex-2 (PRC2) genes Ezh2 and Eed as oncogenotype-dependent cancer genes. Notably, within the same oncogenotype, PRC2 dosage modulates lung tumor homeostasis and critically impacts non-tumor tissue function.

10.
Cancer Cell ; 29(1): 17-31, 2016 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-26766588

RESUMO

Polycomb repressive complexes (PRC) are frequently implicated in human cancer, acting either as oncogenes or tumor suppressors. Here, we show that PRC2 is a critical regulator of KRAS-driven non-small cell lung cancer progression. Modulation of PRC2 by either Ezh2 overexpression or Eed deletion enhances KRAS-driven adenomagenesis and inflammation, respectively. Eed-loss-driven inflammation leads to massive macrophage recruitment and marked decline in tissue function. Additional Trp53 inactivation activates a cell-autonomous epithelial-to-mesenchymal transition program leading to an invasive mucinous adenocarcinoma. A switch between methylated/acetylated chromatin underlies the tumor phenotypic evolution, prominently involving genes controlled by Hippo/Wnt signaling. Our observations in the mouse models were conserved in human cells. Importantly, PRC2 inactivation results in context-dependent phenotypic alterations, with implications for its therapeutic application.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Transição Epitelial-Mesenquimal/genética , Complexo Repressor Polycomb 2/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Acetilação , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Proliferação de Células/genética , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Proteína Potenciadora do Homólogo 2 de Zeste , Histonas/metabolismo , Humanos , Inflamação/genética , Inflamação/metabolismo , Camundongos Transgênicos , Complexo Repressor Polycomb 2/genética , Proteínas Proto-Oncogênicas p21(ras)/genética
11.
Nat Protoc ; 9(12): 2880-902, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25411954

RESUMO

Loss-of-function (LOF) experiments targeting multiple genes during tumorigenesis can be implemented using pooled shRNA libraries. RNAi screens in animal models rely on the use of multiple shRNAs to simultaneously disrupt gene function, as well as to serve as barcodes for cell fate outcomes during tumorigenesis. Here we provide a protocol for performing RNAi screens in orthotopic mouse tumor models, referring to glioma and lung adenocarcinoma as specific examples. The protocol aims to provide guidelines for applying RNAi to a diverse spectrum of solid tumors and to highlight crucial considerations when designing and performing these studies. It covers shRNA library assembly and packaging into lentiviral particles, and transduction into tumor-initiating cells (TICs), followed by in vivo transplantation, tumor DNA recovery, sequencing and analysis. Depending on the target genes and tumor model, tumor suppressors and oncogenes can be identified or biological pathways can be dissected in 6-9 weeks.


Assuntos
Marcação de Genes/métodos , Técnicas Genéticas , Neoplasias Experimentais/genética , RNA Interferente Pequeno/genética , Adenocarcinoma/genética , Adenocarcinoma de Pulmão , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Feminino , Biblioteca Gênica , Xenoenxertos , Humanos , Neoplasias Pulmonares/genética , Masculino , Camundongos Endogâmicos BALB C , Oncogenes , Interferência de RNA , Transfecção
12.
J Exp Med ; 211(9): 1759-77, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-25113974

RESUMO

Histone ubiquitination at DNA breaks is required for activation of the DNA damage response (DDR) and DNA repair. How the dynamic removal of this modification by deubiquitinating enzymes (DUBs) impacts genome maintenance in vivo is largely unknown. To address this question, we generated mice deficient for Ub-specific protease 3 (USP3; Usp3Δ/Δ), a histone H2A DUB which negatively regulates ubiquitin-dependent DDR signaling. Notably, USP3 deletion increased the levels of histone ubiquitination in adult tissues, reduced the hematopoietic stem cell (HSC) reserves over time, and shortened animal life span. Mechanistically, our data show that USP3 is important in HSC homeostasis, preserving HSC self-renewal, and repopulation potential in vivo and proliferation in vitro. A defective DDR and unresolved spontaneous DNA damage contribute to cell cycle restriction of Usp3Δ/Δ HSCs. Beyond the hematopoietic system, Usp3Δ/Δ animals spontaneously developed tumors, and primary Usp3Δ/Δ cells failed to preserve chromosomal integrity. These findings broadly support the regulation of chromatin ubiquitination as a key pathway in preserving tissue function through modulation of the response to genotoxic stress.


Assuntos
Dano ao DNA/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Animais , Carcinogênese , Proliferação de Células , Senescência Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA/fisiologia , Feminino , Histonas/metabolismo , Homeostase , Linfopenia/etiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteases Específicas de Ubiquitina/deficiência , Proteases Específicas de Ubiquitina/genética , Ubiquitinação
13.
Cancer Cell ; 23(5): 660-76, 2013 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-23680149

RESUMO

In mouse and human neural progenitor and glioblastoma "stem-like" cells, we identified key targets of the Polycomb-group protein BMI1 by combining ChIP-seq with in vivo RNAi screening. We discovered that Bmi1 is important in the cellular response to the transforming growth factor-ß/bone morphogenetic protein (TGF-ß/BMP) and endoplasmic reticulum (ER) stress pathways, in part converging on the Atf3 transcriptional repressor. We show that Atf3 is a tumor-suppressor gene inactivated in human glioblastoma multiforme together with Cbx7 and a few other candidates. Acting downstream of the ER stress and BMP pathways, ATF3 binds to cell-type-specific accessible chromatin preloaded with AP1 and participates in the inhibition of critical oncogenic networks. Our data support the feasibility of combining ChIP-seq and RNAi screens in solid tumors and highlight multiple p16(INK4a)/p19(ARF)-independent functions for Bmi1 in development and cancer.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Estresse do Retículo Endoplasmático , Glioblastoma/genética , Células-Tronco Neoplásicas/metabolismo , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator 3 Ativador da Transcrição/análise , Fator 3 Ativador da Transcrição/genética , Fator 3 Ativador da Transcrição/fisiologia , Animais , Núcleo Celular/metabolismo , Cromatina/metabolismo , Homeostase/genética , Humanos , Camundongos , Células-Tronco Neurais/metabolismo , Complexo Repressor Polycomb 1/química , Complexo Repressor Polycomb 1/fisiologia , Proteínas Proto-Oncogênicas/química , Interferência de RNA , Transdução de Sinais
14.
J Control Release ; 163(3): 293-303, 2012 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-23041543

RESUMO

Efficient endocytosis into a wide range of target cells and low toxicity make the arginine-rich Tat peptide (Tat(11): YGRKKRRQRRR, residues 47-57 of HIV-1 Tat protein) an excellent transporter for delivery purposes. Unfortunately, molecules taken up by endocytosis undergo endosomal entrapment and possible metabolic degradation. Escape from the endosome is therefore actively researched. In this context, antimicrobial peptides (AMPs) provide viable templates for the design of new membrane-disruptive motifs. In particular the Cecropin-A and Melittin hybrids (CMs) are among the smallest and most effective peptides with membrane-perturbing abilities. Here we present a novel chimeric peptide in which the Tat(11) motif is fused to the CM(18) hybrid (KWKLFKKIGAVLKVLTTG, residues 1-7 of Cecropin-A and 2-12 of Melittin). When administered to cells, CM(18)-Tat(11) combines the two desired functionalities: efficient uptake and destabilization of endocytotic-vesicle membranes. We show that this chimeric peptide effectively increases cargo-molecule cytoplasm availability and allows the subsequent intracellular localization of diverse membrane-impermeable molecules (i.e. Tat(11)-EGFP fusion protein, calcein, dextrans, and plasmidic DNA) with no detectable cytotoxicity. The present results open the way to the rational engineering of "modular" cell-penetrating peptides (CPPs) that combine (i) efficient translocation from the extracellular milieu into vesicles and (ii) efficient release of molecules from vesicles into the cytoplasm.


Assuntos
Peptídeos Catiônicos Antimicrobianos/administração & dosagem , Membrana Celular/efeitos dos fármacos , Peptídeos Penetradores de Células/administração & dosagem , Animais , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cricetulus , DNA/metabolismo , Dextranos/metabolismo , Endocitose/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Fluoresceínas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Staphylococcus aureus/efeitos dos fármacos , Transfecção
15.
Anal Bioanal Chem ; 403(8): 2339-51, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22585053

RESUMO

Fluorescence recovery after photobleaching (FRAP) can help unveil subtle dynamical and biochemical properties of intracellular components. A peculiar aspect of this method is that it is based on the change of optical properties only, whereas dynamics and biochemistry of the molecules of interest are not perturbed. This makes FRAP particularly suitable for the study of protein translocation, e.g., between nucleus and cytoplasm. Here we present a comprehensive theoretical treatment of FRAP applied to protein nucleocytoplasmic translocation by passive diffusion and/or energy-driven processes across the nuclear envelope. Our mathematical model is validated by experimental FRAP studies with functionalized fluorescent protein chimeras. Using this approach we demonstrate that molecular crowding at the nuclear pore does not hamper passive diffusion and calculate the dimension of the nuclear pore size (5.33 nm). Additionally, our FRAP analysis reveals the biochemical parameters (maximum translocation rate and dissociation constant of the transport complex in cytoplasm) associated with the active import of a prototypical nuclear localization sequence (NLS of SV40) and related mutants. We demonstrate that transportin binding and active import into the nucleus are independent processes that can be separately modulated. The present results are discussed in light of their potential to help in engineering sequences for intracellular targeted delivery of sensors and/or therapeutic compounds. Finally, the limits of validity of our mathematical model are addressed.


Assuntos
Núcleo Celular/metabolismo , Citoplasma/metabolismo , Recuperação de Fluorescência Após Fotodegradação/métodos , Proteínas de Fluorescência Verde/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Células CHO , Cricetinae , Difusão , Carioferinas/metabolismo , Modelos Biológicos , Sinais de Localização Nuclear/metabolismo , Poro Nuclear/metabolismo
16.
J Biol Chem ; 287(8): 5554-61, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22190681

RESUMO

A quantitative description of carrier-mediated nuclear export in live cells is presented. To this end, we fused a prototypical leucine-rich nuclear export signal (NES) to GFP as a cargo model and expressed the fluorescent chimera in live CHO-K1 cells. By modeling FRAP data, we calculate the NES affinity for the export machinery and the maximum rate of nuclear export achievable at saturation of endogenous carriers. The measured active-export time through the Nuclear Pore Complex (NPC) is 18 ms, remarkably similar to the previously determined active-import rate. Also, our results reveal that active export/import and active export/passive diffusion fluxes are uncoupled, thus complementing previous reports on active import/passive diffusion uncoupling. These findings suggest differential gating at the NPC level.


Assuntos
Núcleo Celular/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Transporte Ativo do Núcleo Celular , Animais , Células CHO , Cricetinae , Cricetulus , Difusão , Cinética , Modelos Biológicos , Sinais de Exportação Nuclear , Sinais de Localização Nuclear , Poro Nuclear/metabolismo , Termodinâmica
17.
J Biol Chem ; 286(14): 12292-9, 2011 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-21321119

RESUMO

A detailed study of nuclear import mediated by the HIV-1 Tat peptide (47YGRKKRRQRRR57, TatRRR) is reported. Fluorescence-based measurements, calibration of protein concentrations, and binding assays are exploited to address the physicochemical mechanisms of Tat peptide recognition by the classical importin α (Impα) and importin ß (Impß) receptors both in vitro and in intact cells. We show that TatRRR is an unconventional nuclear localization sequence that binds directly to both Impα and Impß carriers in the absence of competitors (in vitro), whereas this property is silenced in the actual cellular environment. In the latter case, Impα/ß-dependent nuclear import can be successfully restored by replacing the "RRR" stretch with "GGG". We apply a recently developed method to determine quantitatively TatGGG affinity for each receptor. Based on these results, we can rationalize previous controversial reports on the Tat peptide and provide coherent guidelines for the design of novel intracellular targeting sequences.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Fragmentos de Peptídeos/metabolismo , alfa Carioferinas/metabolismo , beta Carioferinas/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Transporte Ativo do Núcleo Celular/genética , Animais , Células CHO , Cricetinae , Cricetulus , Recuperação de Fluorescência Após Fotodegradação , Fragmentos de Peptídeos/química , Ligação Proteica , alfa Carioferinas/genética , beta Carioferinas/genética , Produtos do Gene tat do Vírus da Imunodeficiência Humana/química
18.
Mol Pharm ; 7(3): 680-8, 2010 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-20394437

RESUMO

The ability of dendrimers to cross cell membranes is of much interest for their application in drug and gene delivery. Recent studies demonstrate that dendrimers are capable to enter cells by endocytosis, but the intracellular pathway following their internalization remains controversial. In this study we use confocal fluorescence microscopy to elucidate the intracellular trafficking properties of PAMAM dendrimers with high spatial and temporal resolution in living HeLa cells. Macromolecules of different chemical functionality (neutral, cationic and lipidated), size (from G2 up to G6) and surface charge are investigated and their internalization properties correlated with the molecular structure. Toxicity and internalization data are discussed that allow the identification of dendrimers maximizing intracellular uptake with the minimum effect on cell viability. Time-lapse imaging and colocalization assays with fluorescent biomarkers for endocytic vesicles demonstrate that dendrimers are internalized by both clathrin-dependent endocytosis and macropinocytosis and are eventually delivered to the lysosomal compartment. Moreover we analyzed the uptake of dendrimers in additional cell lines of practical interest for therapeutic purposes. These measurements together with a direct comparison with TAT peptides demonstrate that PAMAM dendrimers possess similar properties to these widely used cell-penetrating peptides and thanks to their chemical tunability may represent a valid alternative for drug and gene delivery.


Assuntos
Dendrímeros/metabolismo , Animais , Transporte Biológico/fisiologia , Linhagem Celular , Cromatografia em Gel , Dendrímeros/química , Endocitose/fisiologia , Citometria de Fluxo , Células HeLa , Células Hep G2 , Humanos , Microscopia de Fluorescência , Células PC12 , Ratos , Espectrometria de Fluorescência
19.
Biomaterials ; 31(17): 4682-94, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20304485

RESUMO

Interaction between differentiating neurons and the extracellular environment guides the establishment of cell polarity during nervous system development. Developing neurons read the physical properties of the local substrate in a contact-dependent manner and retrieve essential guidance cues. In previous works we demonstrated that PC12 cell interaction with nanogratings (alternating lines of ridges and grooves of submicron size) promotes bipolarity and alignment to the substrate topography. Here, we investigate the role of focal adhesions, cell contractility, and actin dynamics in this process. Exploiting nanoimprint lithography techniques and a cyclic olefin copolymer, we engineered biocompatible nanostructured substrates designed for high-resolution live-cell microscopy. Our results reveal that neuronal polarization and contact guidance are based on a geometrical constraint of focal adhesions resulting in an angular modulation of their maturation and persistence. We report on ROCK1/2-myosin-II pathway activity and demonstrate that ROCK-mediated contractility contributes to polarity selection during neuronal differentiation. Importantly, the selection process confined the generation of actin-supported membrane protrusions and the initiation of new neurites at the poles. Maintenance of the established polarity was independent from NGF stimulation. Altogether our results imply that focal adhesions and cell contractility stably link the topographical configuration of the extracellular environment to a corresponding neuronal polarity state.


Assuntos
Polaridade Celular/fisiologia , Adesões Focais/fisiologia , Neurônios/citologia , Actinas/metabolismo , Animais , Diferenciação Celular/fisiologia , Adesões Focais/metabolismo , Microscopia Confocal , Células PC12 , Ratos , Quinases Associadas a rho/metabolismo
20.
J Am Chem Soc ; 132(1): 85-95, 2010 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-19958004

RESUMO

Reversibly photoswitchable (i.e., photochromic) fluorescent proteins open the way to a number of advanced bioimaging techniques applicable to living-cell studies such as sequential photolabeling of distinct cellular regions, innovative FRET schemes, or nanoscopy. Owing to the relevance of fluorescent proteins from Aequorea victoria (AFPs) for cell biology, a photochromic "toolbox" constituted by several AFPs is highly desirable. Here we introduce four new photochromic AFPs whose reversible photoswitching occurs between the native bright and a dark state at low illumination power, on account of a very efficient cis-trans photoisomerization. Most remarkably, the optical bistability of these AFPs derives from the single E222Q mutation in the primary sequence. Apparently, the E222Q substitution can restore the intrinsic photochromic behavior of the isolated chromophore. The significance of these mutants for high-resolution in vivo cell imaging is shown by means of photochromic FRET experiments.


Assuntos
Substituição de Aminoácidos , Hidrozoários , Luz , Proteínas Luminescentes/química , Proteínas Luminescentes/metabolismo , Absorção , Animais , Cor , Escuridão , Transferência Ressonante de Energia de Fluorescência , Proteínas Luminescentes/genética , Modelos Moleculares , Imagem Molecular , Mutação , Fotodegradação , Conformação Proteica/efeitos da radiação , Dobramento de Proteína , Prótons , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA