Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 151(7): 1474-87, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23260137

RESUMO

DNA Ligase IV is responsible for sealing of double-strand breaks (DSBs) during nonhomologous end-joining (NHEJ). Inhibiting Ligase IV could result in amassing of DSBs, thereby serving as a strategy toward treatment of cancer. Here, we identify a molecule, SCR7 that inhibits joining of DSBs in cell-free repair system. SCR7 blocks Ligase IV-mediated joining by interfering with its DNA binding but not that of T4 DNA Ligase or Ligase I. SCR7 inhibits NHEJ in a Ligase IV-dependent manner within cells, and activates the intrinsic apoptotic pathway. More importantly, SCR7 impedes tumor progression in mouse models and when coadministered with DSB-inducing therapeutic modalities enhances their sensitivity significantly. This inhibitor to target NHEJ offers a strategy toward the treatment of cancer and improvement of existing regimens.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , DNA Ligases/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Pirimidinas/uso terapêutico , Bases de Schiff/uso terapêutico , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , DNA Ligase Dependente de ATP , DNA Ligases/química , DNA Ligases/genética , Modelos Animais de Doenças , Desenho de Fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Linfócitos/efeitos dos fármacos , Linfoma/tratamento farmacológico , Linfoma/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Pirimidinas/síntese química , Pirimidinas/química , Tolerância a Radiação , Ratos , Bases de Schiff/síntese química , Bases de Schiff/química , Alinhamento de Sequência
2.
Sensors (Basel) ; 23(17)2023 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-37687774

RESUMO

This study demonstrates the development of a humanized luciferase imaging reporter based on a recently discovered mushroom luciferase (Luz) from Neonothopanus nambi. In vitro and in vivo assessments showed that human-codon-optimized Luz (hLuz) has significantly higher activity than native Luz in various cancer cell types. The potential of hLuz in non-invasive bioluminescence imaging was demonstrated by human tumor xenografts subcutaneously and by the orthotopic lungs xenograft in immunocompromised mice. Luz enzyme or its unique 3OH-hispidin substrate was found to be non-cross-reacting with commonly used luciferase reporters such as Firefly (FLuc2), Renilla (RLuc), or nano-luciferase (NLuc). Based on this feature, a non-overlapping, multiplex luciferase assay using hLuz was envisioned to surpass the limitation of dual reporter assay. Multiplex reporter functionality was demonstrated by designing a new sensor construct to measure the NF-κB transcriptional activity using hLuz and utilized in conjunction with two available constructs, p53-NLuc and PIK3CA promoter-FLuc2. By expressing these constructs in the A2780 cell line, we unveiled a complex macromolecular regulation of high relevance in ovarian cancer. The assays performed elucidated the direct regulatory action of p53 or NF-κB on the PIK3CA promoter. However, only the multiplexed assessment revealed further complexities as stabilized p53 expression attenuates NF-κB transcriptional activity and thereby indirectly influences its regulation on the PIK3CA gene. Thus, this study suggests the importance of live cell multiplexed measurement of gene regulatory function using more than two luciferases to address more realistic situations in disease biology.


Assuntos
Agaricales , Neoplasias Ovarianas , Humanos , Animais , Feminino , Camundongos , NF-kappa B , Linhagem Celular Tumoral , Proteína Supressora de Tumor p53/genética , Luciferases/genética , Agaricales/genética , Classe I de Fosfatidilinositol 3-Quinases
3.
J Cell Sci ; 132(20)2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31455607

RESUMO

Chasing an intriguing biological question on the disparity of sodium iodide symporter (NIS, officially known as SLC5A5) expression and function in the clinical scenario of breast cancer, this study addresses key molecular defects involved. NIS in cancer patients has primarily been recorded to be a cytoplasmic protein, thus limiting the scope for targeted radio-iodine therapy. We developed NIS transgene-overexpressing MCF-7 breast cancer cells, and found a few clonal derivatives that show predominant expression of NIS in the plasma membrane. The majority of clones, however, showed cytosolic NIS expression over long passages. Cells expressing membranous NIS show unperturbed dynamic trafficking of NIS through secretory pathway organelles when compared to cells expressing cytoplasmic NIS or to parental cells. Further, treatment of cells expressing membranous NIS with specific glycosylation inhibitors highlighted the importance of inherent glycosylation processing and an 84 gene signature glycosylation RT-Profiler array revealed that clones expressing NIS in their membrane cluster separately compared to the other cells. We further confirm a role of three differentially expressed genes, i.e. MAN1B1, MAN1A1 and MAN2A1, in regulating NIS localization by RNA interference. Thus, this study shows the important role of mannosidase in N-glycosylation processing in order to correctly traffic NIS to the plasma membrane in breast cancer cells.This article has an associated First Person interview with the first author of the paper.


Assuntos
Neoplasias da Mama/metabolismo , Manose/metabolismo , Proteínas de Neoplasias/metabolismo , Simportadores/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Glicosilação , Humanos , Células MCF-7 , Manose/genética , Proteínas de Neoplasias/genética , Transporte Proteico , Simportadores/genética
4.
Exp Cell Res ; 396(2): 112313, 2020 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-33002501

RESUMO

Activation of STAT3 via Y705-phosphorylation is well documented across multiple cancer types and thus forms the basis of canonical pathway to judge STAT3 activation. Recently, important roles of two other post translational modification (PTM) sites, i.e. S727-phosphorylation and K685-acetylation, leading to STAT3 activation are reported. However, their critical mode of function in controlling STAT3 dimerization and signaling, independent of canonical activation remains elusive. Therefore, to understand the functional relevance of each STAT3 PTMs in breast cancer (BC), cell models are developed by stable overexpression of PTM-site specific point mutants, i.e. Y705F, S727A or K685R, in a 3'UTR-STAT3 knockdown BC cell background. Results using this model system reveal novel findings showing that phosphorylation at S727 can lead to STAT3 activation independent of phosphoY705. We also demonstrate that loss of pS727 or K685ac significantly affects functional phenotypes such as cell survival and proliferation as well as downstream transcriptional activity (Twist 1, Socs3, c-Myc, Bcl-1 and Mcl-1) of STAT3. Thereafter, by utilizing a BRET biosensor for measuring STAT3 phosphorylation in live cells, a crucial role of pS727 in dictating STAT3 activation and homodimerization formation is uncovered. Further by performing retrospective IHC analysis of total and phospho-forms of STAT3 in a cohort of 76 triple negative breast cancer (TNBC) patient samples, a significant dominant expression of phosphoS727 over phosphoY705 PTM (p < 0.001) is found in STAT3 positive cases. We also focus on validating known STAT3 inhibitor molecules for their action against both pY705 and pS727 activation. This study for the first time demonstrates that an anti-helminth drug compound, Niclosamide, is capable of inactivating both phospho-PTM sites on STAT3 and exhibits excellent anticancer efficacy in preclinical TNBC tumour model.


Assuntos
Neoplasias da Mama/metabolismo , Fosfosserina/metabolismo , Processamento de Proteína Pós-Traducional , Fator de Transcrição STAT3/metabolismo , Animais , Anti-Helmínticos/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Modelos Biológicos , Mutação/genética , Metástase Neoplásica , Niclosamida/farmacologia , Fosforilação/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Nanomedicine ; 37: 102437, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34273597

RESUMO

Photothermal-therapy (PTT) inculcates near-infrared laser guided local heating effect, where high degree of precision is expected, but not well proven to-date. An ex vivo tissue biochemical map with molecular/biochemical response showing the coverage area out of an optimized PTT procedure can reveal precision information. In this work, Raman-microscopic mapping and linear discriminant analysis of spectra of PTT treated and surrounding tissue areas ex vivo are done, revealing three distinct spectral clusters/zones, with minimal overlap between the core treated and adjacent untreated zone. The core treated zone showed intense nucleic-acid, cytochrome/mitochondria and protein damage, an adjacent zone showed lesser degree of damages and far zone showed minimal/no damage. Immunohistochemistry for γH2AX (DNA damage marker protein) in PTT exposed tissue also revealed similar results. Altogether, this study reveals the utility of Raman-microspectroscopy for fine-tuning safety parameters and precision that can be achieved from PTT mediated tumor ablation in preclinical/clinical application.


Assuntos
Nanopartículas Metálicas/química , Neoplasias/terapia , Terapia Fototérmica/métodos , Nanomedicina Teranóstica/tendências , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Ouro/química , Ouro/farmacologia , Histonas/genética , Humanos , Neoplasias/genética , Neoplasias/patologia , Análise Espectral Raman
6.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 196-208, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29055790

RESUMO

Acquired radioresistance accompanied with increased metastatic potential is a major hurdle in effective radiotherapy of breast cancers. However, the nature of their inter-dependence and the underlying mechanism remains largely intangible. By employing radioresistant (RR) cell lines, we herein demonstrate that MCF-7 RR cells display phenotypic and molecular alterations evocative of epithelial to mesenchymal transition (EMT) with increased traction forces and membrane ruffling culminating in boosted invasiveness. We then show that these changes can be attributed to overexpression of alpha-actinin-4 (ACTN4), with ACTN4 knockdown near-completely abrogating both radioresistance and EMT-associated changes. We further found that in MCF-7 RR cells, ACTN4 mediates the observed effects by activating AKT, and downstream AKT/GSK3ß signalling. Though ACTN4 plays a similar role in mediating radioresistance and invasiveness in MDA-MB-231 RR cells, co-immunoprecipitation studies reveal that these changes are effected through increased association with AKT and not by overexpression of AKT. Taken together, our study identifies ACTN4/AKT/GSK3ß as a novel pathway regulating radioresistance coupled invasion which can be further explored to improve the radiotherapeutic gain.


Assuntos
Actinina/fisiologia , Neoplasias da Mama/patologia , Movimento Celular/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tolerância a Radiação/genética , Actinina/genética , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Humanos , Células MCF-7 , Invasividade Neoplásica , Transdução de Sinais/genética
7.
Biochim Biophys Acta Mol Cell Res ; 1865(2): 392-405, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29175377

RESUMO

The failure of chemotherapeutic drugs in treatment of various cancers is attributed to the acquisition of drug resistance. However, the migration mechanisms of drug-resistant cancer cells remain incompletely understood. Here we address this question from a biophysical perspective by mapping the phenotypic alterations in ovarian cancer cells (OCCs) resistant to cisplatin and paclitaxel. We show that cisplatin-resistant (CisR), paclitaxel-resistant (PacR) and dual drug-resistant (i.e., resistant to both drugs) OCCs are more contractile and softer than drug-sensitive cells. Protease inhibition suppresses invasion of CisR cells but not of PacR cells, indicative of a protease-dependent mode of migration in CisR cells and a protease-independent mode of migration in PacR. Despite these differences, actomyosin contractility, mediated by the RhoA-ROCK2-Myosin II signaling pathway, regulates both modes of migration. Confined migration experiments establish the role of myosin IIA and IIB in mediating nuclear translocation and regulation of proteolytic activity. Collectively, our results highlight the importance of myosin II as a potential therapeutic target for treatment of drug-resistant ovarian cancer cells.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Miosina Tipo II/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Miosina Tipo II/genética , Proteínas de Neoplasias/genética , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia
8.
Langmuir ; 35(24): 7805-7815, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-31090425

RESUMO

Integrating the concept of biodegradation and light-triggered localized therapy in a functional nanoformulation is the current approach in onco-nanomedicine. Morphology control with an enhanced photothermal response, minimal toxicity, and X-ray attenuation of polymer-based nanoparticles is a critical concern for image-guided photothermal therapy. Herein, we describe the simple design of cost-effective and degradable polycaprolactone-based plasmonic nanoshells for the integrated photothermolysis as well as localized imaging of cancer cells. The gold-deposited polycaprolactone-based plasmonic nanoshells (AuPCL NS) are synthesized in a scalable and facile way under ambient conditions. The synthesized nanoshells are monodisperse, fairly stable, and highly inert even at five times (250 µg/mL) the therapeutic concentration in a week-long test. AuPCL NS are capable of delivering standalone photothermal therapy for the complete ablation of cancer cells without using any anticancerous drugs and causing toxicity. It delivers the same therapeutic efficacy to different cancer cell lines, irrespective of their chemorefractory status and also works as a potential computed tomography contrast agent for the integrated imaging-directed photothermal cancer therapy. High biocompatibility, degradability, and promising photothermal efficacy of AuPCL NS are attractive aspects of this report that could open new horizons of localized plasmonic photothermal therapy for healthcare applications.


Assuntos
Nanomedicina/economia , Nanomedicina/métodos , Nanoconchas/uso terapêutico , Fototerapia/economia , Fototerapia/métodos , Animais , Linhagem Celular Tumoral , Análise Custo-Benefício , Humanos , Hipertermia Induzida , Polímeros/química
9.
Proc Natl Acad Sci U S A ; 112(26): E3374-83, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26080397

RESUMO

Pre-mRNA splicing is a complex regulatory nexus modulated by various trans-factors and their posttranslational modifications to create a dynamic transcriptome through alternative splicing. Signal-induced phosphorylation and dephosphorylation of trans-factors are known to regulate alternative splicing. However, the role of other posttranslational modifications, such as deacetylation/acetylation, methylation, and ubiquitination, that could modulate alternative splicing in either a signal-dependent or -independent manner remain enigmatic. Here, we demonstrate that Scaffold/matrix-associated region-binding protein 1 (SMAR1) negatively regulates alternative splicing through histone deacetylase 6 (HDAC6)-mediated deacetylation of RNA-binding protein Sam68 (Src-associated substrate during mitosis of 68 kDa). SMAR1 is enriched in nuclear splicing speckles and associates with the snRNAs that are involved in splice site recognition. ERK-MAPK pathway that regulates alternative splicing facilitates ERK-1/2-mediated phosphorylation of SMAR1 at threonines 345 and 360 and localizes SMAR1 to the cytoplasm, preventing its interaction with Sam68. We showed that endogenously, SMAR1 through HDAC6 maintains Sam68 in a deacetylated state. However, knockdown or ERK-mediated phosphorylation of SMAR1 releases the inhibitory SMAR1-HDAC6-Sam68 complex, facilitating Sam68 acetylation and alternative splicing. Furthermore, loss of heterozygosity at the Chr.16q24.3 locus in breast cancer cells, wherein the human homolog of SMAR1 (BANP) has been mapped, enhances Sam68 acetylation and CD44 variant exon inclusion. In addition, tail-vein injections in mice with human breast cancer MCF-7 cells depleted for SMAR1 showed increased CD44 variant exon inclusion and concomitant metastatic propensity, confirming the functional role of SMAR1 in regulation of alternative splicing. Thus, our results reveal the complex molecular mechanism underlying SMAR1-mediated signal-dependent and -independent regulation of alternative splicing via Sam68 deacetylation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Processamento Alternativo/fisiologia , Proteínas de Ciclo Celular/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Histona Desacetilases/metabolismo , Matriz Nuclear/metabolismo , Proteínas Nucleares/fisiologia , Proteínas de Ligação a RNA/metabolismo , Acetilação , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Citoplasma/metabolismo , Proteínas de Ligação a DNA/genética , Técnicas de Silenciamento de Genes , Desacetilase 6 de Histona , Humanos , Receptores de Hialuronatos/genética , Sistema de Sinalização das MAP Quinases , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transporte Proteico
10.
Breast Cancer Res Treat ; 164(3): 603-615, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28528452

RESUMO

PURPOSE: Aberrant expression of human sodium iodide symporter (NIS) in breast cancer (BC) is well documented but the transcription factors (TF) regulating its aberrant expression is poorly known. We identify the presence of three p53 binding sites on the human NIS promoter sequence by conducting genome-wide TF analysis, and further investigate their regulatory role. METHODS: The differences in transcription and translation were measured by real-time PCR, luciferase reporter assay, site-directed mutagenesis, in vivo optical imaging, and chromatin immunoprecipitation. The relation of NIS and p53 in clinical samples was judged by TCGA data analysis and immunohistochemistry. RESULTS: Overexpression of wild-type p53 as a transgene or pharmacological activation by doxorubicin drug treatment shows significant suppression of NIS transcription in multiple BC cell types which also results in lowered NIS protein content and cellular iodide intake. NIS repression by activated p53 is further confirmed by non-invasive bioluminescence imaging in live cell and orthotropic tumor model. Abrogation of p53-binding sites by directional mutagenesis confirms reversal of transcriptional activity in wild-type p53-positive BC cells. We also observe direct binding of p53 to these sites on the human NIS promoter. Importantly, TCGA data analysis of NIS and p53 co-expression registers an inverse relationship between the two candidates. CONCLUSION: Our data for the first time highlight the role of p53 as a negative regulator of functional NIS expression in BC, where the latter is a potential targeted radioiodine therapy candidate. Thus, the study provides an important insight into prospective clinical application of this approach that may significantly impact the patient with mutant versus wild-type p53 profile.


Assuntos
Neoplasias da Mama/genética , Simportadores/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Animais , Sítios de Ligação , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Mutagênese Sítio-Dirigida , Transplante de Neoplasias , Regiões Promotoras Genéticas , Simportadores/química , Simportadores/metabolismo
11.
Int J Mol Sci ; 18(5)2017 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-28452928

RESUMO

The unresolved and paramount challenge in bio-imaging and targeted therapy is to clearly define and demarcate the physical margins of tumor tissue. The ability to outline the healthy vital tissues to be carefully navigated with transection while an intraoperative surgery procedure is performed sets up a necessary and under-researched goal. To achieve the aforementioned objectives, there is a need to optimize design considerations in order to not only obtain an effective imaging agent but to also achieve attributes like favorable water solubility, biocompatibility, high molecular brightness, and a tissue specific targeting approach. The emergence of near infra-red fluorescence (NIRF) light for tissue scale imaging owes to the provision of highly specific images of the target organ. The special characteristics of near infra-red window such as minimal auto-fluorescence, low light scattering, and absorption of biomolecules in tissue converge to form an attractive modality for cancer imaging. Imparting molecular fluorescence as an exogenous contrast agent is the most beneficial attribute of NIRF light as a clinical imaging technology. Additionally, many such agents also display therapeutic potentials as photo-thermal agents, thus meeting the dual purpose of imaging and therapy. Here, we primarily discuss molecular imaging and therapeutic potentials of two such classes of materials, i.e., inorganic NIR dyes and metallic gold nanoparticle based materials.


Assuntos
Nanoestruturas/química , Neoplasias/diagnóstico por imagem , Espectroscopia de Luz Próxima ao Infravermelho , Portadores de Fármacos/química , Corantes Fluorescentes/química , Corantes Fluorescentes/uso terapêutico , Humanos , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Nanoestruturas/uso terapêutico , Neoplasias/tratamento farmacológico , Fotoquimioterapia , Fototerapia
12.
Nano Lett ; 15(2): 842-8, 2015 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-25554860

RESUMO

We report biodegradable plasmon resonant liposome gold nanoparticles (LiposAu NPs) capable of killing cancer cells through photothermal therapy. The pharmacokinetic study of LiposAu NPs performed in a small animal model indicates in situ degradation in hepatocytes and further getting cleared through the hepato-biliary and renal route. Further, the therapeutic potential of LiposAu NPs tested in mouse tumor xenograft model using NIR laser (750 nm) illumination resulted in complete ablation of the tumor mass, thus prolonging disease-free survival.


Assuntos
Materiais Biocompatíveis , Ouro/química , Hipertermia Induzida , Lipossomos , Nanopartículas Metálicas/uso terapêutico , Neoplasias/terapia , Fototerapia , Animais , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Biol Chem ; 289(37): 25431-44, 2014 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-25086032

RESUMO

The evolution of the cancer cell into a metastatic entity is the major cause of death in patients with cancer. It has been acknowledged that aberrant activation of a latent embryonic program, known as the epithelial-mesenchymal transition (EMT), can endow cancer cells with the migratory and invasive capabilities associated with metastatic competence for which E-cadherin switch is a well-established hallmark. Discerning the molecular mechanisms that regulate E-cadherin expression is therefore critical for understanding tumor invasiveness and metastasis. Here we report that SMAR1 overexpression inhibits EMT and decelerates the migratory potential of breast cancer cells by up-regulating E-cadherin in a bidirectional manner. While SMAR1-dependent transcriptional repression of Slug by direct recruitment of SMAR1/HDAC1 complex to the matrix attachment region site present in the Slug promoter restores E-cadherin expression, SMAR1 also hinders E-cadherin-MDM2 interaction thereby reducing ubiquitination and degradation of E-cadherin protein. Consistently, siRNA knockdown of SMAR1 expression in these breast cancer cells results in a coordinative action of Slug-mediated repression of E-cadherin transcription, as well as degradation of E-cadherin protein through MDM2, up-regulating breast cancer cell migration. These results indicate a crucial role for SMAR1 in restraining breast cancer cell migration and suggest the candidature of this scaffold matrix-associated region-binding protein as a tumor suppressor.


Assuntos
Neoplasias da Mama/genética , Caderinas/biossíntese , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ligação a DNA/biossíntese , Transição Epitelial-Mesenquimal/genética , Proteínas Nucleares/biossíntese , Neoplasias da Mama/patologia , Caderinas/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Humanos , Metástase Neoplásica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteólise , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Fatores de Transcrição da Família Snail , Fatores de Transcrição/genética
14.
Proc Natl Acad Sci U S A ; 108(29): 12060-5, 2011 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-21730157

RESUMO

Identifying protein-protein interactions (PPIs) is essential for understanding various disease mechanisms and developing new therapeutic approaches. Current methods for assaying cellular intermolecular interactions are mainly used for cells in culture and have limited use for the noninvasive assessment of small animal disease models. Here, we describe red light-emitting reporter systems based on bioluminescence resonance energy transfer (BRET) that allow for assaying PPIs both in cell culture and deep tissues of small animals. These BRET systems consist of the recently developed Renilla reniformis luciferase (RLuc) variants RLuc8 and RLuc8.6, used as BRET donors, combined with two red fluorescent proteins, TagRFP and TurboFP635, as BRET acceptors. In addition to the native coelenterazine luciferase substrate, we used the synthetic derivative coelenterazine-v, which further red-shifts the emission maxima of Renilla luciferases by 35 nm. We show the use of these BRET systems for ratiometric imaging of both cells in culture and deep-tissue small animal tumor models and validate their applicability for studying PPIs in mice in the context of rapamycin-induced FK506 binding protein 12 (FKBP12)-FKBP12 rapamycin binding domain (FRB) association. These red light-emitting BRET systems have great potential for investigating PPIs in the context of drug screening and target validation applications.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Luciferases de Renilla , Proteínas Luminescentes , Ligação Proteica , Serina-Treonina Quinases TOR/metabolismo , Proteína 1A de Ligação a Tacrolimo/metabolismo , Animais , Western Blotting , Avaliação Pré-Clínica de Medicamentos/métodos , Imidazóis , Camundongos , Plasmídeos/genética , Pirazinas , Proteína Vermelha Fluorescente
15.
ACS Pharmacol Transl Sci ; 7(2): 375-383, 2024 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-38357276

RESUMO

Noninvasive, real-time, longitudinal imaging of protein functions in living systems with unprecedented specificity is one of the critical challenges of modern biomedical research. Toward that goal, here, we report a platform fusion technology called activity-based protein profiling-bioluminescence resonance energy transfer (ABPP-BRET). This method provides an opportunity to study the post-translational modification of a target protein in real time in living systems in a longitudinal manner. This semisynthetic BRET biosensor method is used for target engagement studies and further for inhibitor profiling in live cells. The simplicity of this method coupled with the critical physical distance-dependent BRET readout turned out to be a powerful method, thus pushing the activity-based protein profiling technology to the next level.

16.
Heliyon ; 10(4): e25553, 2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38384550

RESUMO

Bioluminescence resonance energy transfer (BRET) is one of the most promising approaches used for noninvasive imaging of protein-protein interactions in vivo. Recently, our team has discovered a genetically encodable bioluminescent system from the fungus Neonothopanus nambi and identified a novel luciferase that represents an imaging tool orthogonal to other luciferin-luciferase systems. We demonstrated the possibility of using the fungal luciferase as a new BRET donor by creating fused pairs with acceptor red fluorescent proteins, of which tdTomato provided the highest BRET efficiency. Using this new BRET system, we also designed a mTOR pathway specific rapamycin biosensor by integrating the FRB and FKBP12 protein dimerization system. We demonstrated the specificity and efficacy of the new fungal luciferase-based BRET combination for application in mammalian cell culture that will provide the unique opportunity to perform multiplexed BRET assessment in the future.

17.
Int J Biol Macromol ; 242(Pt 2): 124765, 2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-37156315

RESUMO

Among the EGFR family of receptors, HER3 is considered as a pseudo-kinase which primarily interacts with HER2 in presence of heregulin-1ß. We identified two hotspot mutations i.e. G284R and D297Y and one double mutant HER2-S310F/HER3-G284R in breast cancer patients. Long term MDS (7.5 µs) revealed that HER3-D297Y and HER2-S310F:HER3-G284R do not allow the interaction with HER2 as these mutations cause dramatic conformational changes in its flanking regions. This results in formation of an unstable HER2-WT:HER3-D297Y heterodimer, thereby abrogating the downstream signalling by AKT. We found that His228 and Ser300 of HER3-D297Y form stable interactions with Glu245 and Tyr270 of EGFR-WT, in the presence of either EGF or heregulin-1ß. Applying TRIM-ing mediated direct knockdown of endogenous EGFR protein, specificity of the unconventional EGFR:HER3-D297Y interaction was validated. Due to this unusual ligand mediated interaction, cancer cells were found susceptible to EGFR targeted therapeutics i.e. Gefitinib and Erlotinib. Further, in TCGA analysis, BC patients harbouring HER3-D297Y mutation showed increased p-EGFR levels as compared to the patients harbouring HER3-WT and HER3-G284R mutations. For the first time, this comprehensive study showed the importance of specific hotspot mutations in HER3 dimerization domain can defy the Trastuzumab therapy, rather cells become susceptible to the EGFR inhibitors.


Assuntos
Neoplasias da Mama , Receptor ErbB-3 , Feminino , Humanos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Dimerização , Receptores ErbB/genética , Receptores ErbB/metabolismo , Mutação , Neuregulina-1/genética , Neuregulina-1/metabolismo , Neuregulina-1/farmacologia , Receptor ErbB-2/metabolismo , Receptor ErbB-3/genética , Receptor ErbB-3/metabolismo
18.
Explor Target Antitumor Ther ; 4(6): 1227-1248, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38213543

RESUMO

A generalized therapeutic strategy for various disease conditions, including cancer, is to deplete or inactivate harmful protein targets. Various forms of protein or gene silencing molecules, e.g., small molecule inhibitors, RNA interference (RNAi), and microRNAs (miRNAs) have been used against druggable targets. Over the past few years, targeted protein degradation (TPD) approaches have been developed for direct degradation of candidate proteins. Among the TPD approaches, proteolysis targeting chimeras (PROTACs) have emerged as one of the most promising approaches for the selective elimination of proteins via the ubiquitin-proteasome system. Other than PROTACs, TPD methods with potential therapeutic use include intrabody-mediated protein knockdown and tripartite motif-21 (TRIM-21) mediated TRIM-Away. In this review, protein knockdown approaches, their modes of action, and their advantages over conventional gene knockdown approaches are summarized. In cancers, disease-associated protein functions are often executed by specific post-translational modifications (PTMs). The role of TRIM-Away is highlighted in the direct knockdown of PTM forms of target proteins. Moreover, the application challenges and the prospective clinical use of TPD approaches in various diseases are also discussed.

19.
Nanoscale ; 15(3): 1273-1288, 2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36541678

RESUMO

Photothermal therapy (PTT) has emerged as a very potent therapeutic approach in the treatment of tumors. Gold nanoparticles have gained considerable scientific interest as a photosensitizer due to their absorbance in the near-infrared regions. However, their biodegradation and excretion from the body is a challenge. Various biodegradable systems consisting of liposomes and polymers have been synthesized, but their precise manufacturing and decomposition mechanisms have not yet been explored. Using zein nanoparticles as a template, we have fabricated a glutathione-functionalized gold core shell type of formulation. The scalability of the one-step seedless gold coating process is also reported. The synthesis procedure of these tunable nanoparticles is understood with TEM. The thermal degradation of the material under the physiological conditions is thoroughly examined using UV and TEM. In vitro PTT effectiveness on breast cancer cells is assessed after an extensive in vitro toxicity research. The mechanism of cell death is studied using ROS and cell cycle analysis. The material exhibited good efficacy as a PTT agent in mice and showed non-toxicity up to 14 days. The renal clearance study of the material in mice shows its disintegration into renal clearable minute gold seeds. All the findings suggest biodegradable glutathione-functionalized gold core-shell nanoparticles as potential photothermal cancer treatment agents.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Fotoquimioterapia , Animais , Camundongos , Ouro/farmacologia , Ouro/uso terapêutico , Nanopartículas Metálicas/uso terapêutico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia , Linhagem Celular Tumoral
20.
Arterioscler Thromb Vasc Biol ; 31(11): e72-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21836062

RESUMO

OBJECTIVE: Stem cell therapy for angiogenesis and vascular regeneration has been investigated using adult or embryonic stem cells. In the present study, we investigated the potential of endothelial cells (ECs) derived from human induced pluripotent stem cells (hiPSCs) to promote the perfusion of ischemic tissue in a murine model of peripheral arterial disease. METHODS AND RESULTS: Endothelial differentiation was initiated by culturing hiPSCs for 14 days in differentiation media supplemented with BMP-4 and vascular endothelial growth factor. The hiPSC-ECs exhibited endothelial characteristics by forming capillary-like structures in matrigel and incorporating acetylated-LDL. They stained positively for EC markers such as KDR, CD31, CD144, and eNOS. In vitro exposure of hiPSC-ECs to hypoxia resulted in increased expression of various angiogenic related cytokines and growth factors. hiPSC-ECs were stably transduced with a double fusion construct encoded by the ubiquitin promoter, firefly luciferase for bioluminescence imaging and green fluorescence protein for fluorescent detection. The hiPSC-ECs (5×10(5)) were delivered by intramuscular injection into the ischemic hindlimb of SCID mice at day 0 and again on day 7 after femoral artery ligation (n=8). Bioluminescence imaging showed that hiPSC-ECs survived in the ischemic limb for at least 2 weeks. In addition, laser Doppler imaging showed that the ratio of blood perfusion was increased by hiPSC-EC treatment by comparison to the saline-treated group (0.58±0.12 versus 0.44±0.04; P=0.005). The total number of capillaries in the ischemic limb of mice receiving hiPSC-EC injections was greater than those in the saline-treated group (1284±155 versus 797±206 capillaries/mm(2)) (P<0.002). CONCLUSION: This study is a first step toward development of a regenerative strategy for peripheral arterial disease based on the use of ECs derived from hiPSCs.


Assuntos
Capilares/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Endotélio Vascular/citologia , Endotélio Vascular/transplante , Doença Arterial Periférica/fisiopatologia , Doença Arterial Periférica/terapia , Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Membro Posterior/irrigação sanguínea , Humanos , Isquemia/terapia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neovascularização Fisiológica/fisiologia , Transplante de Células-Tronco/métodos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA