Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Microbiol Immunol ; 68(5): 165-178, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38444370

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic is an ongoing global public health crisis. The causative agent, the SARS-CoV-2 virus, enters host cells via molecular interactions between the viral spike protein and the host cell ACE2 surface protein. The SARS-CoV-2 spike protein is extensively decorated with up to 66 N-linked glycans. Glycosylation of viral proteins is known to function in immune evasion strategies but may also function in the molecular events of viral entry into host cells. Here, we show that N-glycosylation at Asn331 and Asn343 of SARS-CoV-2 spike protein is required for it to bind to ACE2 and for the entry of pseudovirus harboring the SARS-CoV-2 spike protein into cells. Interestingly, high-content glycan binding screening data have shown that N-glycosylation of Asn331 and Asn343 of the RBD is important for binding to the specific glycan molecule G4GN (Galß-1,4 GlcNAc), which is critical for spike-RBD-ACE2 binding. Furthermore, IL-6 was identified through antibody array analysis of conditioned media of the corresponding pseudovirus assay. Mutation of N-glycosylation of Asn331 and Asn343 sites of the spike receptor-binding domain (RBD) significantly reduced the transcriptional upregulation of pro-inflammatory signaling molecule IL-6. In addition, IL-6 levels correlated with spike protein levels in COVID-19 patients' serum. These findings establish the importance of RBD glycosylation in SARS-CoV-2 pathogenesis, which can be exploited for the development of novel therapeutics for COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Interleucina-6 , Ligação Proteica , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Internalização do Vírus , Glicoproteína da Espícula de Coronavírus/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Humanos , Glicosilação , Enzima de Conversão de Angiotensina 2/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/fisiologia , Interleucina-6/metabolismo , COVID-19/virologia , COVID-19/metabolismo , Células HEK293 , Asparagina/metabolismo , Polissacarídeos/metabolismo
2.
J Biol Chem ; 300(1): 105583, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38141770

RESUMO

Membrane polyphosphoinositides (PPIs) are lipid-signaling molecules that undergo metabolic turnover and influence a diverse range of cellular functions. PPIs regulate the activity and/or spatial localization of a number of actin-binding proteins (ABPs) through direct interactions; however, it is much less clear whether ABPs could also be an integral part in regulating PPI signaling. In this study, we show that ABP profilin1 (Pfn1) is an important molecular determinant of the cellular content of PI(4,5)P2 (the most abundant PPI in cells). In growth factor (EGF) stimulation setting, Pfn1 depletion does not impact PI(4,5)P2 hydrolysis but enhances plasma membrane (PM) enrichment of PPIs that are produced downstream of activated PI3-kinase, including PI(3,4,5)P3 and PI(3,4)P2, the latter consistent with increased PM recruitment of SH2-containing inositol 5' phosphatase (SHIP2) (a key enzyme for PI(3,4)P2 biosynthesis). Although Pfn1 binds to PPIs in vitro, our data suggest that Pfn1's affinity to PPIs and PM presence in actual cells, if at all, is negligible, suggesting that Pfn1 is unlikely to directly compete with SHIP2 for binding to PM PPIs. Additionally, we provide evidence for Pfn1's interaction with SHIP2 in cells and modulation of this interaction upon EGF stimulation, raising an alternative possibility of Pfn1 binding as a potential restrictive mechanism for PM recruitment of SHIP2. In conclusion, our findings challenge the dogma of Pfn1's binding to PM by PPI interaction, uncover a previously unrecognized role of Pfn1 in PI(4,5)P2 homeostasis and provide a new mechanistic avenue of how an ABP could potentially impact PI3K signaling byproducts in cells through lipid phosphatase control.


Assuntos
Fosfatidilinositóis , Profilinas , Fator de Crescimento Epidérmico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases/metabolismo , Fosfatidilinositóis/metabolismo , Humanos , Células HEK293 , Profilinas/metabolismo
3.
Comput Biol Chem ; 106: 107933, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37536229

RESUMO

This study aims to investigate the potential therapeutic application of Ixeridium dentatum (ID) in treating atopic dermatitis (AD) through network pharmacology, molecular docking, and molecular dynamic simulation. We employed GC-MS techniques and identified 40 bioactive compounds present in the ID and determined their targets by accessing public databases. The convergence of compounds and dermatitis related targets led to the identification of 32 common genes. Among them, IL1B, PTGS2, IL6, IL2, and RELA, were found to be significant targets which were analyzed using Cytoscape network topology. The KEGG pathway evaluation revealed that these targets were significantly enriched in the C-type lectin receptor signaling pathway. The therapeutic efficacy of Stigmasta-5,22-dien-3-ol, Urea, n-Heptyl-, and 3-Epimoretenol was demonstrated in molecular docking assay, as evidenced by their presence in the core compounds of the compound-target network. Furthermore, these compounds exhibited significant kinetic stability and chemical reactivity in DFT quantum analysis when compared to their co-crystallized ligands and reference drug, indicating their potential as key targets for future research. Among the top three docking complexes, namely IL6-3-Epimoretenol, and IL2- Stigmasta-5,22-dien-3-ol, both demonstrated exceptional dynamic characteristics in molecular dynamics simulations at 100 ns. The feasibility of these compounds could be attributed to the prior traditional interrelationship between ID and AD. Overall, this research elucidates the interplay between AD-associated signaling pathways and target receptors with the bioactive ID. The proposal posits the utilization of antecedent compounds as a substitute for the customary pharmaceutical intervention that obstructs the discharge of cytokines, which incite dermal inflammation in the C-type lectin receptor signaling pathway of atopic dermatitis.


Assuntos
Dermatite Atópica , Medicamentos de Ervas Chinesas , Humanos , Dermatite Atópica/tratamento farmacológico , Interleucina-2 , Interleucina-6 , Simulação de Acoplamento Molecular , Lectinas Tipo C
4.
J Nanobiotechnology ; 20(1): 393, 2022 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-36045375

RESUMO

High-quality point-of-care is critical for timely decision of disease diagnosis and healthcare management. In this regard, biosensors have revolutionized the field of rapid testing and screening, however, are confounded by several technical challenges including material cost, half-life, stability, site-specific targeting, analytes specificity, and detection sensitivity that affect the overall diagnostic potential and therapeutic profile. Despite their advances in point-of-care testing, very few classical biosensors have proven effective and commercially viable in situations of healthcare emergency including the recent COVID-19 pandemic. To overcome these challenges functionalized magnetic nanoparticles (MNPs) have emerged as key players in advancing the biomedical and healthcare sector with promising applications during the ongoing healthcare crises. This critical review focus on understanding recent developments in theranostic applications of functionalized magnetic nanoparticles (MNPs). Given the profound global economic and health burden, we discuss the therapeutic impact of functionalized MNPs in acute and chronic diseases like small RNA therapeutics, vascular diseases, neurological disorders, and cancer, as well as for COVID-19 testing. Lastly, we culminate with a futuristic perspective on the scope of this field and provide an insight into the emerging opportunities whose impact is anticipated to disrupt the healthcare industry.


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19 , Nanopartículas de Magnetita , Nanopartículas , COVID-19/diagnóstico , Teste para COVID-19 , Doença Crônica , Humanos , Nanopartículas de Magnetita/uso terapêutico , Nanomedicina , Pandemias
5.
Molecules ; 27(2)2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-35056661

RESUMO

Cell cycle kinases represent an important component of the cell machinery that controls signal transduction involved in cell proliferation, growth, and differentiation. Nek2 is a mitotic Ser/Thr kinase that localizes predominantly to centrosomes and kinetochores and orchestrates centrosome disjunction and faithful chromosomal segregation. Its activity is tightly regulated during the cell cycle with the help of other kinases and phosphatases and via proteasomal degradation. Increased levels of Nek2 kinase can promote centrosome amplification (CA), mitotic defects, chromosome instability (CIN), tumor growth, and cancer metastasis. While it remains a highly attractive target for the development of anti-cancer therapeutics, several new roles of the Nek2 enzyme have recently emerged: these include drug resistance, bone, ciliopathies, immune and kidney diseases, and parasitic diseases such as malaria. Therefore, Nek2 is at the interface of multiple cellular processes and can influence numerous cellular signaling networks. Herein, we provide a critical overview of Nek2 kinase biology and discuss the signaling roles it plays in both normal and diseased human physiology. While the majority of research efforts over the last two decades have focused on the roles of Nek2 kinase in tumor development and cancer metastasis, the signaling mechanisms involving the key players associated with several other notable human diseases are highlighted here. We summarize the efforts made so far to develop Nek2 inhibitory small molecules, illustrate their action modalities, and provide our opinion on the future of Nek2-targeted therapeutics. It is anticipated that the functional inhibition of Nek2 kinase will be a key strategy going forward in drug development, with applications across multiple human diseases.


Assuntos
Doenças Ósseas/patologia , Inibidores Enzimáticos/farmacologia , Doenças do Sistema Imunitário/patologia , Nefropatias/patologia , Malária/patologia , Quinases Relacionadas a NIMA/antagonistas & inibidores , Neoplasias/patologia , Doenças Ósseas/tratamento farmacológico , Doenças Ósseas/enzimologia , Resistência a Medicamentos , Humanos , Doenças do Sistema Imunitário/tratamento farmacológico , Doenças do Sistema Imunitário/enzimologia , Nefropatias/tratamento farmacológico , Nefropatias/enzimologia , Malária/tratamento farmacológico , Malária/enzimologia , Metástase Neoplásica , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia
6.
Bioorg Chem ; 117: 105463, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34753058

RESUMO

Human cathepsin B is a cysteine-dependent protease whose roles in both normal and diseased cellular states remain yet to be fully delineated. This is primarily due to overlapping substrate specificities and lack of unambiguously annotated physiological functions. In this work, a selective, cell-permeable, clickable and tagless small molecule cathepsin B probe, KDA-1, is developed and kinetically characterized. KDA-1 selectively targets active site Cys25 residue of cathepsin B for labeling and can detect active cellular cathepsin B in proteomes derived from live human MDA-MB-231 breast cancer cells and HEK293 cells. It is anticipated that KDA-1 probe will find suitable applications in functional proteomics involving human cathepsin B enzyme.


Assuntos
Catepsina B/química , Sondas Moleculares/química , Catepsina B/genética , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Sondas Moleculares/síntese química , Estrutura Molecular , Relação Estrutura-Atividade
7.
R Soc Open Sci ; 8(10): 211003, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34659780

RESUMO

Oil pollution is of increasing concern for environmental safety and the use of microbial surfactants in oil remediation has become inevitable for their efficacy and ecofriendly nature. In this work, biosurfactants of bacteria isolated from oil-contaminated soil have been characterized. Four potent biosurfactant-producing strains (SD4, SD11, SD12 and SD13) were selected from 27 isolates based on drop collapse assay and emulsification index, and identified as species belonging to Bacillus, Burkholderia, Providencia and Klebsiella, revealed from their 16S rRNA gene-based analysis. Detailed morphological and biochemical characteristics of each selected isolate were determined. Their growth conditions for maximum biosurfactant production were optimized and found quite similar among the four isolates with a pH of 3.0 and temperature 37°C after 6 or 7 days of growth on kerosene. The biosurfactants of SD4, SD11 and SD12 appeared to be glycolipids and that of SD13 a lipopeptide. Emulsification activity of most of the biosurfactants was stable at low and high temperatures (4-100°C), a wide range of pH (2-10) and salt concentrations (2-7% NaCl). Each biosurfactant showed antimicrobial activity against two or more pathogenic bacteria. The biosurfactants were well-capable of emulsifying kerosene, diesel and soya bean, and could efficiently degrade diesel.

8.
Anticancer Res ; 37(4): 1655-1663, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28373426

RESUMO

BACKGROUND/AIM: One of the major hurdles in the treatment of breast cancers is the inability of anti-cancer drugs to eliminate the breast cancer stem cells (BCSCs) population, which leads to disease relapse. The dearth in anti-cancer drugs that target BCSCs can be attributed to the absence of in vitro screening models that can not only recapitulate the tumor microenvironment consisting of BCSCs but also preserve the 3-dimensional (3D) architecture of in vivo tumors. MATERIALS AND METHODS: In our present study, we have developed a 3D cell culture system that shows: (i) enrichment of BCSCs, (ii) increased drug resistance, and (iii) generation of hypoxic conditions similar to tumors. RESULTS: Using this model, we were able to screen a FDA-approved diversity set and identify as well as validate actinomycin D as a potential anti-breast cancer agent. Interestingly, we show that actinomycin D specifically targets and down-regulates the expression of the stem cell transcription factor, Sox-2. Additionally, down-regulation of Sox-2 leads to depletion of the stem-cell population resulting in the inability of breast cancer cells to initiate tumor progression. CONCLUSION: This study demonstrates the utility of an in vivo-like 3D cell culture system for the identification and validation of anti-cancer agents that will have a better probability of success in the clinic.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Dactinomicina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Fatores de Transcrição SOXB1/antagonistas & inibidores , Antibióticos Antineoplásicos/farmacologia , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Células Tumorais Cultivadas
9.
Sci Rep ; 6: 34400, 2016 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-27694854

RESUMO

Analysis of individual cells at the subcellular level is important for understanding diseases and accelerating drug discovery. Nanoscale endoscopes allow minimally invasive probing of individual cell interiors. Several such instruments have been presented previously, but they are either too complex to fabricate or require sophisticated external detectors because of low signal collection efficiency. Here we present a nanoendoscope that can locally excite fluorescence in labelled cell organelles and collect the emitted signal for spectral analysis. Finite Difference Time Domain (FDTD) simulations have shown that with an optimized nanoendoscope taper profile, the light emission and collection was localized within ~100 nm. This allows signal detection to be used for nano-photonic sensing of the proximity of fluorophores. Upon insertion into the individual organelles of living cells, the nanoendoscope was fabricated and resultant fluorescent signals collected. This included the signal collection from the nucleus of Acridine orange labelled human fibroblast cells, the nucleus of Hoechst stained live liver cells and the mitochondria of MitoTracker Red labelled MDA-MB-231 cells. The endoscope was also inserted into a live organism, the yellow fluorescent protein producing nematode Caenorhabditis elegans, and a fluorescent signal was collected. To our knowledge this is the first demonstration of in vivo, local fluorescence signal collection on the sub-organelle level.


Assuntos
Caenorhabditis elegans/metabolismo , Mitocôndrias Hepáticas/metabolismo , Imagem Molecular/métodos , Nanotecnologia/métodos , Imagem Óptica/métodos , Animais , Linhagem Celular Tumoral , Endoscopia , Humanos
10.
Elife ; 52016 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-27371829

RESUMO

Mitosis occurs efficiently, but when it is disturbed or delayed, p53-dependent cell death or senescence is often triggered after mitotic exit. To characterize this process, we conducted CRISPR-mediated loss-of-function screens using a cell-based assay in which mitosis is consistently disturbed by centrosome loss. We identified 53BP1 and USP28 as essential components acting upstream of p53, evoking p21-dependent cell cycle arrest in response not only to centrosome loss, but also to other distinct defects causing prolonged mitosis. Intriguingly, 53BP1 mediates p53 activation independently of its DNA repair activity, but requiring its interacting protein USP28 that can directly deubiquitinate p53 in vitro and ectopically stabilize p53 in vivo. Moreover, 53BP1 can transduce prolonged mitosis to cell cycle arrest independently of the spindle assembly checkpoint (SAC), suggesting that while SAC protects mitotic accuracy by slowing down mitosis, 53BP1 and USP28 function in parallel to select against disturbed or delayed mitosis, promoting mitotic efficiency.


Assuntos
Pontos de Checagem do Ciclo Celular , Centrossomo/metabolismo , Mitose , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Ubiquitina Tiolesterase/metabolismo , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Marcação de Genes , Testes Genéticos , Humanos
11.
Sci Transl Med ; 8(327): 327ra24, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26912903

RESUMO

Conventional cancer treatment strategies assume that maximum patient benefit is achieved through maximum killing of tumor cells. However, by eliminating the therapy-sensitive population, this strategy accelerates emergence of resistant clones that proliferate unopposed by competitors-an evolutionary phenomenon termed "competitive release." We present an evolution-guided treatment strategy designed to maintain a stable population of chemosensitive cells that limit proliferation of resistant clones by exploiting the fitness cost of the resistant phenotype. We treated MDA-MB-231/luc triple-negative and MCF7 estrogen receptor-positive (ER(+)) breast cancers growing orthotopically in a mouse mammary fat pad with paclitaxel, using algorithms linked to tumor response monitored by magnetic resonance imaging. We found that initial control required more intensive therapy with regular application of drug to deflect the exponential tumor growth curve onto a plateau. Dose-skipping algorithms during this phase were less successful than variable dosing algorithms. However, once initial tumor control was achieved, it was maintained with progressively smaller drug doses. In 60 to 80% of animals, continued decline in tumor size permitted intervals as long as several weeks in which no treatment was necessary. Magnetic resonance images and histological analysis of tumors controlled by adaptive therapy demonstrated increased vascular density and less necrosis, suggesting that vascular normalization resulting from enforced stabilization of tumor volume may contribute to ongoing tumor control with lower drug doses. Our study demonstrates that an evolution-based therapeutic strategy using an available chemotherapeutic drug and conventional clinical imaging can prolong the progression-free survival in different preclinical models of breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Modelos Animais de Doenças , Feminino , Humanos , Imageamento por Ressonância Magnética , Camundongos , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Análise de Sobrevida , Resultado do Tratamento
12.
Int J Cancer ; 136(4): E188-96, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25156304

RESUMO

ATP-binding cassette (ABC) drug transporters consuming ATPs for drug efflux is a common mechanism by which clinical cancers develop multidrug resistance (MDR). We hypothesized that MDR phenotypes could be suppressed by administration of "ersatzdroges," nonchemotherapy drugs that are, nevertheless, ABC substrates. We reasoned that, through prolonged activation of the ABC pumps, ersatzdroges will force MDR cells to divert limited resources from proliferation and invasion thus delaying disease progression. We evaluated ABC substrates as ersatzdroge by comparing their effects on proliferation and survival of MDR cell lines (MCF-7/Dox and 8226/Dox40) with the effects on the drug-sensitive parental lines (MCF-7 and 8226/s, respectively) in glucose-limited condition. The changes in glucose and energy demands were also examined in vitro and in vivo. MCF-7/Dox showed higher ATP demand and susceptibility to glucose resource limitation. Ersatzdroges significantly decreased proliferation of MCF-7/Dox when the culture media contained physiological glucose concentrations (1.0 g/L) or less, but had no effect on MCF-7. Similar evidence was obtained from 8226/Dox40 and 8226/s comparison. In vivo 18F-FDG-PET imaging demonstrated that glucose uptake was increased by systemic administration of an ersatzdroge in tumors composed of MDR. These results suggest that administration of ersatzdroges, by increasing the metabolic cost of resistance, can suppress proliferation of drug-resistance phenotypes. This provides a novel and relatively simple application model of evolution-based strategy, which can exploit the cost of resistance to delay proliferation of drug-resistant cancer phenotypes. Furthermore, suggested is the potential of ersatzdroges to identify tumors or regions of tumors that express the MDR phenotype.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Antineoplásicos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Claritromicina/farmacologia , Claritromicina/uso terapêutico , Ciclosporina/farmacologia , Ciclosporina/uso terapêutico , Resistência a Múltiplos Medicamentos , Eritromicina/farmacologia , Eritromicina/uso terapêutico , Feminino , Expressão Gênica , Humanos , Células MCF-7 , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos Nus , Terapia de Alvo Molecular , Carga Tumoral , Verapamil/farmacologia , Verapamil/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Womens Health (Lond) ; 10(4): 423-30, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25259902

RESUMO

Conventional systemic therapy for disseminated breast cancer is based on the general assumption that the greatest patient benefit is achieved by killing the maximum number of tumor cells. While this strategy often achieves a significant reduction in tumor burden, most patients with metastatic breast cancer ultimately die from their disease as therapy fails because tumor cells evolve resistance. We propose that the conventional maximum dose/maximum cell kill cancer therapy, when viewed from an evolutionary vantage, is suboptimal and likely even harmful as it accelerates evolution and growth of the resistant phenotypes that ultimately cause patient death. As an alternative, we are investigating evolutionary therapeutic strategies that shift the treatment goal from killing the maximum number of cancer cells to maximizing patient survival. Here we introduce two novel approaches for systemic therapy for metastatic breast cancer, considering the evolutionary nature of tumor progression; adaptive therapy and double-bind therapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/fisiopatologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/biossíntese , Trifosfato de Adenosina/metabolismo , Animais , Neoplasias da Mama/patologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Metástase Neoplásica , Fenótipo , Receptor ErbB-2/biossíntese , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Fatores de Tempo
14.
Proc Natl Acad Sci U S A ; 107(50): 21547-52, 2010 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-21115820

RESUMO

Profilin1, a ubiquitously expressed actin-binding protein, plays a critical role in cell migration through actin cytoskeletal regulation. Given the traditional view of profilin1 as a promigratory molecule, it is difficult to reconcile observations that profilin1 is down-regulated in various invasive adenocarcinomas and that reduced profilin1 expression actually confers increased motility to certain adenocarcinoma cells. In this study, we show that profilin1 negatively regulates lamellipodin targeting to the leading edge in MDA-MB-231 breast cancer cells and normal cells; profilin1 depletion increases lamellipodin concentration at the lamellipodial tip (where it binds Ena/VASP), and this mediates the hypermotility. We report that the molecular mechanism underlying profilin1's modulation of lamellipodin localization relates to phosphoinositide control. Specifically, we show that phosphoinositide binding of profilin1 inhibits the motility of MDA-MB-231 cells by negatively regulating PI(3,4)P(2) at the membrane and thereby limiting recruitment of lamellipodin [a PI(3,4)P(2)-binding protein] and Ena/VASP to the leading edge. In summary, this study uncovers a unique biological consequence of profilin1-phosphoinositide interaction, thus providing direct evidence of profilin1's regulation of cell migration independent of its actin-related activity.


Assuntos
Proteínas de Transporte/metabolismo , Movimento Celular/fisiologia , Proteínas de Membrana/metabolismo , Fosfatidilinositóis/metabolismo , Profilinas/metabolismo , Proteínas de Transporte/genética , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Humanos , Proteínas de Membrana/genética , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Profilinas/genética
15.
J Cell Physiol ; 218(2): 436-43, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18937284

RESUMO

Profilin-1 (Pfn1), a ubiquitously expressed actin-binding protein, has been regarded as a tumor-suppressor molecule for breast cancer. Since AKT signaling impacts cell survival and proliferation, in this study we investigated whether AKT activation in breast cancer cells is sensitive to perturbation of Pfn1 expression. We found that even a moderate overexpression of Pfn1 leads to a significant reduction in phosphorylation of AKT in MDA-MB-231 breast cancer cells. We further demonstrated that Pfn1 overexpression in MDA-MB-231 cells is associated with a significant reduction in the level of the phosphoinositide regulator of AKT, PIP(3), and impaired membrane translocation of AKT that is required for AKT activation, in response to EGF stimulation. Interestingly, Pfn1-overexpressing cells showed post-transcriptional upregulation of PTEN. Furthermore, when PTEN expression was silenced, AKT phosphorylation was rescued, suggesting PTEN upregulation is responsible for Pfn1-dependent attenuation of AKT activation in MDA-MB-231 cells. Pfn1 overexpression induced PTEN upregulation and reduced AKT activation were also reproducible features of BT474 breast cancer cells. These findings may provide mechanistic insights underlying at least some of the tumor-suppressive properties of Pfn1.


Assuntos
Neoplasias da Mama/enzimologia , PTEN Fosfo-Hidrolase/genética , Profilinas/genética , Profilinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Regulação para Cima/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Membrana Celular/enzimologia , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Humanos , Fosfatos de Fosfatidilinositol/metabolismo
16.
Fertil Steril ; 87(2): 343-50, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17097647

RESUMO

OBJECTIVE: To evaluate the significance of an estrogen-upregulated 64.0-kDa human uterine fluid (hUF) glycoprotein in relation to promotion of fertility. DESIGN: Prospective study. SETTING: Department of Assisted Reproductive Biology Unit, Institute of Reproductive Medicine, Kolkata. India. PATIENT(S): Sixty-three women with unexplained infertility having normal ovulatory cycle with normal endocrine profile and 18 parous women. INTERVENTION(S): Women either received no stimulation (n = 35) or were stimulated with clomiphene citrate (CC) alone (100 mg/day from day 3 to day 7; n = 56) or in combination with pure FSH (75 IU on day 3 and day 8; n = 54) and were subjected to IUI. Parous women (n = 18) receiving no treatment served as control and were followed-up for spontaneous ovulation. MAIN OUTCOME MEASURE(S): Uterine fluid protein profile, relative intensity of 64.0-kDa protein, number of mature follicles, endometrial thickness, and pregnancy. RESULT(S): Expression of the 64.0-kDa protein exhibited positive correlation with prevailing estradiol levels, but the degree of the protein response to estrogen was comparatively blunted in the CC-incorporated cycles. Endometrial thickness and pregnancy outcomes correlated positively with the expression of the protein. CONCLUSION(S): The 64.0-kDa hUF protein perhaps plays a role in endometrial receptivity to support pregnancy. Failure of pregnancy despite documented ovulation in CC-stimulated cycles may be due to its attenuating effects on expression of the protein.


Assuntos
Estrogênios/metabolismo , Líquido Folicular/metabolismo , Glicoproteínas/análise , Infertilidade Feminina/metabolismo , Infertilidade Feminina/terapia , Indução da Ovulação/estatística & dados numéricos , Serpinas/análise , Adulto , Biomarcadores/análise , Feminino , Glicoproteínas/química , Humanos , Índia/epidemiologia , Infertilidade Feminina/epidemiologia , Peso Molecular , Serpinas/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA