Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Cell Sci ; 131(23)2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30404828

RESUMO

α-Synuclein is associated with Parkinson's disease, and is mainly localized in presynaptic terminals and regulates exocytosis, but its physiological roles remain controversial. Here, we studied the effects of soluble and aggregated α-synuclein on exocytosis, and explored the molecular mechanism by which α-synuclein interacts with regulatory proteins, including Rab3A, Munc13-1 (also known as Unc13a) and Munc18-1 (also known as STXBP1), in order to regulate exocytosis. Through fluorescence recovery after photobleaching experiments, overexpressed α-synuclein in PC12 cells was found to be in a monomeric form, which promotes exocytosis. In contrast, aggregated α-synuclein induced by lactacystin treatment inhibits exocytosis. Our results show that α-synuclein is involved in vesicle priming and fusion. α-Synuclein and phorbol 12-myristate 13-acetate (PMA), which is known to enhance vesicle priming mediated by Rab3A, Munc13-1 and Munc18-1, act on the same population of vesicles, but regulate priming independently. Furthermore, the results show a novel effects of α-synuclein on mobilizing Ca2+ release from thapsigargin-sensitive Ca2+ pools to enhance the ATP-induced [Ca2+]i increase, which enhances vesicle fusion. Our results provide a detailed understanding of the action of α-synuclein during the final steps of exocytosis.


Assuntos
Cálcio/metabolismo , Exocitose/fisiologia , Tapsigargina/farmacologia , alfa-Sinucleína/metabolismo , Animais , Fusão de Membrana/fisiologia , Células PC12 , Ratos , Tapsigargina/metabolismo , Transfecção , Proteína rab3A de Ligação ao GTP/genética , Proteína rab3A de Ligação ao GTP/metabolismo
2.
Sensors (Basel) ; 20(6)2020 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-32204388

RESUMO

The harmful impact of the heavy metal lead on human health has been known for years. However, materials that contain lead remain in the environment. Measuring the blood lead level (BLL) is the only way to officially evaluate the degree of exposure to lead. The so-called "safe value" of the BLL seems to unreliably represent the secure threshold for children. In general, lead's underlying toxicological mechanism remains unclear and needs to be elucidated. Therefore, we developed a novel genetically encoded fluorescence resonance energy transfer (FRET)-based lead biosensor, Met-lead, and applied it to transgenic Drosophila to perform further investigations. We combined Met-lead with the UAS-GAL4 system to the sensor protein specifically expressed within certain regions of fly brains. Using a suitable imaging platform, including a fast epifluorescent or confocal laser-scanning/two-photon microscope with high resolution, we recorded the changes in lead content inside fly brains ex vivo and in vivo and at different life stages. The blood-brain barrier was found to play an important role in the protection of neurons in the brain against damage due to the heavy metal lead, either through food or microinjection into the abdomen. Met-lead has the potential to be a powerful tool for the sensing of lead within living organisms by employing either a fast epi-FRET microscope or high-resolution brain imaging.


Assuntos
Técnicas Biossensoriais , Drosophila melanogaster/química , Chumbo/isolamento & purificação , Metais Pesados/isolamento & purificação , Animais , Chumbo/química , Metais Pesados/química
3.
BMC Bioinformatics ; 16: 117, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25888019

RESUMO

BACKGROUND: Unsupervised analyses such as clustering are the essential tools required to interpret time-series expression data from microarrays. Several clustering algorithms have been developed to analyze gene expression data. Early methods such as k-means, hierarchical clustering, and self-organizing maps are popular for their simplicity. However, because of noise and uncertainty of measurement, these common algorithms have low accuracy. Moreover, because gene expression is a temporal process, the relationship between successive time points should be considered in the analyses. In addition, biological processes are generally continuous; therefore, the datasets collected from time series experiments are often found to have an insufficient number of data points and, as a result, compensation for missing data can also be an issue. RESULTS: An affinity propagation-based clustering algorithm for time-series gene expression data is proposed. The algorithm explores the relationship between genes using a sliding-window mechanism to extract a large number of features. In addition, the time-course datasets are resampled with spline interpolation to predict the unobserved values. Finally, a consensus process is applied to enhance the robustness of the method. Some real gene expression datasets were analyzed to demonstrate the accuracy and efficiency of the algorithm. CONCLUSION: The proposed algorithm has benefitted from the use of cubic B-splines interpolation, sliding-window, affinity propagation, gene relativity graph, and a consensus process, and, as a result, provides both appropriate and effective clustering of time-series gene expression data. The proposed method was tested with gene expression data from the Yeast galactose dataset, the Yeast cell-cycle dataset (Y5), and the Yeast sporulation dataset, and the results illustrated the relationships between the expressed genes, which may give some insights into the biological processes involved.


Assuntos
Algoritmos , Gráficos por Computador , Perfilação da Expressão Gênica/métodos , Regulação Fúngica da Expressão Gênica , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Ciclo Celular/fisiologia , Análise por Conglomerados , Sequência Consenso , Galactose/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Esporos Fúngicos/fisiologia , Fatores de Tempo
4.
Pharmaceutics ; 14(7)2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35890212

RESUMO

Ligand-targeting drug conjugates are a class of clinically validated biopharmaceutical drugs constructed by conjugating cytotoxic drugs with specific disease antigen targeting ligands through appropriate linkers. The integrated linker-drug motif embedded within such a system can prevent the premature release during systemic circulation, thereby allowing the targeting ligand to engage with the disease antigen and selective accumulation. We have designed and synthesized new thioester-linked maytansinoid conjugates. By performing in vitro cytotoxicity, targeting ligand binding assay, and in vivo pharmacokinetic studies, we investigated the utility of this new linker-drug moiety in the small molecule drug conjugate (SMDC) system. In particular, we conjugated the thioester-linked maytansinoids to the phosphatidylserine-targeting small molecule zinc dipicolylamine and showed that Zn8_DM1 induced tumor regression in the HCC1806 triple-negative breast cancer xenograft model. Moreover, in a spontaneous sorafenib-resistant liver cancer model, Zn8_DM1 exhibited potent antitumor growth efficacy. From quantitative mRNA analysis of Zn8_DM1 treated-tumor tissues, we observed the elevation of gene expressions associated with a "hot inflamed tumor" state. With the identification and validation of a plethora of cancer-associated antigens in the "omics" era, this work provided the insight that antibody- or small molecule-based targeting ligands can be conjugated similarly to generate new ligand-targeting drug conjugates.

5.
J Med Chem ; 65(19): 12802-12824, 2022 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-36153998

RESUMO

Ligand-targeting drug delivery systems have made significant strides for disease treatments with numerous clinical approvals in this era of precision medicine. Herein, we report a class of small molecule-based immune checkpoint-targeting maytansinoid conjugates. From the ligand targeting ability, pharmacokinetics profiling, in vivo anti-pancreatic cancer, triple-negative breast cancer, and sorafenib-resistant liver cancer efficacies with quantitative mRNA analysis of treated-tumor tissues, we demonstrated that conjugate 40a not only induced lasting regression of tumor growth, but it also rejuvenated the once immunosuppressive tumor microenvironment to an "inflamed hot tumor" with significant elevation of gene expressions that were not accessible in the vehicle-treated tumor. In turn, the immune checkpoint-targeting small molecule drug conjugate from this work represents a new pharmacodelivery strategy that can be expanded with combination therapy with existing immune-oncology treatment options.


Assuntos
Fosfatidilserinas , Neoplasias de Mama Triplo Negativas , Humanos , Ligantes , RNA Mensageiro , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Microambiente Tumoral
6.
Biomed J ; 44(6 Suppl 2): S210-S217, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-35300943

RESUMO

BACKGROUND: Previous studies showed inconsistent Results of the effects of dipeptidyl peptidase (DPP)-IV inhibitors on syngeneic mouse islet transplantation. We hypothesized that the implanted islet numbers are critical for the effects of DPP-IV inhibitors on the outcomes of transplantation. METHODS: One hundred and fifty or three hundred islets were syngeneically transplanted under the renal capsule of each streptozocin-diabetic C57BL/6 mouse and recipients were then treated without or with LAF237 (10 mg/kg/day, po) for 6 weeks. After transplantation, recipients' blood glucose, body weight and intraperitoneal glucose tolerance test (IPGTT) were followed-up periodically. The graft was removed for the measurement of ß-cell mass at 6 weeks. RESULTS: In recipients with 150 islets, it was not significantly different between the LAF237- treated group (n = 14) and control group (n = 14) in terms of the blood glucose, body weight, glucose tolerance at 2, 4 and 6 weeks or the graft ß-cell mass at 6 weeks. In contrast, in recipients with 300 islets, the LAF237-treated group (n = 24) did have a lower area under the curve of the IPGTT at 4 weeks (p = 0.0237) and 6 weeks (p = 0.0113) as well as more graft ß-cell mass at 6 weeks (0.655 ± 0.008 mg vs. 0.435 ± 0.006 mg, p = 0.0463) than controls (n = 24). CONCLUSIONS: Our findings revealed 6-week treatment of LAF237 improves glucose tolerance and increases graft ß-cell mass in diabetic mice transplanted with a sufficient number but not a marginal number of islets. These indicate that the effects of DPP-IV inhibitors are influenced by the implanted islet mass.


Assuntos
Diabetes Mellitus Experimental , Inibidores da Dipeptidil Peptidase IV , Adamantano/análogos & derivados , Animais , Glicemia , Peso Corporal , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/cirurgia , Inibidores da Dipeptidil Peptidase IV/farmacologia , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Dipeptidil Peptidases e Tripeptidil Peptidases , Humanos , Hipoglicemiantes/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Pirrolidinas
7.
Front Chem ; 9: 822587, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35242746

RESUMO

An efficient Ugi multicomponent reaction with strain promoted azide-alkyne cycloaddition protocol has been utilized in concert or independently to prepare a small family of bioactive zinc(II) dipicolylamine (ZnDPA)-based SN-38 conjugates. With sequential click chemistry coupling between the cytotoxic payload and phosphatidylserine-targeting ZnDPA ligand derived from structurally diverse carboxylic acids, aldehyde or ketones, and isocyanides, we demonstrated that this convergent synthetic strategy could furnish conjugates harnessing diversified linkers that exhibited different pharmacokinetic profiles in systemic circulation in vivo. Among the eight new conjugates, comparative studies on in vitro cytotoxicities, plasma stabilities, in vivo pharmacokinetic properties, and maximum tolerated doses were then carried out to identify a potent ZnDPA-based SN-38 conjugate that resulted in pancreatic cancer growth regression with an 80% reduction of cytotoxic payload used when compared to that of the marketed irinotecan. Our work provided the roadmap to construct a variety of theranostic agents in a similar manner for cancer treatment.

8.
Transl Oncol ; 14(1): 100897, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33069101

RESUMO

Zinc(II)-dipicolylamine (Zn-DPA) has been shown to specifically identify and bind to phosphatidylserine (PS), which exists in bulk in the tumor microenvironment. BPRDP056, a Zn-DPA-SN38 conjugate was designed to provide PS-targeted drug delivery of a cytotoxic SN38 to the tumor microenvironment, thereby allowing a lower dosage of SN38 that induces apoptosis in cancer cells. Micro-Western assay showed that BPRDP056 exhibited apoptotic signal levels similar to those of CPT-11 in the treated tumors growing in mice. Pharmacokinetic study showed that BPRDP056 has excellent systemic stability in circulation in mice and rats. BPRDP056 is accumulated in tumors and thus increases the cytotoxic effects of SN38. The in vivo antitumor activities of BPRDP056 have been shown to be significant in subcutaneous pancreas, prostate, colon, liver, breast, and glioblastoma tumors, included an orthotopic pancreatic tumor, in mice. BPRDP056 shrunk tumors at a lower (~20% only) dosing intensity of SN38 compared to that of SN38 conjugated in CPT-11 in all tumor models tested. A wide spectrum of antitumor activities is expected to treat all cancer types of PS-rich tumor microenvironments. BPRDP056 is a first-in-class small molecule drug conjugate for cancer therapy.

9.
Microsc Microanal ; 16(3): 313-26, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20377928

RESUMO

The Orai1-STIM1 constructed store-operated Ca2+ channels (SOCs) have been found to exert several essential Ca2+ entry/signaling cascades, e.g., the generation of immune response in T lymphocytes. Although biochemical and novel imaging evidence appear to indicate that Orai1 and STIM1 interact with each other to achieve store-operated Ca2+ entry (SOCE), the detailed mechanism of functional SOCE in situ has yet to be fully understood. In this study, green fluorescence protein (EGFP as donor) targeted to either the N- or C-terminal of Orai1 (wild type or delta1-90+delta267-301 double deletion type) and mOrange (as acceptor) tagged STIM1 were used to comprise a fluorescence resonance energy transfer (FRET) pair within living PC12 cells. The fluorescence lifetime map and histogram/distribution of each single cell, determined by one-photon excitation fluorescence lifetime imaging microscopy (FLIM), was used to visualize FRET and show the Orai1 homodimer and Orai1-STIM1 binding. Both the color-coded lifetime map and the distribution of EGFP-tagged Orai1 significantly changed after the administration of thapsigargin, the SOCE stimulating agent. The FRET efficiency from each experimental set was also calculated and compared using double exponential analysis. In summary, we show the detailed interactions Orai1-Orai1 and Orai1-STIM1 within intact living cells by using the FLIM-FRET technique.


Assuntos
Canais de Cálcio/metabolismo , Dimerização , Processamento de Imagem Assistida por Computador/métodos , Glicoproteínas de Membrana/metabolismo , Microscopia de Fluorescência/métodos , Animais , Linhagem Celular , Transferência Ressonante de Energia de Fluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteína ORAI1 , Ligação Proteica , Ratos , Coloração e Rotulagem/métodos , Molécula 1 de Interação Estromal
10.
Sci Rep ; 10(1): 2838, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32071352

RESUMO

A major obstacle to nanodrugs-mediated cancer therapy is their rapid uptake by the reticuloendothelial system that decreases the systemic exposure of the nanodrugs to tumors and also increases toxicities. Intralipid has been shown to reduce nano-oxaliplatin-mediated toxicity while improving bioavailability. Here, we have found that Intralipid reduces the cytotoxicity of paclitaxel for human monocytic cells, but not for breast, lung, or pancreatic cancer cells. Intralipid also promotes the polarization of macrophages to the anti-cancer M1-like phenotype. Using a xenograft breast cancer mouse model, we have found that Intralipid pre-treatment significantly increases the amount of paclitaxel reaching the tumor and promotes tumor apoptosis. The combination of Intralipid with half the standard clinical dose of Abraxane reduces the tumor growth rate as effectively as the standard clinical dose. Our findings suggest that pre-treatment of Intralipid has the potential to be a powerful agent to enhance the tumor cytotoxic effects of Abraxane and to reduce its off-target toxicities.


Assuntos
Paclitaxel Ligado a Albumina/farmacologia , Neoplasias da Mama/tratamento farmacológico , Imunidade Inata/efeitos dos fármacos , Fosfolipídeos/farmacologia , Óleo de Soja/farmacologia , Animais , Antineoplásicos , Apoptose/efeitos dos fármacos , Disponibilidade Biológica , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Emulsões/farmacologia , Feminino , Xenoenxertos , Humanos , Camundongos , Nanopartículas/química , Oxaliplatina/farmacologia , Paclitaxel/química , Paclitaxel/farmacologia , Fosfolipídeos/imunologia , Óleo de Soja/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Biosens Bioelectron ; 168: 112571, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32892119

RESUMO

Forms of lead (Pb) have been insidiously invading human life for thousands of years without obvious signs of their considerable danger to human health. Blood lead level (BLL) is the routine measure used for diagnosing the degree of lead intoxication, although it is unclear whether there is any safe range of BLL. To develop a practical detection tool for living organisms, we engineered a genetically encoded fluorescence resonance energy transfer (FRET)-based Pb2+ biosensor, 'Met-lead 1.44 M1', with excellent performance. Met-lead 1.44 M1 has an apparent dissociation constant (Kd) of 25.97 nM, a detection limit (LOD) of 10 nM (2.0 ppb/0.2 µg/dL), and an enhancement dynamic ratio of nearly ~ 5-fold upon Pb2+ binding. The 10 nM sensitivity of Met-lead 1.44 M1 is five times below the World Health Organization-permitted level of lead in tap water (10 ppb; WHO, 2017), and fifteen times lower than the maximum BLL for children (3 µg/dL). We deployed Met-lead 1.44 M1 to measure Pb2+ concentrations in different living models, including two general human cell lines and one specific line, induced pluripotent stem cell (iPSC)-derived cardiomyocytes, as well as in widely used model species in plant (Arabidopsis thaliana) and animal (Drosophila melanogaster) research. Our results suggest that this new biosensor is suitable for lead toxicological research in vitro and in vivo, and will pave the way toward potential applications for both low BLL measures and rapid detection of environmental lead in its divalent form.


Assuntos
Técnicas Biossensoriais , Chumbo , Animais , Drosophila melanogaster , Transferência Ressonante de Energia de Fluorescência , Chumbo/toxicidade
12.
J Med Chem ; 62(13): 6047-6062, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31181158

RESUMO

We report that compound 13, a novel phosphatidylserine-targeting zinc(II) dipicolylamine drug conjugate, readily triggers a positive feedback therapeutic loop through the in situ generation of phosphatidylserine in the tumor microenvironment. Linker modifications, pharmacokinetics profiling, in vivo antitumor studies, and micro-Western array of treated-tumor tissues were employed to show that this class of conjugates induced regeneration of apoptotic signals, which facilitated subsequent recruitment of the circulating conjugates through the zinc(II) dipicolylamine-phosphatidylserine association and resulted in compounding antitumor efficacy. Compared to the marketed compound 17, compound 13 not only induced regressions in colorectal and pancreatic tumor models, it also exhibited at least 5-fold enhancement in antitumor efficacy with only 40% of the drug employed during treatment, culminating in a >12.5-fold increase in therapeutic potential. Our study discloses a chemically distinct apoptosis-targeting theranostic, with built-in complementary functional moieties between the targeting module and the drug mechanism to expand the arsenal of antitumor therapy.


Assuntos
Antineoplásicos/uso terapêutico , Complexos de Coordenação/uso terapêutico , Indolizinas/uso terapêutico , Neoplasias/tratamento farmacológico , Fosfatidilserinas/metabolismo , Picolinas/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Complexos de Coordenação/síntese química , Complexos de Coordenação/química , Desenho de Fármacos , Humanos , Indolizinas/síntese química , Indolizinas/química , Masculino , Camundongos Endogâmicos ICR , Camundongos Nus , Estrutura Molecular , Picolinas/síntese química , Picolinas/química , Relação Estrutura-Atividade , Inibidores da Topoisomerase I/síntese química , Inibidores da Topoisomerase I/química , Inibidores da Topoisomerase I/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Zinco/química
13.
Bioinformatics ; 23(24): 3374-81, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17956879

RESUMO

MOTIVATION: Determining locations of protein expression is essential to understand protein function. Advances in green fluorescence protein (GFP) fusion proteins and automated fluorescence microscopy allow for rapid acquisition of large collections of protein localization images. Recognition of these cell images requires an automated image analysis system. Approaches taken by previous work concentrated on designing a set of optimal features and then applying standard machine-learning algorithms. In fact, trends of recent advances in machine learning and computer vision can be applied to improve the performance. One trend is the advances in multiclass learning with error-correcting output codes (ECOC). Another trend is the use of a large number of weak detectors with boosting for detecting objects in images of real-world scenes. RESULTS: We take advantage of these advances to propose a new learning algorithm, AdaBoost.ERC, coupled with weak and strong detectors, to improve the performance of automatic recognition of protein subcellular locations in cell images. We prepared two image data sets of CHO and Vero cells and downloaded a HeLa cell image data set in the public domain to evaluate our new method. We show that AdaBoost.ERC outperforms other AdaBoost extensions. We demonstrate the benefit of weak detectors by showing significant performance improvements over classifiers using only strong detectors. We also empirically test our method's capability of generalizing to heterogeneous image collections. Compared with previous work, our method performs reasonably well for the HeLa cell images. AVAILABILITY: CHO and Vero cell images, their corresponding feature sets (SSLF and WSLF), our new learning algorithm, AdaBoost.ERC, and Supplementary Material are available at http://aiia.iis.sinica.edu.tw/


Assuntos
Algoritmos , Inteligência Artificial , Interpretação de Imagem Assistida por Computador/métodos , Microscopia de Fluorescência/métodos , Reconhecimento Automatizado de Padrão/métodos , Proteínas/metabolismo , Frações Subcelulares/metabolismo , Proteínas/ultraestrutura , Frações Subcelulares/ultraestrutura
14.
Microsc Res Tech ; 71(4): 305-14, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18069668

RESUMO

Systemic analysis of subcellular protein localization (location proteomics) provides clues for understanding gene functions and physiological condition of the cells. However, recognition of cell images of subcellular structures highly depends on experience and becomes the rate-limiting step when classifying subcellular protein localization. Several research groups have extracted specific numerical features for the recognition of subcellular protein localization, but these recognition systems are restricted to images of single particular cell line acquired by one specific imaging system and not applied to recognize a range of cell image sources. In this study, we establish a single system for automated subcellular structure recognition to identify cell images from various sources. Two different sources of cell images, 317 Vero (http://gfp-cdna.embl.de) and 875 CHO cell images of subcellular structures, were used to train and test the system. When the system was trained by a single source of images, the recognition rate is high and specific to the trained source. The system trained by the CHO cell images gave high average recognition accuracy for CHO cells of 96%, but this was reduced to 46% with Vero images. When we trained the system using a mixture of CHO and Vero cell images, an average accuracy of recognition reached 86.6% for both CHO and Vero cell images. The system can reject images with low confidence and identify the cell images correctly recognized to avoid manual reconfirmation. In summary, we have established a single system that can recognize subcellular protein localizations from two different sources for location-proteomic studies. studies.


Assuntos
Microscopia de Fluorescência/métodos , Reconhecimento Automatizado de Padrão/métodos , Proteínas/metabolismo , Frações Subcelulares/classificação , Frações Subcelulares/ultraestrutura , Algoritmos , Animais , Células CHO , Chlorocebus aethiops , Cricetinae , Cricetulus , Proteínas de Fluorescência Verde/metabolismo , Interpretação de Imagem Assistida por Computador , Frações Subcelulares/metabolismo , Células Vero
15.
Microsc Res Tech ; 70(2): 119-34, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17146761

RESUMO

Recent technological improvements have made it possible to examine the dynamics of individual vesicles at a very high temporal and spatial resolution. Quantification of the dynamic properties of secretory vesicles is labor-intensive and therefore it is crucial to develop software to automate the process of analyzing vesicle dynamics. Dual-threshold and binary image conversion were applied to enhance images and define the areas of objects of interest that were to be tracked. The movements, changes in fluorescence intensity, and changes in the area of each tracked object were measured using a new software system named the Protein Tracking system (PTrack). Simulations revealed that the system accurately recognized tracked objects and measured their dynamic properties. Comparison of the results from tracking real time-lapsed images manually with those automatically obtained using PTrack revealed similar patterns for changes in fluorescence intensity and a high accuracy (<89%). According to tracking results, PTrack can distinguish different vesicular organelles that are similar in shape, based on their unique dynamic properties. In conclusion, the novel tracking system, PTrack, should facilitate automated quantification of the dynamic properties of vesicles that are important when classifying vesicular protein locations.


Assuntos
Exocitose/fisiologia , Vesículas Secretórias/fisiologia , Animais , Transporte Biológico Ativo , Proteínas de Fluorescência Verde/metabolismo , Processamento de Imagem Assistida por Computador , Microscopia de Fluorescência/instrumentação , Microscopia de Fluorescência/métodos , Neuropeptídeo Y/metabolismo , Células PC12 , Peroxissomos/fisiologia , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Vesículas Secretórias/ultraestrutura , Proteína rab3A de Ligação ao GTP/metabolismo
16.
Ann Clin Transl Neurol ; 3(11): 854-865, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27844031

RESUMO

OBJECTIVE: Charcot-Marie-Tooth disease type X1 (CMTX1), which is caused by mutations in the gap junction (GJ) protein beta-1 gene (GJB1), is the second most common form of Charcot-Marie-Tooth disease (CMT). GJB1 encodes the GJ beta-1 protein (GJB1), which forms GJs within the myelin sheaths of peripheral nerves. The process by which GJB1 mutants cause neuropathy has not been fully elucidated. This study evaluated the biophysical characteristics of GJB1 mutants and their correlations with the clinical features of CMTX1 patients. METHODS: All patients with a validated GJB1 mutation were assessed using the Charcot-Marie-Tooth disease neuropathy score version 2 (CMTNS). The impacts of the mutations on the biophysical functions of GJB1 were characterized by assessing intracellular localization, expression patterns, and GJ Ca2+ permeability. RESULT: Nineteen GJB1 mutations were identified in 24 patients with a clinical diagnosis of CMT. Six are novel mutations: p.L6S, p.I20F, p.I101Rfs*8, p.F153L, p.R215P, and p.D278V. Diverse pathological effects of the mutations were demonstrated, including reduced GJB1 expression, intracellular mislocalization, and altered GJ functions. GJB1 mutations that caused a complete loss of GJ Ca2+ permeability appeared to be associated with an earlier disease onset, whereas those resulting in preservation of GJ permeability and with predominant cell membrane expression tended to have a later onset and a milder phenotype. INTERPRETATION: This study demonstrated that the degree of loss of GJ function caused by the GJB1 mutations might contribute to the onset and severity of neuropathic symptoms in CMTX1.

17.
PLoS One ; 8(6): e65853, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23776557

RESUMO

Cd(2+) causes damages to several human tissues. Although the toxicological and carcinogenetic mechanisms of Cd(2+) have been previously established, some basic questions on this toxicant remain unclear. In this study, we constructed Met-cad 1.57, a new fluorescent resonance energy transfer (FRET)-based Cd(2+) indicator, which contains a portion of a Cd(2+)-binding protein (CadR) obtained from Pseudomonas putida as the Cd(2+) sensing key. We produced a human embryonic kidney cell line HEK-MCD157 which stably expresses the Met-cad 1.57 for further investigations. Both fluorescence spectroscopy and FRET microscopic ratio imaging were used to monitor the Cd(2+) concentration within the living HEK-MCD157 cells. The dissociation constant of Met-cad 1.57 was approximately 271 nM. The function of Ca(2+) channels as a potential Cd(2+) entry gateway was further confirmed in the HEK-MCD157 cells. The organelle-targeted property of the protein-based Cd(2+) indicator directly reveals the nucleus accumulation phenomena. In summary, a human kidney cell line that stably expresses the FRET-based Cd(2+) indicator Met-cad 1.57 was constructed for reliable and convenient investigations to determine the Cd(2+) concentration within living cells, including the identification of the entry pathway of Cd(2+) and sub-cellular sequestration.


Assuntos
Cádmio/metabolismo , Transferência Ressonante de Energia de Fluorescência , Proteínas de Bactérias/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Humanos , Espectrometria de Fluorescência , Fatores de Transcrição/metabolismo
18.
PLoS One ; 8(9): e75884, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24073285

RESUMO

The DOC-2/DAB2 interactive protein (DAB2IP) is a new member of the Ras GTPase-activating protein family. Recent studies have shown that, in addition to its tumor suppressive role in various tumors, DAB2IP also plays an important role in regulating neuronal migration and positioning during brain development. In this study, we determined the roles of DAB2IP in the neuronal differentiation of human mesenchymal stem cells (hMSCs). We found that lentiviral short hairpin RNA (shRNA)-mediated knockdown of DAB2IP promoted the mesenchymal-to-neuroepithelial stem cell transition (MtNeST) and neuronal differentiation, which were accompanied by a reduction of cell proliferation but not apoptosis or cellular senescence. This suggests that DAB2IP plays an important role in the neuronal induction of hMSCs. Moreover, our finding that reduction of glycogen synthase kinase 3 beta (GSK3ß) activity upon LiCl pretreatment inhibited both the MtNeST and production of MAP2-positive cells upon DAB2IP knockdown suggests that this transition is most likely mediated by regulation of the GSK3ß signaling pathway. Our study demonstrates that DAB2IP participates in the first step of neuron induction of hMSCs, which implies a potentially important role for DAB2IP in the MtNeST during neurogenesis.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Células Neuroepiteliais/citologia , Neurônios/citologia , Proteínas Ativadoras de ras GTPase/metabolismo , Apoptose , Western Blotting , Proliferação de Células , Células Cultivadas , Regulação para Baixo , Citometria de Fluxo , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Lentivirus/genética , Células-Tronco Mesenquimais/metabolismo , Microscopia de Fluorescência , Células Neuroepiteliais/metabolismo , Neurônios/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Ativadoras de ras GTPase/antagonistas & inibidores , Proteínas Ativadoras de ras GTPase/genética
19.
Toxicol Sci ; 126(2): 436-45, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22240981

RESUMO

Lead ion (Pb(2+)) is one of the most hazardous heavy metals to almost all life forms. The components of store-operated Ca(2+) entry as a molecular gateway have been previously found to participate in the cytotoxic entry of Pb(2+). However, the safe levels of intracellular Pb(2+) hiding in blood Pb(2+) levels are still not determined with full certainty. The present study aimed to construct protein-based Pb(2+) indicators to help establish a reliable setting for the content monitoring of intracellular Pb(2+). A series of Pb(2+) indicators based on fluorescence resonance energy transfer, Met-leads, were developed. The Pb(2+)-binding protein PbrR (from Cupriavidus metallidurans CH34) was applied between the fluorescent protein pair ECFP(ΔC11) and cp173Venus. The spectral patterns and sensing ranges of all Met-leads were characterized in vitro. Among these constructs, Met-lead 1.59 had relatively high ion selectivity and broad dynamic range (3.3-5.7). Consequently, this Met-lead was adopted in the cellular Pb(2+) biosensing. The intracellular Pb(2+) content in human embryonic kidney cells was successfully monitored using Met-lead 1.59 under both short- and long-term treatments. The existence of intracellular Pb(2+) can be significantly sensed using Met-lead 1.59 after 3 h 0.5µM (10 µg/dl) exposure, which is 200 times more improved than previous live-cell indicators. In summary, a new Pb(2+) indicator, Met-lead 1.59, was successfully developed for advanced research on Pb(2+) toxicology.


Assuntos
Chumbo/análise , Proteínas/química , Sequência de Aminoácidos , Técnicas Biossensoriais , Linhagem Celular , Humanos , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
20.
Toxicol Sci ; 110(2): 353-62, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19429662

RESUMO

Pb(2+) ions cause severe damages to living cells. In particular, our previous study showed that the Orai-STIM1 (stromal interacting protein 1)-formed store-operated Ca(2+) channels (SOCs) allow Pb(2+) entry. In relation to this, the present study investigates the molecular gating mechanism of Pb(2+) entry by Orai1 with STIM1, as well as the resulting cytotoxicity on human embryonic kidney HEK293 cells. The store-operated Ca(2+) entry (SOCE, activity of SOCs) and Pb(2+) entry were measured using the fura-2 imaging method and indo-1 quenching strategy, as well as through an atomic absorption spectrophotometer. The results of RT-PCR, Western blot, fast confocal, and fluorescent lifetime imaging microscopy indicated the endogenous expression of Orai1 and STIM1 in HEK cells and the functional interaction between these two proteins during SOCE. Both SOCE and Pb(2+) entry largely increased when Orai1 and STIM1 were overexpressed (3- and 1.64-folds, respectively) compared with nonfluorescent cells, and they were significantly attenuated when the E106Q mutation of Orail with STIM1 was cotransfected (6- and 2.25-folds decrease, respectively) compared with Orai1-STIM1 coexpressed cells. The ion gating for Pb(2+) could be governed by the E106 region of Orai1. After sorting and subsequent cultures, the Orai1-STIM1 positive expressed cells behaved more sensitively to Pb(2+) than the Orai1-STIM1 negative cells. In summary, the data suggest that Orai1, together with STIM1, plays a critical role in Pb(2+) entry and the toxicity of Pb(2+).


Assuntos
Canais de Cálcio/metabolismo , Citosol/metabolismo , Chumbo/metabolismo , Chumbo/toxicidade , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animais , Transporte Biológico , Western Blotting , Cálcio/metabolismo , Canais de Cálcio/genética , Proliferação de Células/efeitos dos fármacos , Separação Celular , Sobrevivência Celular/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Humanos , Proteínas de Membrana/genética , Microscopia Confocal , Microscopia de Fluorescência , Mutação , Proteínas de Neoplasias/genética , Proteína ORAI1 , Células PC12 , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Espectrofotometria Atômica , Molécula 1 de Interação Estromal , Tapsigargina/farmacologia , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA