Статті в журналах з теми "Cancer Cell Imaging - Gold Nanoclusters"

Щоб переглянути інші типи публікацій з цієї теми, перейдіть за посиланням: Cancer Cell Imaging - Gold Nanoclusters.

Оформте джерело за APA, MLA, Chicago, Harvard та іншими стилями

Оберіть тип джерела:

Ознайомтеся з топ-50 статей у журналах для дослідження на тему "Cancer Cell Imaging - Gold Nanoclusters".

Біля кожної праці в переліку літератури доступна кнопка «Додати до бібліографії». Скористайтеся нею – і ми автоматично оформимо бібліографічне посилання на обрану працю в потрібному вам стилі цитування: APA, MLA, «Гарвард», «Чикаго», «Ванкувер» тощо.

Також ви можете завантажити повний текст наукової публікації у форматі «.pdf» та прочитати онлайн анотацію до роботи, якщо відповідні параметри наявні в метаданих.

Переглядайте статті в журналах для різних дисциплін та оформлюйте правильно вашу бібліографію.

1

Chattoraj, Shyamtanu, Md Asif Amin, Saswat Mohapatra, Surajit Ghosh, and Kankan Bhattacharyya. "Cancer Cell Imaging Using in Situ Generated Gold Nanoclusters." ChemPhysChem 17, no. 1 (October 23, 2015): 61–68. http://dx.doi.org/10.1002/cphc.201500731.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
2

Zheng, Youkun, Weiwei Liu, Yun Chen, Hui Jiang, and Xuemei Wang. "Mercaptopyrimidine-directed gold nanoclusters: a suitable fluorescent probe for intracellular glutathione imaging and selective cancer cell identification." Journal of Materials Chemistry B 6, no. 22 (2018): 3650–54. http://dx.doi.org/10.1039/c8tb00791h.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
3

Kuo, Jui-Chi, Tsung-Rong Kuo, Fajar Rinawati, Erna Susilowati, Sucipto, and Dyah Ika Krisnawati. "Inhibition of cancer cells using target-specific 2A3 antibody-conjugated gold nanoclusters." Acta Biochimica Indonesiana 4, no. 2 (March 1, 2022): 69. http://dx.doi.org/10.32889/actabioina.69.

Повний текст джерела
Анотація:
Background: Metal nanoclusters (NCs) with outstanding structural and optical properties have been intensively validated for applications in nanomedicine and nanotechnology. Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) is overexpressed in many cancer cells. Objective: The gold nanoclusters conjugated with a single domain antibody targeting CEACAM6 of 2A3 (2A3-AuNCs) were synthesized for the inhibition of cancer cells. Methods: 2A3-AuNCs were prepared via a facile hydrothermal approach. The cell viability was measured by resazurin dye reduction assay. The cell death was analyzed by fluorescence imaging. Results: Structural and optical characterizations demonstrated the successful synthesis of 2A3-AuNCs with a roughly spherical shape and a size of 2.35 nm. The 2A3-AuNCs revealed a maximum fluorescence intensity at 350 nm with a fluorescence quantum yield of 4.0%. The cell viability assay indicated that 2A3-AuNCs could inhibit the growths of cancer cells with overexpressed CEACAM6, including breast cancer MDA-MB-231 and MDA-MB-468 cells. The fluorescence imaging results also demonstrated that 2A3-AuNCs could inhibit the growth of cancer cells with MDA-MB-231 and MDA-MB-468 cells. Conclusion: Combination with the results of cell viability assay and fluorescence imaging, the surface ligand of 2A3 antibody on 2A3-AuNCs exhibited promising inhibition of CEACAM6 overexpressed cancer cells. Our work provides a potential application of AuNCs in cancer therapy.
Стилі APA, Harvard, Vancouver, ISO та ін.
4

Wang, Jianling, Leifeng Chen, Jing Ye, Zhiyong Li, Hui Jiang, Hong Yan, Marina Yu Stogniy, Igor B. Sivaev, Vladimir I. Bregadze, and Xuemei Wang. "Carborane Derivative Conjugated with Gold Nanoclusters for Targeted Cancer Cell Imaging." Biomacromolecules 18, no. 5 (April 4, 2017): 1466–72. http://dx.doi.org/10.1021/acs.biomac.6b01845.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
5

Pan, Yiting, Qinzhen Li, Qin Zhou, Wan Zhang, Ping Yue, Changzhi Xu, Ximing Qin, Haizhu Yu, and Manzhou Zhu. "Cancer cell specific fluorescent methionine protected gold nanoclusters for in-vitro cell imaging studies." Talanta 188 (October 2018): 259–65. http://dx.doi.org/10.1016/j.talanta.2018.05.079.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
6

Lee, Eun Sung, Byung Seok Cha, Seokjoon Kim, and Ki Soo Park. "Synthesis of Exosome-Based Fluorescent Gold Nanoclusters for Cellular Imaging Applications." International Journal of Molecular Sciences 22, no. 9 (April 23, 2021): 4433. http://dx.doi.org/10.3390/ijms22094433.

Повний текст джерела
Анотація:
In recent years, fluorescent metal nanoclusters have been used to develop bioimaging and sensing technology. Notably, protein-templated fluorescent gold nanoclusters (AuNCs) are attracting interest due to their excellent fluorescence properties and biocompatibility. Herein, we used an exosome template to synthesize AuNCs in an eco-friendly manner that required neither harsh conditions nor toxic chemicals. Specifically, we used a neutral (pH 7) and alkaline (pH 11.5) pH to synthesize two different exosome-based AuNCs (exo-AuNCs) with independent blue and red emission. Using field-emission scanning electron microscopy, energy dispersive X-ray microanalysis, nanoparticle tracking analysis, and X-ray photoelectron spectroscopy, we demonstrated that AuNCs were successfully formed in the exosomes. Red-emitting exo-AuNCs were found to have a larger Stokes shift and a stronger fluorescence intensity than the blue-emitting exo-AuNCs. Both exo-AuNCs were compatible with MCF-7 (human breast cancer), HeLa (human cervical cancer), and HT29 (human colon cancer) cells, although blue-emitting exo-AuNCs were cytotoxic at high concentrations (≥5 mg/mL). Red-emitting exo-AuNCs successfully stained the nucleus and were compatible with membrane-staining dyes. This is the first study to use exosomes to synthesize fluorescent nanomaterials for cellular imaging applications. As exosomes are naturally produced via secretion from almost all types of cell, the proposed method could serve as a strategy for low-cost production of versatile nanomaterials.
Стилі APA, Harvard, Vancouver, ISO та ін.
7

Qiao, Juan, Xiaoyu Mu, Li Qi, Jingjing Deng, and Lanqun Mao. "Folic acid-functionalized fluorescent gold nanoclusters with polymers as linkers for cancer cell imaging." Chemical Communications 49, no. 73 (2013): 8030. http://dx.doi.org/10.1039/c3cc44256j.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
8

Chattoraj, Shyamtanu, Md Asif Amin, Saswat Mohapatra, Surajit Ghosh, and Kankan Bhattacharyya. "Inside Cover: Cancer Cell Imaging Using in Situ Generated Gold Nanoclusters (ChemPhysChem 1/2016)." ChemPhysChem 17, no. 1 (January 2016): 2. http://dx.doi.org/10.1002/cphc.201501148.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
9

Ye, Jing, Tianxiang Hu, Yanqi Wu, Hui Chen, Qianqian Qiu, Rongqing Geng, Hui Ding, and Xiaojuan Zhao. "Near-Infrared Liposome-Capped Au-Rare Earth Bimetallic Nanoclusters for Fluorescence Imaging of Tumor Cells." Journal of Biomedical Nanotechnology 18, no. 9 (September 1, 2022): 2113–22. http://dx.doi.org/10.1166/jbn.2022.3423.

Повний текст джерела
Анотація:
Early detection of cancer can effectively improve the survival rate of cancer patients. Fluorescence imaging has the advantages of high sensitivity and rapid imaging, and is widely used in the precise imaging detection of tumors. In this study, five kinds of Au-rare earth bimetallic nanoclusters (Au/Ln NCs) were prepared by template method using five representative rare earth elements doped with gold. The morphologies, surface charges, sizes, fluorescence quantum yields and maximum fluorescence emission wavelengths of these five kinds of Au/Ln NCs were characterized and contrasted. The findings indicated that the Au/Ce nanoclusters (Au/Ce NCs) prepared by Ce doping have the longest fluorescence emission wavelength (695 nm) and higher quantum yield, which could effectively avoid the interference of autofluorescence, and was suitable for fluorescence imaging of tumor cells. In order to improve the specific accumulation of nanoclusters in tumor cells, Au/Ce NCs were coated with folic acid modified liposomes (lip-FA) to constructed a targeted fluorescent imaging probe with near-infrared response (Au/Ce@lip-FA), which was successfully used for fluorescence imaging of tumor cells. The probe has the characteristics of stable fluorescence signal, good targeting, easy internalization, and safe metabolism, and can provide high-resolution and high-brightness imaging information, which is expected to play an important role in the clinical diagnosis and surgical treatment of tumors.
Стилі APA, Harvard, Vancouver, ISO та ін.
10

Wang, Chensu, Jingyuan Li, Christian Amatore, Yu Chen, Hui Jiang, and Xue-Mei Wang. "Gold Nanoclusters and Graphene Nanocomposites for Drug Delivery and Imaging of Cancer Cells." Angewandte Chemie International Edition 50, no. 49 (October 11, 2011): 11644–48. http://dx.doi.org/10.1002/anie.201105573.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
11

Wang, Chensu, Jingyuan Li, Christian Amatore, Yu Chen, Hui Jiang, and Xue-Mei Wang. "Gold Nanoclusters and Graphene Nanocomposites for Drug Delivery and Imaging of Cancer Cells." Angewandte Chemie 123, no. 49 (October 11, 2011): 11848–52. http://dx.doi.org/10.1002/ange.201105573.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
12

Bian, Pingping, Jing Zhou, Yueying Liu, and Zhanfang Ma. "One-step fabrication of intense red fluorescent gold nanoclusters and their application in cancer cell imaging." Nanoscale 5, no. 13 (2013): 6161. http://dx.doi.org/10.1039/c3nr01282d.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
13

Hada, Alexandru-Milentie, Ana-Maria Craciun, Monica Focsan, Raluca Borlan, Olga Soritau, Milica Todea, and Simion Astilean. "Folic acid functionalized gold nanoclusters for enabling targeted fluorescence imaging of human ovarian cancer cells." Talanta 225 (April 2021): 121960. http://dx.doi.org/10.1016/j.talanta.2020.121960.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
14

Zhu, Shuxian, Xiaoyu Wang, Lu Liu, and Lidong Li. "Gold nanocluster grafted conjugated polymer nanoparticles for cancer cell imaging and photothermal killing." Colloids and Surfaces A: Physicochemical and Engineering Aspects 597 (July 2020): 124764. http://dx.doi.org/10.1016/j.colsurfa.2020.124764.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
15

Zhang, Wanjun, Jing Ye, Yuanyuan Zhang, Qiwei Li, Xiawei Dong, Hui Jiang, and Xuemei Wang. "One-step facile synthesis of fluorescent gold nanoclusters for rapid bio-imaging of cancer cells and small animals." RSC Advances 5, no. 78 (2015): 63821–26. http://dx.doi.org/10.1039/c5ra11321k.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
16

Sha, Qiuyue, Ruixue Guan, Huiying Su, Liang Zhang, Bi-Feng Liu, Zhaoyu Hu, and Xin Liu. "Carbohydrate-protein template synthesized high mannose loading gold nanoclusters: A powerful fluorescence probe for sensitive Concanavalin A detection and specific breast cancer cell imaging." Talanta 218 (October 2020): 121130. http://dx.doi.org/10.1016/j.talanta.2020.121130.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
17

Xu, Shouming, Hong Yang, Kang Zhao, Jianguo Li, Liyun Mei, Yun Xie, and Anping Deng. "Simple and rapid preparation of orange-yellow fluorescent gold nanoclusters using dl-homocysteine as a reducing/stabilizing reagent and their application in cancer cell imaging." RSC Advances 5, no. 15 (2015): 11343–48. http://dx.doi.org/10.1039/c4ra13288b.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
18

Purohit, Rahul, and Sanjay Singh. "Fluorescent gold nanoclusters for efficient cancer cell targeting." International Journal of Nanomedicine Volume 13 (March 2018): 15–17. http://dx.doi.org/10.2147/ijn.s125003.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
19

Zhang, Qing, Mingying Yang, Ye Zhu, and Chuanbin Mao. "Metallic Nanoclusters for Cancer Imaging and Therapy." Current Medicinal Chemistry 25, no. 12 (April 19, 2018): 1379–96. http://dx.doi.org/10.2174/0929867324666170331122757.

Повний текст джерела
Анотація:
Background: Nanoclusters are made of a few to tens of atoms with a size below 2 nm. Compared with nanoparticles, they exhibited excellent properties, such as tunable fluorescence, ease of conjugation, high quantum yield and biocompatibility, which are highly desired in the development of cancer nanotheranostics. Hence, the metallic nanoclusters have emerged as a newcomer in cancer nanomedicines. This review aims to summarize recently developed approaches to preparing metallic nanoclusters, highlight their applications in cancer theranostics, and provide a brief outlook for the future developments of nanoclusters in nanomedicine. Method: We carried out a thorough literature search using online databases. The search was focused on a centered question. Irrelevant articles were excluded after further examination and directly relevant articles were included. The relevant articles were classified by the subjects and the information from these articles was synthesized. Results: One hundred and forty-three articles were included in this review. About eighty articles outlined the development in the synthetic methods of nanoclusters. The synthesis approaches include chemical reduction, photoreduction and so on. The progress in the application of gold and silver nanoclusters to cancer theranostics was described in fifteen and eight articles, respectively. The rest articles were about the advancements in the use of other metal nanoclusters and nanocluster nanocomposites as cancer theranostic agents. Conclusion: This review summarizes the synthesis and use of metallic nanoclusters or their nanocomposites as cancer theranostic agents. It confirms their importance, advantages and potentials in serving as a new generation of cancer theranostics in clinics.
Стилі APA, Harvard, Vancouver, ISO та ін.
20

Zhang, Jian, Yi Fu, Cecil V. Conroy, Zhenghua Tang, Ge Li, Richard Y. Zhao, and Gangli Wang. "Fluorescence Intensity and Lifetime Cell Imaging with Luminescent Gold Nanoclusters." Journal of Physical Chemistry C 116, no. 50 (December 6, 2012): 26561–69. http://dx.doi.org/10.1021/jp306036y.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
21

Jarockyte, Greta, Vilius Poderys, Virginijus Barzda, Vitalijus Karabanovas, and Ricardas Rotomskis. "Blood Plasma Stabilized Gold Nanoclusters for Personalized Tumor Theranostics." Cancers 14, no. 8 (April 8, 2022): 1887. http://dx.doi.org/10.3390/cancers14081887.

Повний текст джерела
Анотація:
Personalized cancer theranostics has a potential to increase efficiency of early cancer diagnostics and treatment, and to reduce negative side-effects. Protein-stabilized gold nanoclusters may serve as theranostic agents. To make gold nanoclusters personalized and highly biocompatible, the clusters were stabilized with human plasma proteins. Optical properties of synthesized nanoclusters were investigated spectroscopically, and possible biomedical application was evaluated using standard cell biology methods. The spectroscopic investigations of human plasma proteins stabilized gold nanoclusters revealed that a wide photoluminescence band in the optical tissue window is suitable for cancer diagnostics. High-capacity generation of singlet oxygen and other reactive oxygen species was also observed. Furthermore, the cluster accumulation in cancer cells and the photodynamic effect were evaluated. The results demonstrate that plasma proteins stabilized gold nanoclusters that accumulate in breast cancer cells and are non-toxic in the dark, while appear phototoxic under irradiation with visible light. The results positively confirm the utility of plasma protein stabilized gold nanoclusters for the use in cancer diagnostics and treatment.
Стилі APA, Harvard, Vancouver, ISO та ін.
22

Feng, Bo, Yanan Xing, Jinze Lan, Zhi Su, and Fu Wang. "Synthesis of MUC1 aptamer-stabilized gold nanoclusters for cell-specific imaging." Talanta 212 (May 2020): 120796. http://dx.doi.org/10.1016/j.talanta.2020.120796.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
23

Venkatesh, V., Akansha Shukla, Sri Sivakumar, and Sandeep Verma. "Purine-Stabilized Green Fluorescent Gold Nanoclusters for Cell Nuclei Imaging Applications." ACS Applied Materials & Interfaces 6, no. 3 (January 29, 2014): 2185–91. http://dx.doi.org/10.1021/am405345h.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
24

Han, Weiwei, Wenjiang Yang, Fuping Gao, Pengju Cai, Jianjun Wang, Sijia Wang, Jingquan Xue, Xueyun Gao, and Yu Liu. "Iodine-124 Labeled Gold Nanoclusters for Positron Emission Tomography Imaging in Lung Cancer Model." Journal of Nanoscience and Nanotechnology 20, no. 3 (March 1, 2020): 1375–82. http://dx.doi.org/10.1166/jnn.2020.17169.

Повний текст джерела
Анотація:
This work reports the synthesis, radiolabeling and imaging studies of iodine-124 labeled peptide modified gold nanoclusters (AuNCs) as positron emission tomography (PET) tracer for lung cancer. The novel modified Au nanoclusters were successfully synthesized by conjugation of tumortargeting peptide luteinizing hormone releasing hormone (LHRH) to human serum albumin (HAS) as a scaffold, resulting in 73% labeling yield of 124I-LHRH-HSA AuNCs. After rapid purification, the radiochemical purity was above 98%. Dynamic PET study in normal rats showed high liver accumulation and rapid lung clearance. Both the PET and fluorescence imaging in A549 xenografted tumor model demonstrated certain amount of tumor uptake. In orthotopic lung cancer model, the tumor sites could be clearly visualized between 2 to 5 hours in PET images. The higher radioactivity concentration in the left lung which inoculated orthotopic tumor than right lung also exhibited the targeting properties. The biological properties of this iodine-124 labeled nanoclusters afford potential applications for early diagnosis of lung cancer with PET.
Стилі APA, Harvard, Vancouver, ISO та ін.
25

Ga, Lu, and Jun Ai. "In Situ Imaging of Cancer Cell via Cancer Aptamer and its Fluorescent Clusters." Advanced Materials Research 998-999 (July 2014): 256–59. http://dx.doi.org/10.4028/www.scientific.net/amr.998-999.256.

Повний текст джерела
Анотація:
Silver nanoclusters synthesized using aptamer as matrices attracted more attention in the fields of medicine, pharmacology and biology. In this paper, we report a novel approach to sgc-8-C8-templated formation of fluorescent Ag nanoclusters and its application to bioimaging. This was further confirmed by flow cytometry. The cell toxicity (3-[4, 5-dimethylthiazolyl-2]-2, 5 -diphenyltetrazolium bromide, MTT) assay demonstrated that the silver nanoclusters has only little affect on the cytotoxicity to the cells. These results demonstrated that this aptamer-based method for labeling and imaging cellular protein is facile, effective.
Стилі APA, Harvard, Vancouver, ISO та ін.
26

Khandelia, Rumi, Satyapriya Bhandari, Uday Narayan Pan, Siddhartha Sankar Ghosh, and Arun Chattopadhyay. "Gold Nanocluster Embedded Albumin Nanoparticles for Two-Photon Imaging of Cancer Cells Accompanying Drug Delivery." Small 11, no. 33 (May 4, 2015): 4075–81. http://dx.doi.org/10.1002/smll.201500216.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
27

Hada, Alexandru-Milentie, Ana-Maria Craciun, and Simion Astilean. "Gold nanoclusters performing as contrast agents for non-invasive imaging of tissue-like phantoms via two-photon excited fluorescence lifetime imaging." Analyst 146, no. 23 (2021): 7126–30. http://dx.doi.org/10.1039/d1an01394g.

Повний текст джерела
Анотація:
Photoluminescent bovine serum albumin stabilized gold nanoclusters as reliable label-free FLIM contrast agents under near-infrared two-photon excitation inside cancer tissue-mimicking agarose phantoms.
Стилі APA, Harvard, Vancouver, ISO та ін.
28

Basu, Kingshuk, Kousik Gayen, Tulika Mitra, Abhishek Baral, Sib Sankar Roy, and Arindam Banerjee. "Different Color Emissive Copper Nanoclusters for Cancer Cell Imaging." ChemNanoMat 3, no. 11 (September 5, 2017): 808–14. http://dx.doi.org/10.1002/cnma.201700162.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
29

Suo, Zhiguang, Xialing Hou, Yu Liu, Feifei Xing, Yingying Chen та Lingyan Feng. "β-Lactoglobulin amyloid fibril-templated gold nanoclusters for cellular multicolor fluorescence imaging and colorimetric blood glucose assay". Analyst 145, № 21 (2020): 6919–27. http://dx.doi.org/10.1039/d0an01357a.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
30

Lu, Hui, Bin Chen, Yu Li, Jiye Shi, Jiang Li, Lihua Wang, Shihua Luo, Chunhai Fan, Jianlei Shen, and Jing Chen. "Benzyl-rich ligand engineering of the photostability of atomically precise gold nanoclusters." Chemical Communications 58, no. 14 (2022): 2395–98. http://dx.doi.org/10.1039/d1cc06467c.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
31

Yang, Ying, Shuxin Wang, Changzhi Xu, Anjian Xie, Yuhua Shen, and Manzhou Zhu. "Improved fluorescence imaging and synergistic anticancer phototherapy of hydrosoluble gold nanoclusters assisted by a novel two-level mesoporous canal structured silica nanocarrier." Chemical Communications 54, no. 22 (2018): 2731–34. http://dx.doi.org/10.1039/c8cc00685g.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
32

Shiang, Yen-Chun, Chih-Ching Huang, Wei-Yu Chen, Po-Cheng Chen, and Huan-Tsung Chang. "Fluorescent gold and silver nanoclusters for the analysis of biopolymers and cell imaging." Journal of Materials Chemistry 22, no. 26 (2012): 12972. http://dx.doi.org/10.1039/c2jm30563a.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
33

Nebu, John, J. S. Anjali Devi, R. S. Aparna, K. Abha, and George Sony. "Erlotinib conjugated gold nanocluster enveloped magnetic iron oxide nanoparticles–A targeted probe for imaging pancreatic cancer cells." Sensors and Actuators B: Chemical 257 (March 2018): 1035–43. http://dx.doi.org/10.1016/j.snb.2017.11.017.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
34

Ding, Changqin, and Yang Tian. "Gold nanocluster-based fluorescence biosensor for targeted imaging in cancer cells and ratiometric determination of intracellular pH." Biosensors and Bioelectronics 65 (March 2015): 183–90. http://dx.doi.org/10.1016/j.bios.2014.10.034.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
35

Zhang, Lu, BenChao Zheng, Rui Guo, Ying Miao, and Biao Li. "Bone marrow mesenchymal stem cell-mediated ultrasmall gold nanoclusters and hNIS gene synergize radiotherapy for breast cancer." Journal of Materials Chemistry B 9, no. 12 (2021): 2866–76. http://dx.doi.org/10.1039/d1tb00186h.

Повний текст джерела
Анотація:
BMSC can carry both human sodium iodide symporter (hNIS) and ultrasmall gold nanoclusters (usAuNCs) to target triple negative breast cancer (TNBC), achieving synergistically radioactive enhancement of 131I therapy.
Стилі APA, Harvard, Vancouver, ISO та ін.
36

Xue, J., Y. Liu, W. Han, and W. Yang. "Dual-modality liver function imaging agent: neolactosylated albumin stabilized gold nanoclusters." Nuclear Medicine and Biology 72-73 (July 2019): S41—S42. http://dx.doi.org/10.1016/s0969-8051(19)30303-8.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
37

Xu, Man-Man, Tong-Tong Jia, Bingjie Li, Wang Ma, Xiaoyuan Chen, Xueli Zhao, and Shuang-Quan Zang. "Tuning the properties of atomically precise gold nanoclusters for biolabeling and drug delivery." Chemical Communications 56, no. 62 (2020): 8766–69. http://dx.doi.org/10.1039/d0cc03498c.

Повний текст джерела
Анотація:
The fluorescent properties of atomically precise nanoclusters have been regulated by solvent-induced atomic structure transformation and cationic polymer-induced self-assembly for cell imaging and drug delivery.
Стилі APA, Harvard, Vancouver, ISO та ін.
38

Broekgaarden, Mans, Anne-Laure Bulin, Estelle Porret, Benjamin Musnier, Benoit Chovelon, Corinne Ravelet, Lucie Sancey, et al. "Surface functionalization of gold nanoclusters with arginine: a trade-off between microtumor uptake and radiotherapy enhancement." Nanoscale 12, no. 13 (2020): 6959–63. http://dx.doi.org/10.1039/d0nr01138j.

Повний текст джерела
Анотація:
Gold nanoclusters (AuNC) have strong potential for cancer imaging and therapy. We demonstrate that optimizing the surface chemistry of AuNCs for increased tumor uptake can significantly affect its potential to augment radiotherapy outcomes.
Стилі APA, Harvard, Vancouver, ISO та ін.
39

Wu, Xiaoxia, Yan Peng, Xiaomei Duan, Lingyan Yang, Jinze Lan, and Fu Wang. "Homologous Gold Nanoparticles and Nanoclusters Composites with Enhanced Surface Raman Scattering and Metal Fluorescence for Cancer Imaging." Nanomaterials 8, no. 10 (October 11, 2018): 819. http://dx.doi.org/10.3390/nano8100819.

Повний текст джерела
Анотація:
A large number of deaths from cancer can be attributed to the lack of effective early-stage diagnostic techniques. Thus, accurate and effective early diagnosis is a major research goal worldwide. With the unique phenomenon of localized surface plasmon resonance (LSPR), plasmonic nanomaterials have attracted considerable attention for applications in surface-enhanced Raman scattering (SERS) and metal-enhanced fluorescence (MEF). Both SERS and MEF are ultra-sensitive methods for the detection and identification of early tumor at molecular level. To combine the merits of the fast and accurate imaging of MEF and the stable and clear imaging of SERS, we propose a novel dual functional imaging nanoprobe based on gold nanoparticles and gold nanocluster composites (denoted AuNPC-RGD). The gold nanoparticles are used as LSPR substrates to realized enhancement of Raman or fluorescence signal, while the gold nanoclusters serve as a fluorophore for MEF imaging, and exhibit better biocompatibility and stability. Furthermore, target molecule of cyclic Arg-Gly-Asp (cRGD) is incorporated into the composite to improve delivery efficiency, selectivity and imaging accuracy. These integrated properties endow AuNPC-RGD composites with outstanding biocompatibility and excellent imaging abilities, which could be used to achieve accurate and effective diagnosis for early cancer.
Стилі APA, Harvard, Vancouver, ISO та ін.
40

Tian, Ye, Ming Wei, Lijun Wang, Yuankai Hong, Dan Luo, and Yinlin Sha. "Two-Photon Time-Gated In Vivo Imaging of Dihydrolipoic-Acid-Decorated Gold Nanoclusters." Materials 14, no. 24 (December 15, 2021): 7744. http://dx.doi.org/10.3390/ma14247744.

Повний текст джерела
Анотація:
Due to the unique advantages of two-photon technology and time-resolved imaging technology in the biomedical field, attention has been paid to them. Gold clusters possess excellent physicochemical properties and low biotoxicity, which make them greatly advantageous in biological imaging, especially for in vivo animal imaging. A gold nanocluster was coupled with dihydrolipoic acid to obtain a functionalized nanoprobe; the material displayed significant features, including a large two-photon absorption cross-section (up to 1.59 × 105 GM) and prolonged fluorescence lifetime (>300 ns). The two-photon and time-resolution techniques were used to perform cell imaging and in vivo imaging.
Стилі APA, Harvard, Vancouver, ISO та ін.
41

Chen, Haiyan, Bowen Li, Chuan Wang, Xin Zhang, Zhengqi Cheng, Xi Dai, Rui Zhu, and Yueqing Gu. "Characterization of a fluorescence probe based on gold nanoclusters for cell and animal imaging." Nanotechnology 24, no. 5 (January 11, 2013): 055704. http://dx.doi.org/10.1088/0957-4484/24/5/055704.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
42

Duan, Ying, Ruiping Duan, Rui Liu, Man Guan, Wenjuan Chen, Jingjing Ma, Mingmao Chen, Bo Du, and Qiqing Zhang. "Chitosan-Stabilized Self-Assembled Fluorescent Gold Nanoclusters for Cell Imaging and Biodistribution in Vivo." ACS Biomaterials Science & Engineering 4, no. 3 (February 9, 2018): 1055–63. http://dx.doi.org/10.1021/acsbiomaterials.7b00975.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
43

Yang, Xuan, Linfeng Gan, Lei Han, Dan Li, Jin Wang, and Erkang Wang. "Facile preparation of chiral penicillamine protected gold nanoclusters and their applications in cell imaging." Chemical Communications 49, no. 23 (2013): 2302. http://dx.doi.org/10.1039/c3cc00200d.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
44

Kong, Yifei, Jun Chen, Feng Gao, Rik Brydson, Benjamin Johnson, George Heath, Yue Zhang, Lin Wu, and Dejian Zhou. "Near-infrared fluorescent ribonuclease-A-encapsulated gold nanoclusters: preparation, characterization, cancer targeting and imaging." Nanoscale 5, no. 3 (2013): 1009–17. http://dx.doi.org/10.1039/c2nr32760k.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
45

Volotskova, O., Y. Osakada, G. Pratx, B. Cui, and L. Xing. "Development of a Novel Optical Imaging Probe for Cancer Detection: X-ray Activated Gold Nanoclusters." International Journal of Radiation Oncology*Biology*Physics 87, no. 2 (October 2013): S646. http://dx.doi.org/10.1016/j.ijrobp.2013.06.1710.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
46

Sood, Kritika, and Asifkhan Shanavas. "The Role of Gold Nanoclusters as Emerging Theranostic Agents for Cancer Management." Current Pathobiology Reports 9, no. 2 (May 25, 2021): 33–42. http://dx.doi.org/10.1007/s40139-021-00222-4.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
47

Zhao, Yongfeng, Deborah Sultan, Lisa Detering, Hannah Luehmann, and Yongjian Liu. "Facile synthesis, pharmacokinetic and systemic clearance evaluation, and positron emission tomography cancer imaging of64Cu–Au alloy nanoclusters." Nanoscale 6, no. 22 (2014): 13501–9. http://dx.doi.org/10.1039/c4nr04569f.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
48

Fahmi, Mochamad Zakki, Keng-Liang Ou, Jem-Kun Chen, Ming-Hua Ho, Shin-Hwa Tzing, and Jia-Yaw Chang. "Development of bovine serum albumin-modified hybrid nanoclusters for magnetofluorescence imaging and drug delivery." RSC Adv. 4, no. 62 (2014): 32762–72. http://dx.doi.org/10.1039/c4ra05785f.

Повний текст джерела
Анотація:
Magnetofluorescent nanoclusters containing oil-soluble nanoparticles of MnFe2O4 and AgInS2–ZnS QDs are prepared. The nanoclusters possess photoluminescent and magnetic properties as well as an excellent specific targeting and drug delivery capability on HeLa cancer cell.
Стилі APA, Harvard, Vancouver, ISO та ін.
49

Tan, Shih-Hua, Sibidou Yougbaré, Hsueh-Liang Chu, Tsung-Rong Kuo, and Tsai-Mu Cheng. "Hemoglobin-Conjugated Gold Nanoclusters for Qualitative Analysis of Haptoglobin Phenotypes." Polymers 12, no. 10 (September 29, 2020): 2242. http://dx.doi.org/10.3390/polym12102242.

Повний текст джерела
Анотація:
Designing a facile and rapid detection method for haptoglobin (Hp) phenotypes in human blood plasma is urgently needed to meet clinic requirements in hemolysis theranostics. In this work, a novel approach to qualitatively analyze Hp phenotypes was developed using a fluorescent probe of gold nanoclusters (AuNCs). Hemoglobin-conjugated (Hb)-AuNCs were successfully synthesized with blue-green fluorescence and high biocompatibility via one-pot synthesis. The fluorescence of Hb-AuNCs comes from the ligand-metal charge transfer between surface ligands of Hb and the gold cores with high oxidation states. The biocompatibility assays including cell viability and fluorescence imaging, demonstrated high biocompatibility of Hb-AuNCs. For the qualitative analysis, three Hp phenotypes in plasma, Hp 1-1, Hp 2-1, and Hp 2-2, were successfully discriminated according to changes in the fluorescence intensity and peak position of the maximum intensity of Hb-AuNCs. Our work provides a practical method with facile and rapid properties for the qualitative analysis of three Hp phenotypes in human blood plasma.
Стилі APA, Harvard, Vancouver, ISO та ін.
50

Khandelia, Rumi, Satyapriya Bhandari, Uday Narayan Pan, Siddhartha Sankar Ghosh, and Arun Chattopadhyay. "Drug Delivery: Gold Nanocluster Embedded Albumin Nanoparticles for Two-Photon Imaging of Cancer Cells Accompanying Drug Delivery (Small 33/2015)." Small 11, no. 33 (September 2015): 4074. http://dx.doi.org/10.1002/smll.201570201.

Повний текст джерела
Стилі APA, Harvard, Vancouver, ISO та ін.
Ми пропонуємо знижки на всі преміум-плани для авторів, чиї праці увійшли до тематичних добірок літератури. Зв'яжіться з нами, щоб отримати унікальний промокод!

До бібліографії