引文搜索指导
查看引文搜索小技巧我们的引文数据库包含引用Promega产品的同行评审期刊文章。有以下多种方法可以搜索引文:
- 在文本字段中直接输入关键词(包括目录号、产品名称或作者)。
- 在“选择应用方向”列表中选择一个或多个“应用方向”。
- 选择应用方向并输入关键词。直接按照文章发表年份搜索。
对于使用特定Promega产品的引文,我们建议您按产品名称或目录号进行搜索。对于范围更广的搜索,建议您选择一个或两个应用方向查看结果,然后通过向搜索条件添加关键词来缩小焦点。
符合搜索条件的引文将按时间倒序显示(较新的引用文章优先显示)。关于特定产品的更多信息,请参阅 引文及技术文章分析或 操作说明。
筛选项清空
- 研究/应用方向
- 产品/技术
选择引文起止时间
-
-
In vitro and in vivo analysis of extracellular vesicle-mediated metastasis using a bright, red-shifted bioluminescent reporter protein
Advanced Genetics | 2022 | 查看原文 |
作者:Gloria I Perez, David Broadbent, Ahmed A Zarea, Benedikt Dolgikh, Matthew P Bernard, Alicia Withrow, Amelia McGill, Victoria Toomajian, Lukose K Thampy, Jack Harkema, Joel R Walker, Thomas A Kirkland&
- 摘要:Cancer cells produce heterogeneous extracellular vesicles (EVs) as mediators of intercellular communication. This study focuses on a novel method to image EV subtypes and their biodistribution in vivo. A red-shifted bioluminescence resonance energy transfer (BRET) EV reporter is developed, called PalmReNL, which allows for highly sensitive EV tracking in vitro and in vivo. PalmReNL enables the authors to study the common surface molecules across EV subtypes that determine EV organotropism and their functional differences in cancer progression. Regardless of injection routes, whether retro-orbital or intraperitoneal, PalmReNL positive EVs, isolated from murine mammary carcinoma cells, localized to the lungs. The early appearance of metastatic foci in the lungs of mammary tumor-bearing mice following multiple intraperitoneal injections of the medium and large EV (m/lEV)-enriched fraction derived from mammary carcinoma cells is demonstrated. In addition, the results presented here show that tumor cell-derived m/lEVs act on distant tissues through upregulating LC3 expression within the lung.展开
关键词:外泌体,生物分布,生物发光共振能量转移,Nano-Glo Fluorofurimazine 活体成像底物,FFz
应用产品:CellTiter-Fluor™ Cell Viability AssayNano-Glo® Fluorofurimazine In Vivo Substrate (FFz)
-
-
-
Pharmacokinetics and biodistribution of extracellular vesicles administered intravenously and intranasally to Macaca nemestrina
journal of extracelluar Biol | 2022 | 查看原文 |
作者:Tom Driedonks, Linglei Jiang, Bess Carlson, Zheng Han, Guanshu Liu, Suzanne E Queen, Erin N Shirk, Olesia Gololobova, Zhaohao Liao, Lyle H Nyberg, Gabriela Lima, Liliia Paniushkina, Marta Garcia-
- 摘要:Extracellular vesicles (EVs) have potential in disease treatment since they can be loaded with therapeutic molecules and engineered for retention by specific tissues. However, questions remain on optimal dosing, administration, and pharmacokinetics. Previous studies have addressed biodistribution and pharmacokinetics in rodents, but little evidence is available for larger animals. Here, we investigated the pharmacokinetics and biodistribution of Expi293F-derived EVs labelled with a highly sensitive nanoluciferase reporter (palmGRET) in a non-human primate model (Macaca nemestrina), comparing intravenous (IV) and intranasal (IN) administration over a 125-fold dose range. We report that EVs administered IV had longer circulation times in plasma than previously reported in mice and were detectable in cerebrospinal fluid (CSF) after 30-60 minutes. EV association with PBMCs, especially B-cells, was observed as early as one minute post-administration. EVs were detected in liver and spleen within one hour of IV administration. However, IN delivery was minimal, suggesting that pretreatment approaches may be needed in large animals. Furthermore, EV circulation times strongly decreased after repeated IV administration, possibly due to immune responses and with clear implications for xenogeneic EV-based therapeutics. We hope that our findings from this baseline study in macaques will help to inform future research and therapeutic development of EVs.展开
关键词:biodistribution; drug delivery; extracellular vesicles; macaques; nanomedicine; pharmacokinetics; therapeutics
应用产品:Nano-Glo® Fluorofurimazine In Vivo Substrate (FFz)Nano-Glo® Luciferase Assay
-
-
-
Orthogonal inducible control of Cas13 circuits enables programmable RNA regulation in mammalian cells
bioRxiv | 2023 | 查看原文 |
作者:Yage Ding, Cristina Tous, Jaehoon Choi, Jingyao Chen, Wilson W Wong
- 摘要:RNA plays an indispensable role in mammalian cell functions. Cas13, a class of RNA-guided ribonuclease, is a flexible tool for modifying and regulating coding and non-coding RNAs, with enormous potential for creating new cell functions. However, the lack of control over Cas13 activity has limited its cell engineering capability. Here, we present the CRISTAL ( C ontrol of R NA with Inducible S pli T C A s13 Orthologs and Exogenous L igands) platform. CRISTAL is powered by a collection (10 total) of orthogonal split inducible Cas13s that can be turned ON or OFF via small molecules in multiple cell types, providing precise temporal control. Also, we engineered Cas13 logic circuits that can respond to endogenous signaling and exogenous small molecule inputs. Furthermore, the orthogonality, low leakiness, and high dynamic range of our inducible Cas13d and Cas13b enable the design and construction of a robust incoherent feedforward loop, leading to near-perfect and tunable adaptation response. Finally, using our inducible Cas13s, we achieve simultaneous multiplexed control of multiple genes in vitro and in mice. Together, our CRISTAL design represents a powerful platform for precisely regulating RNA dynamics to advance cell engineering and elucidate RNA biology.展开
关键词:核糖核酸酶,Cas13,RNA调控,Nano-Glo Fluorofurimazine 活体成像底物
-
-
-
Application of bioluminescence resonance energy transfer-based cell tracking approach in bone tissue engineering
Sage Journals Home | 2021 | 查看原文 |
作者:Lufei Wang, Dong Joon Lee, Han Han, Lixing Zhao, Hiroshi Tsukamoto, Yong-IL Kim, Adele M Musicant, Kshitij Parag-Sharma, Xiangxiang Hu, Henry C Tseng, Jen-Tsan Chi, Zhengyan Wang, Antonio L Ameli
- 摘要:Bioluminescent imaging (BLI) has emerged as a popular in vivo tracking modality in bone regeneration studies stemming from its clear advantages: non-invasive, real-time, and inexpensive. We recently adopted bioluminescence resonance energy transfer (BRET) principle to improve BLI cell tracking and generated the brightest bioluminescent signal known to date, which thus enables more sensitive real-time cell tracking at deep tissue level. In the present study, we brought BRET-based cell tracking strategy into the field of bone tissue engineering for the first time. We labeled rat mesenchymal stem cells (rMSCs) with our in-house BRET-based GpNLuc reporter and evaluated the cell tracking efficacy both in vitro and in vivo. In scaffold-free spheroid 3D culture system, using BRET-based GpNLuc labeling resulted in significantly better correlation to cell numbers than a fluorescence based approach. In scaffold-based 3D culture system, GpNLuc-rMSCs displayed robust bioluminescence signals with minimal background noise. Furthermore, a tight correlation between BLI signal and cell number highlighted the robust reliability of using BRET-based BLI. In calvarial critical sized defect model, robust signal and the consistency in cell survival evaluation collectively supported BRET-based GpNLuc labeling as a reliable approach for non-invasively tracking MSC. In summary, BRET-based GpNLuc labeling is a robust, reliable, and inexpensive real-time cell tracking method, which offers a promising direction for the technological innovation of BLI and even non-invasive tracking systems, in the field of bone tissue engineering.展开
关键词:Bioluminescent imaging; bioluminescence resonance energy transfer; bone tissue engineering; cell tracking; mesenchymal stem cells.
-
-
-
Locoregionally administered B7-H3-targeted CAR T cells for treatment of atypical teratoid/rhabdoid tumors
Nature Medicine volume | 2020 | 查看原文 |
作者:Johanna Theruvath, Elena Sotillo, Christopher W. Mount, Claus Moritz Graef, Alberto Delaidelli, Sabine Heitzeneder, Louai Labanieh, Shaurya Dhingra, Amaury Leruste, Robbie G. Majzner, Peng Xu, Sabine
- 摘要:Atypical teratoid/rhabdoid tumors (ATRTs) typically arise in the central nervous system (CNS) of children under 3 years of age. Despite intensive multimodal therapy (surgery, chemotherapy and, if age permits, radiotherapy), median survival is 17 months1,2. We show that ATRTs robustly express B7-H3/CD276 that does not result from the inactivating mutations in SMARCB1 (refs. 3,4), which drive oncogenesis in ATRT, but requires residual SWItch/Sucrose Non-Fermentable (SWI/SNF) activity mediated by BRG1/SMARCA4. Consistent with the embryonic origin of ATRT5,6, B7-H3 is highly expressed on the prenatal, but not postnatal, brain. B7-H3.BB.z-chimeric antigen receptor (CAR) T cells administered intracerebroventricularly or intratumorally mediate potent antitumor effects against cerebral ATRT xenografts in mice, with faster kinetics, greater potency and reduced systemic levels of inflammatory cytokines compared to CAR T cells administered intravenously. CAR T cells administered ICV also traffic from the CNS into the periphery; following clearance of ATRT xenografts, B7-H3.BB.z-CAR T cells administered intracerebroventricularly or intravenously mediate antigen-specific protection from tumor rechallenge, both in the brain and periphery. These results identify B7-H3 as a compelling therapeutic target for this largely incurable pediatric tumor and demonstrate important advantages of locoregional compared to systemic delivery of CAR T cells for the treatment of CNS malignancies.展开
关键词:CAR-T,B7-H3,儿科肿瘤,中枢神经系统恶性肿瘤,
-
-
-
Visualizing cell-cell communication using synthetic notch activated MRI
bioRxiv https://pubmed.ncbi.nlm.nih.gov/36893267/ | 2023 | 查看原文 |
作者:TianDuo Wang, Yuanxin Chen, Nivin N Nystrom, Shirley Liu, Yanghao Fu, Francisco M Martinez, Timothy J Scholl, John A Ronald
- 摘要:Cell–cell communication plays a fundamental role in multicellular organisms. Cell-based cancer immunotherapies rely on the ability of innate or engineered receptors on immune cells to engage specific antigens on cancer cells to induce tumor kill. To improve the development and translation of these therapies, imaging tools capable of noninvasively and spatiotemporally visualizing immune-cancer cell interactions would be highly valuable. Using the synthetic Notch (SynNotch) system, we engineered T cells that upon interaction with a chosen antigen (CD19) on neighboring cancer cells induce the expression of optical reporter genes and the human-derived, magnetic resonance imaging (MRI) reporter gene organic anion transporting polypeptide 1B3 (OATP1B3). Administration of engineered T cells induced the antigen-dependent expression of all our reporter genes in mice bearing CD19-positive tumors but not CD19-negative tumors. Notably, due to the high spatial resolution and tomographic nature of MRI, contrast-enhanced foci within CD19-positive tumors representing OATP1B3-expressing T cells were clearly visible and their distribution was readily mapped. We then extended this technology onto human natural killer-92 (NK-92) cells, observing similar CD19-dependent reporter activity in tumor-bearing mice. Furthermore, we show that when delivered intravenously, engineered NK-92 cells can be detected via bioluminescence imaging in a systemic cancer model. With continued work, this highly modular imaging strategy could aid in the monitoring of cell therapies in patients and, beyond this, augment our understanding of how different cell populations interact within the body during normal physiology or disease.展开
关键词:MRI; cell therapy; cell–cell communication; reporter gene; synthetic Notch
-
-
-
Enhancing anti-EGFRvIII CAR T cell therapy against glioblastoma with a paracrine SIRPγ-derived CD47 blocker
BioRxiv | 2023 | 查看原文 |
作者:Tomás A. MartinsNazanin TatariDeniz KaymakSabrina HoganEwelina M. BartoszekRonja WieboldtMarie-Françoise RitzAlicia BuckMarta McDaidAlexandra GerberAisha BeshirovaManina M. EtterAnja HeiderTala ShekarianHayget MohamedPhilip SchmassmannInes AbelL
- 摘要:A major challenge for chimeric antigen receptor (CAR) T cell therapy against glioblastoma (GBM) is its immunosuppressive tumor microenvironment (TME), which is densely populated and supported by protumoral glioma-associated microglia and macrophages (GAMs). Targeting of CD47, a “don’t-eat-me” signal overexpressed by tumor cells, disrupts the CD47-SIRPα axis and induces GAM phagocytic function. However, antibody-mediated CD47 blockade monotherapy is associated with toxicity and low bioavailability in solid tumors. To overcome these limitations, we combined local CAR T cell therapy with paracrine GAM modulation for more effective elimination of GBM. To this end, we engineered a new CAR T cell against epidermal growth factor receptor variant III (EGFRvIII) that constitutively secretes a SIRPγ-related protein (SGRP) with high affinity to CD47. Anti-EGFRvIII-SGRP CAR T cells eliminated EGFRvIII+GBM in a dose-dependent mannerin vitroand eradicated orthotopically xenografted EGFRvIII-mosaic GBM by locoregional applicationin vivo.This resulted in significant tumor-free long-term survival, followed by partial tumor control upon tumor re-challenge. The combination of anti-CD47 antibodies with anti-EGFRvIII CAR T cells failed to achieve a similar therapeutic effect, underscoring the importance of sustained paracrine GAM modulation. Multidimensional brain immunofluorescence microscopy and in-depth spectral flow cytometry on GBM-xenografted brains showed that anti-EGFRvIII-SGRP CAR T cells accelerated GBM clearance, increased CD68+cell trafficking to tumor scar sites, and induced myeloid-mediated tumor cell uptake. Additionally, in a peripheral lymphoma mouse xenograft model, anti-CD19-SGRP CAR T cells had superior efficacy compared to conventional anti-CD19 CAR T cells. Validation on human GBM explants revealed that anti-EGFRvIII-SGRP CAR T cells had similar tumor-killing capacity to anti-EGFRvIII CAR monotherapy, but showed a slight improvement in maintenance of tumor-infiltrated CD14+myeloid cells. Thus, local anti-EGFRvIII-SGRP CAR T cell therapy combines the potent antitumor effect of engineered T cells with the modulation of the surrounding innate immune TME, resulting in the additive elimination of bystander EGFRvIII-tumor cells in a manner that overcomes major mechanisms of CAR T cell therapy resistance, including tumor innate immune suppression and antigen escape.展开
关键词:GBM, EGFRvIII, CAR T cell therapy, CD47, GAM, microglia modulation, SGRP, immunotherapy
应用产品:Nano-Glo® Fluorofurimazine In Vivo Substrate (FFz)Trypsin Gold, Mass Spectrometry Grade
-
-
-
NanoBiT System and Hydrofurimazine for Optimized Detection of Viral Infection in Mice—A Novel in Vivo Imaging Platform
International journal of Molecular Science | 2020 | 查看原文 |
作者:Natasa Gaspar,Giorgia Zambito,Iris J. C. Dautzenberg,Steve J. Cramer,Rob C. Hoeben,Clemens Lowik,Joel R. Walker,Thomas A. Kirkland,Thomas P. Smith,Wytske M. van Weerden,Jeroen de Vrij and Laura Mez
- 摘要:Reporter genes are used to visualize intracellular biological phenomena, including viral infection. Here we demonstrate bioluminescent imaging of viral infection using the NanoBiT system in combination with intraperitoneal injection of a furimazine analogue, hydrofurimazine. This recently developed substrate has enhanced aqueous solubility allowing delivery of higher doses for in vivo imaging. The small high-affinity peptide tag (HiBiT), which is only 11 amino-acids in length, was engineered into a clinically used oncolytic adenovirus, and the complementary large protein (LgBiT) was constitutively expressed in tumor cells. Infection of the LgBiT expressing cells with the HiBiT oncolytic virus will reconstitute NanoLuc in the cytosol of the cell, providing strong bioluminescence upon treatment with substrate. This new bioluminescent system served as an early stage quantitative viral transduction reporter in vitro and also in vivo in mice, for longitudinal monitoring of oncolytic viral persistence in infected tumor cells. This platform provides novel opportunities for studying the biology of viruses in animal models.展开
关键词:bioluminescence imaging; hibit tag; hydrofurimazine; nanobit system; oncolytic virus
-
-
-
Highly Sensitive Real-Time In Vivo Imaging of an Influenza Reporter Virus Reveals Dynamics of Replication and Spread
journal of Virology | 2013 | 查看原文 |
作者:Vy Tran, Lindsey A. Moser, Daniel S. Poole, Andrew Mehle
- 摘要:The continual public health threat posed by the emergence of novel influenza viruses necessitates the ability to rapidly monitor infection and spread in experimental systems. To analyze real-time infection dynamics, we have created a replication-competent influenza reporter virus suitable for in vivo imaging. The reporter virus encodes the small and bright NanoLuc luciferase whose activity serves as an extremely sensitive readout of viral infection. This virus stably maintains the reporter construct and replicates in culture and in mice with near-native properties. Bioluminescent imaging of the reporter virus permits serial observations of viral load and dissemination in infected animals, even following clearance of a sublethal challenge. We further show that the reporter virus recapitulates known restrictions due to host range and antiviral treatment, suggesting that this technology can be applied to studying emerging influenza viruses and the impact of antiviral interventions on infections in vivo. These results describe a generalizable method to quickly determine the replication and pathogenicity potential of diverse influenza strains in animals.展开
关键词:流感病毒,NanoLuc荧光素酶,生物发光成像
-
-
-
In Vivo Live Imaging of Oncolytic Mammalian Orthoreovirus Expressing NanoLuc Luciferase in Tumor Xenograft Mice
journal of Virology | 2019 | 查看原文 |
作者:Yuta Kanai, Takahiro Kawagishi, Yoshiharu Matsuura, Takeshi Kobayashi
- 摘要:Wild-type mammalian reoviruses (MRVs) have been evaluated as oncolytic agents against various cancers; however, genetic modification methods for improving MRV agents have not been exploited fully. In the present study, using MRV strain T1L, we generated a reporter MRV that expresses a NanoLuc luciferase (NLuc) gene and used it for noninvasive imaging of MRV infection in tumor xenograft mice. NLuc and a P2A self-cleaving peptide gene cassette were placed upstream of the L1 gene open reading frame to enable bicistronic expression of NLuc and the L1 gene product. BALB/c nude mice intranasally infected with MRV expressing NLuc (rsT1L-NLuc) displayed bioluminescent signals in the chest area at 4 days postinfection (dpi), which is consistent with natural MRV infection in the lung. Furthermore, to monitor tumor-selective infection by MRV, nude mice bearing human cancer xenografts were infected intravenously with rsT1L-NLuc. Bioluminescent signals were detected in tumors as early as 3 dpi and persisted for 2 months. The results demonstrate the utility of an autonomous replicating reporter MRV for noninvasive live imaging of replicating oncolytic MRV agents.展开
关键词:呼肠孤病毒(MRVs),NanoLuc荧光素酶,生物发光成像
-
- 技术支持
-
电话: 010-58256268
邮箱: info@promega.com