22 results on '"Fabrice Laroche"'
Search Results
2. Extracellular Nanovesicle Enhanced Gene Transfection Using Polyethyleneimine in HEK293T Cells and Zebrafish Embryos
- Author
-
Zhenzhen Zhang, Kai Wen, Chao Zhang, Fabrice Laroche, Zhenglong Wang, Qiang Zhou, Zunfeng Liu, Jan Pieter Abrahams, and Xiang Zhou
- Subjects
gene transfection ,extracellular nanovesicles ,zebra fish ,polyethyleneimine ,efficiency ,Biotechnology ,TP248.13-248.65 - Abstract
It is a hot topic to improve efficiency and decrease toxicity of gene transfection reagents. The extracellular nanovesicles (EVs) that are released by cells play an important role in intercellular communication and are naturally designed for genetic exchange between cells. Here, we show that the EVs have a large beneficial effect in polyethyleneimine (PEI)-mediated transfection of a GFP-encoding plasmid into HEK293T cells. An improvement of transfection efficiency of ~500% and a decrease in toxicity were observed in a specific concentration range of PEI. The EVs also greatly improved the transfection of the same plasmid into zebrafish embryos. To verify the generality of this gene transfection approach, we also tested the cell viability and gene transfection efficiency using two other plasmids (EpTEN and ELuc) and in another cell line (A549). The measured increase in transfection efficiency makes EV a promising candidate for enhancement of the quality of current PEI-based transfection technique.
- Published
- 2020
- Full Text
- View/download PDF
3. Functional and genomic analyses reveal therapeutic potential of targeting β-catenin/CBP activity in head and neck cancer
- Author
-
Vinay K. Kartha, Khalid A. Alamoud, Khikmet Sadykov, Bach-Cuc Nguyen, Fabrice Laroche, Hui Feng, Jina Lee, Sara I. Pai, Xaralabos Varelas, Ann Marie Egloff, Jennifer E. Snyder-Cappione, Anna C. Belkina, Manish V. Bais, Stefano Monti, and Maria A. Kukuruzinska
- Subjects
HNSCC ,β-Catenin/CBP transcriptional activity ,Aggressive tumor cells ,ICG-001 ,TCGA ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy characterized by tumor heterogeneity, locoregional metastases, and resistance to existing treatments. Although a number of genomic and molecular alterations associated with HNSCC have been identified, they have had limited impact on the clinical management of this disease. To date, few targeted therapies are available for HNSCC, and only a small fraction of patients have benefited from these treatments. A frequent feature of HNSCC is the inappropriate activation of β-catenin that has been implicated in cell survival and in the maintenance and expansion of stem cell-like populations, thought to be the underlying cause of tumor recurrence and resistance to treatment. However, the therapeutic value of targeting β-catenin activity in HNSCC has not been explored. Methods We utilized a combination of computational and experimental profiling approaches to examine the effects of blocking the interaction between β-catenin and cAMP-responsive element binding (CREB)-binding protein (CBP) using the small molecule inhibitor ICG-001. We generated and annotated in vitro treatment gene expression signatures of HNSCC cells, derived from human oral squamous cell carcinomas (OSCCs), using microarrays. We validated the anti-tumorigenic activity of ICG-001 in vivo using SCC-derived tumor xenografts in murine models, as well as embryonic zebrafish-based screens of sorted stem cell-like subpopulations. Additionally, ICG-001-inhibition signatures were overlaid with RNA-sequencing data from The Cancer Genome Atlas (TCGA) for human OSCCs to evaluate its association with tumor progression and prognosis. Results ICG-001 inhibited HNSCC cell proliferation and tumor growth in cellular and murine models, respectively, while promoting intercellular adhesion and loss of invasive phenotypes. Furthermore, ICG-001 preferentially targeted the ability of subpopulations of stem-like cells to establish metastatic tumors in zebrafish. Significantly, interrogation of the ICG-001 inhibition-associated gene expression signature in the TCGA OSCC human cohort indicated that the targeted β-catenin/CBP transcriptional activity tracked with tumor status, advanced tumor grade, and poor overall patient survival. Conclusions Collectively, our results identify β-catenin/CBP interaction as a novel target for anti-HNSCC therapy and provide evidence that derivatives of ICG-001 with enhanced inhibitory activity may serve as an effective strategy to interfere with aggressive features of HNSCC.
- Published
- 2018
- Full Text
- View/download PDF
4. Towards Resolving the Pro- and Anti-Tumor Effects of the Aryl Hydrocarbon Receptor
- Author
-
Supraja Narasimhan, Elizabeth Stanford Zulick, Olga Novikov, Ashley J. Parks, Jennifer J. Schlezinger, Zhongyan Wang, Fabrice Laroche, Hui Feng, Francesca Mulas, Stefano Monti, and David H. Sherr
- Subjects
aryl hydrocarbon receptor ,cancer ,AHR agonist ,AHR antagonist ,cancer therapeutics ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
We have postulated that the aryl hydrocarbon receptor (AHR) drives the later, more lethal stages of some cancers when chronically activated by endogenous ligands. However, other studies have suggested that, under some circumstances, the AHR can oppose tumor aggression. Resolving this apparent contradiction is critical to the design of AHR-targeted cancer therapeutics. Molecular (siRNA, shRNA, AHR repressor, CRISPR-Cas9) and pharmacological (AHR inhibitors) approaches were used to confirm the hypothesis that AHR inhibition reduces human cancer cell invasion (irregular colony growth in 3D Matrigel cultures and Boyden chambers), migration (scratch wound assay) and metastasis (human cancer cell xenografts in zebrafish). Furthermore, these assays were used for a head-to-head comparison between AHR antagonists and agonists. AHR inhibition or knockdown/knockout consistently reduced human ER−/PR−/Her2− and inflammatory breast cancer cell invasion, migration, and metastasis. This was associated with a decrease in invasion-associated genes (e.g., Fibronectin, VCAM1, Thrombospondin, MMP1) and an increase in CDH1/E-cadherin, previously associated with decreased tumor aggression. Paradoxically, AHR agonists (2,3,7,8-tetrachlorodibenzo-p-dioxin and/or 3,3′-diindolylmethane) similarly inhibited irregular colony formation in Matrigel and blocked metastasis in vivo but accelerated migration. These data demonstrate the complexity of modulating AHR activity in cancer while suggesting that AHR inhibitors, and, under some circumstances, AHR agonists, may be useful as cancer therapeutics.
- Published
- 2018
- Full Text
- View/download PDF
5. Ultrabright fluorescent silica nanoparticles for in vivo targeting of xenografted human tumors and cancer cells in zebrafish
- Author
-
Xiaodan Qin, Fabrice Laroche, Berney Peng, Igor M. Sokolov, Shajesh Palantavida, Maxim Dokukin, Saquib Ahmed M. A. Peerzade, and Hui Feng
- Subjects
Transplantation, Heterologous ,02 engineering and technology ,Polyethylene glycol ,010402 general chemistry ,01 natural sciences ,Article ,Polyethylene Glycols ,HeLa ,chemistry.chemical_compound ,Folic Acid ,In vivo ,Neoplasms ,PEG ratio ,Animals ,Humans ,General Materials Science ,Particle Size ,Zebrafish ,biology ,Optical Imaging ,Silicon Dioxide ,021001 nanoscience & nanotechnology ,biology.organism_classification ,Fluorescence ,0104 chemical sciences ,Transplantation ,chemistry ,Cancer cell ,Biophysics ,Nanoparticles ,Female ,Particle size ,0210 nano-technology ,Porosity ,HeLa Cells - Abstract
New ultrabright fluorescent silica nanoparticles capable of the fast targeting of epithelial tumors in vivo are presented. The as-synthesized folate-functionalized ultrabright particles of 30-40 nm are 230 times brighter than quantum dots (QD450) and 50% brighter than the polymer dots with similar spectra (excitation 365 nm and emission 486 nm). To decrease non-specific targeting, particles are coated with polyethylene glycol (PEG). We demonstrate the in vivo targeting of xenographic human cervical epithelial tumors (HeLa cells) using zebrafish as a model system. The particles target tumors (and probably even individual HeLa cells) as small as 10-20 microns within 20-30 minutes after blood injection. To demonstrate the advantages of ultrabrightness, we repeated the experiments with similar but 200× less bright particles. Compared to those, ultrabright particles showed ∼3× faster tumor detection and ∼2× higher relative fluorescent contrast of tumors/cancer cells.
- Published
- 2019
6. UFD1 contributes to MYC-mediated leukemia aggressiveness through suppression of the proapoptotic unfolded protein response
- Author
-
Guang Lan Zhang, Michelle A. Kelliher, L. Wang, Ching-Ti Liu, Leah N Huiting, Justine E. Roderick, B. Li, Jae Won Choi, Y. W. Wang, Y. Samaha, Hui Feng, Nicole M. Anderson, and Fabrice Laroche
- Subjects
Male ,Transcriptional Activation ,0301 basic medicine ,Cancer Research ,Transcription, Genetic ,Down-Regulation ,Apoptosis ,Endoplasmic-reticulum-associated protein degradation ,Endoplasmic Reticulum ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins c-myc ,eIF-2 Kinase ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein kinase A ,Zebrafish ,Transcription Factor CHOP ,EIF-2 kinase ,biology ,Ubiquitin ,Chemistry ,Endoplasmic reticulum ,Intracellular Signaling Peptides and Proteins ,Proteins ,Hematology ,Endoplasmic Reticulum Stress ,Up-Regulation ,Cell biology ,Adaptor Proteins, Vesicular Transport ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Disease Progression ,Unfolded Protein Response ,Unfolded protein response ,biology.protein ,Female ,Carcinogenesis - Abstract
Despite the pivotal role of MYC in tumorigenesis, the mechanisms by which it promotes cancer aggressiveness remain incompletely understood. Here, we show that MYC transcriptionally upregulates the ubiquitin fusion degradation 1 (UFD1) gene in T-cell acute lymphoblastic leukemia (T-ALL). Allelic loss of ufd1 in zebrafish induces tumor cell apoptosis and impairs MYC-driven T-ALL progression but does not affect general health. As the E2 component of an endoplasmic reticulum (ER)-associated degradation (ERAD) complex, UFD1 facilitates the elimination of misfolded/unfolded proteins from the ER. We found that UFD1 inactivation in human T-ALL cells impairs ERAD, exacerbates ER stress, and induces apoptosis. Moreover, we show that UFD1 inactivation promotes the proapoptotic unfolded protein response (UPR) mediated by protein kinase RNA-like ER kinase (PERK). This effect is demonstrated by an upregulation of PERK and its downstream effector C/EBP homologous protein (CHOP), as well as a downregulation of BCL2 and BCLxL. Indeed, CHOP inactivation or BCL2 overexpression is sufficient to rescue tumor cell apoptosis induced by UFD1 knockdown. Together, our studies identify UFD1 as a critical regulator of the ER stress response and a novel contributor to MYC-mediated leukemia aggressiveness, with implications for targeted therapy in T-ALL and likely other MYC-driven cancers.
- Published
- 2018
7. Ultrabright fluorescent cellulose acetate nanoparticles for imaging tumors through systemic and topical applications
- Author
-
Shajesh Palantavida, Maxim Dokukin, Berney Peng, Mohammad Almeqdadi, Hui Feng, Fabrice Laroche, Igor M. Sokolov, Ömer H. Yilmaz, Jatin Roper, Massachusetts Institute of Technology. Department of Biological Engineering, and Koch Institute for Integrative Cancer Research at MIT
- Subjects
Chemistry ,Mechanical Engineering ,Nanoparticle ,02 engineering and technology ,010402 general chemistry ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,01 natural sciences ,Cellulose acetate ,Fluorescence ,Article ,0104 chemical sciences ,Nanomaterials ,chemistry.chemical_compound ,Mechanics of Materials ,In vivo ,Biophysics ,General Materials Science ,Viscose ,Cellulose ,0210 nano-technology ,Preclinical imaging - Abstract
Cellulose acetate (CA), viscose, or artificial silk are biocompatible human-benign derivatives of cellulose, one of the most abundant biopolymers on earth. While various optical materials have been developed from CA, optical CA nanomaterials are nonexistent. Here we report on the assembly of a new family of extremely bright fluorescent CA nanoparticles (CA-dots), which are fully suitable for in vivo imaging/targeting applications. CA-dots can encapsulate a variety of molecular fluorophores. Using various commercially available fluorophores, we demonstrate that the fluorescence of CA-dots can be tuned within the entire UV–VIS-NIR spectrum. We also demonstrate excellent specific targeting of tumors in vivo, when injected in zebrafish (xenograft model of human cervical epithelial cancer), and unusually strong exvivo topical labeling of colon cancer in mice utilizing CA folate-functionalized nanoparticles., National Science Foundation (U.S.) (Grant CBET 1745530)
- Published
- 2019
8. Data on ultrabright fluorescent cellulose acetate nanoparticles for imaging tumors through systemic and topical applications
- Author
-
Ömer H. Yilmaz, Shajesh Palantavida, Berney Peng, Igor M. Sokolov, Hui Feng, Fabrice Laroche, Maxim Dokukin, Jatin Roper, Mohammad Almeqdadi, Massachusetts Institute of Technology. Department of Biology, and Koch Institute for Integrative Cancer Research at MIT
- Subjects
0303 health sciences ,Multidisciplinary ,Materials Science ,Nanoparticle ,Poloxamer ,lcsh:Computer applications to medicine. Medical informatics ,Cellulose acetate ,Fluorescence ,3. Good health ,03 medical and health sciences ,chemistry.chemical_compound ,symbols.namesake ,0302 clinical medicine ,chemistry ,Folic acid ,Copolymer ,symbols ,lcsh:R858-859.7 ,Research article ,Raman spectroscopy ,lcsh:Science (General) ,030217 neurology & neurosurgery ,030304 developmental biology ,Nuclear chemistry ,lcsh:Q1-390 - Abstract
Characterization data of fluorescent nanoparticles made of cellulose acetate (CA-dots) are shown. The data in this article accompanies the research article "Ultrabright fluorescent cellulose acetate nanoparticles for imaging tumors through systemic and topical applications" [1]. The measurements and calculation of brightness of individual CA-dots are presented. The description of conjugation procedure Pluronic F127-Folic Acid copolymer and folic acid is shown. Identification of composition of CA dots using Raman and absorbance spectroscopy is demonstrated. The methods for image analysis of efficiency of CA-dot targeting of epithelial tumors xenografted in zebrafish is presented.
- Published
- 2019
9. The TCA cycle transferase DLST is important for MYC-mediated leukemogenesis
- Author
-
Michelle A. Kelliher, A T Look, M Aioub, H.L. Peng, Y. Samaha, Evisa Gjini, Daniel Helman, Donna Neuberg, T Cheng, Marc R. Mansour, Itrat Harrold, Travis T. Denton, Adam Amsterdam, Dun Li, Takaomi Sanda, Justine E. Roderick, Nicole M. Anderson, Fabrice Laroche, Anurag K. Singh, Hui Feng, and Le Meng
- Subjects
0301 basic medicine ,Cancer Research ,Carcinogenesis ,Cell Survival ,Citric Acid Cycle ,Apoptosis ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,0302 clinical medicine ,RNA interference ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Viability assay ,Zebrafish ,Gene knockdown ,Hematology ,biology.organism_classification ,Cell biology ,Citric acid cycle ,030104 developmental biology ,Oncology ,Biochemistry ,Cell culture ,030220 oncology & carcinogenesis ,Ketoglutaric Acids ,Acyl Coenzyme A ,Acyltransferases - Abstract
Despite the pivotal role of MYC in the pathogenesis of T-cell acute lymphoblastic leukemia (T-ALL) and many other cancers, the mechanisms underlying MYC-mediated tumorigenesis remain inadequately understood. Here we utilized a well-characterized zebrafish model of Myc-induced T-ALL for genetic studies to identify novel genes contributing to disease onset. We found that heterozygous inactivation of a tricarboxylic acid (TCA) cycle enzyme, dihydrolipoamide S-succinyltransferase (Dlst), significantly delayed tumor onset in zebrafish without detectable effects on fish development. DLST is the E2 transferase of the α-ketoglutarate (α-KG) dehydrogenase complex (KGDHC), which converts α-KG to succinyl-CoA in the TCA cycle. RNAi knockdown of DLST led to decreased cell viability and induction of apoptosis in human T-ALL cell lines. Polar metabolomics profiling revealed that the TCA cycle was disrupted by DLST knockdown in human T-ALL cells, as demonstrated by an accumulation of α-KG and a decrease of succinyl-CoA. Addition of succinate, the downstream TCA cycle intermediate, to human T-ALL cells was sufficient to rescue defects in cell viability caused by DLST inactivation. Together, our studies uncovered an important role for DLST in MYC-mediated leukemogenesis and demonstrated the metabolic dependence of T-lymphoblasts on the TCA cycle, thus providing implications for targeted therapy.
- Published
- 2016
10. Functional and genomic analyses reveal therapeutic potential of targeting β-catenin/CBP activity in head and neck cancer
- Author
-
Stefano Monti, Jin-A Lee, Manish V. Bais, Khikmet Sadykov, Khalid Alamoud, Hui Feng, Jennifer E. Snyder-Cappione, Ann Marie Egloff, Maria A. Kukuruzinska, Bach-Cuc Nguyen, Xaralabos Varelas, Vinay K. Kartha, Anna C. Belkina, Fabrice Laroche, and Sara I. Pai
- Subjects
0301 basic medicine ,Cell ,lcsh:Medicine ,HNSCC ,Tumor Status ,Molecular Targeted Therapy ,Neoplasm Metastasis ,Wnt Signaling Pathway ,Zebrafish ,beta Catenin ,Genetics (clinical) ,β-Catenin/CBP transcriptional activity ,ICG-001 ,biology ,Genomics ,3. Good health ,Gene Expression Regulation, Neoplastic ,Phenotype ,medicine.anatomical_structure ,Head and Neck Neoplasms ,Carcinoma, Squamous Cell ,Disease Progression ,Neoplastic Stem Cells ,Molecular Medicine ,Mouth Neoplasms ,lcsh:QH426-470 ,Cell Survival ,Sialoglycoproteins ,Mice, Nude ,Pyrimidinones ,03 medical and health sciences ,Cell Line, Tumor ,Cell Adhesion ,Genetics ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Molecular Biology ,Cell Proliferation ,Cell growth ,Research ,Gene Expression Profiling ,lcsh:R ,Head and neck cancer ,Epithelial Cells ,TCGA ,Bridged Bicyclo Compounds, Heterocyclic ,biology.organism_classification ,medicine.disease ,Survival Analysis ,Head and neck squamous-cell carcinoma ,Peptide Fragments ,Mice, Inbred C57BL ,lcsh:Genetics ,stomatognathic diseases ,030104 developmental biology ,Tumor progression ,Catenin ,Cancer research ,Aggressive tumor cells - Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is an aggressive malignancy characterized by tumor heterogeneity, locoregional metastases, and resistance to existing treatments. Although a number of genomic and molecular alterations associated with HNSCC have been identified, they have had limited impact on the clinical management of this disease. To date, few targeted therapies are available for HNSCC, and only a small fraction of patients have benefited from these treatments. A frequent feature of HNSCC is the inappropriate activation of β-catenin that has been implicated in cell survival and in the maintenance and expansion of stem cell-like populations, thought to be the underlying cause of tumor recurrence and resistance to treatment. However, the therapeutic value of targeting β-catenin activity in HNSCC has not been explored. Methods We utilized a combination of computational and experimental profiling approaches to examine the effects of blocking the interaction between β-catenin and cAMP-responsive element binding (CREB)-binding protein (CBP) using the small molecule inhibitor ICG-001. We generated and annotated in vitro treatment gene expression signatures of HNSCC cells, derived from human oral squamous cell carcinomas (OSCCs), using microarrays. We validated the anti-tumorigenic activity of ICG-001 in vivo using SCC-derived tumor xenografts in murine models, as well as embryonic zebrafish-based screens of sorted stem cell-like subpopulations. Additionally, ICG-001-inhibition signatures were overlaid with RNA-sequencing data from The Cancer Genome Atlas (TCGA) for human OSCCs to evaluate its association with tumor progression and prognosis. Results ICG-001 inhibited HNSCC cell proliferation and tumor growth in cellular and murine models, respectively, while promoting intercellular adhesion and loss of invasive phenotypes. Furthermore, ICG-001 preferentially targeted the ability of subpopulations of stem-like cells to establish metastatic tumors in zebrafish. Significantly, interrogation of the ICG-001 inhibition-associated gene expression signature in the TCGA OSCC human cohort indicated that the targeted β-catenin/CBP transcriptional activity tracked with tumor status, advanced tumor grade, and poor overall patient survival. Conclusions Collectively, our results identify β-catenin/CBP interaction as a novel target for anti-HNSCC therapy and provide evidence that derivatives of ICG-001 with enhanced inhibitory activity may serve as an effective strategy to interfere with aggressive features of HNSCC. Electronic supplementary material The online version of this article (10.1186/s13073-018-0569-7) contains supplementary material, which is available to authorized users.
- Published
- 2018
11. Towards Resolving the Pro- and Anti-Tumor Effects of the Aryl Hydrocarbon Receptor
- Author
-
Olga Novikov, Supraja Narasimhan, Elizabeth Stanford Zulick, Jennifer J. Schlezinger, Hui Feng, Francesca Mulas, Stefano Monti, Zhongyan Wang, Ashley J. Parks, David H. Sherr, and Fabrice Laroche
- Subjects
0301 basic medicine ,Cell ,Metastasis ,CDH1 ,lcsh:Chemistry ,Small hairpin RNA ,Cell Movement ,Neoplasms ,lcsh:QH301-705.5 ,Spectroscopy ,Zebrafish ,Gene knockdown ,biology ,Chemistry ,aryl hydrocarbon receptor ,General Medicine ,respiratory system ,3. Good health ,Computer Science Applications ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,AHR agonist ,Female ,Breast Neoplasms ,Catalysis ,Article ,Inorganic Chemistry ,AHR antagonist ,03 medical and health sciences ,cancer therapeutics ,Cell Line, Tumor ,medicine ,cancer ,Animals ,Humans ,Neoplasm Invasiveness ,Gene Silencing ,Physical and Theoretical Chemistry ,Molecular Biology ,Matrigel ,Organic Chemistry ,Cancer ,Aryl hydrocarbon receptor ,medicine.disease ,respiratory tract diseases ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Receptors, Aryl Hydrocarbon ,Cancer research ,biology.protein - Abstract
We have postulated that the aryl hydrocarbon receptor (AHR) drives the later, more lethal stages of some cancers when chronically activated by endogenous ligands. However, other studies have suggested that, under some circumstances, the AHR can oppose tumor aggression. Resolving this apparent contradiction is critical to the design of AHR-targeted cancer therapeutics. Molecular (siRNA, shRNA, AHR repressor, CRISPR-Cas9) and pharmacological (AHR inhibitors) approaches were used to confirm the hypothesis that AHR inhibition reduces human cancer cell invasion (irregular colony growth in 3D Matrigel cultures and Boyden chambers), migration (scratch wound assay) and metastasis (human cancer cell xenografts in zebrafish). Furthermore, these assays were used for a head-to-head comparison between AHR antagonists and agonists. AHR inhibition or knockdown/knockout consistently reduced human ER−/PR−/Her2− and inflammatory breast cancer cell invasion, migration, and metastasis. This was associated with a decrease in invasion-associated genes (e.g., Fibronectin, VCAM1, Thrombospondin, MMP1) and an increase in CDH1/E-cadherin, previously associated with decreased tumor aggression. Paradoxically, AHR agonists (2,3,7,8-tetrachlorodibenzo-p-dioxin and/or 3,3′-diindolylmethane) similarly inhibited irregular colony formation in Matrigel and blocked metastasis in vivo but accelerated migration. These data demonstrate the complexity of modulating AHR activity in cancer while suggesting that AHR inhibitors, and, under some circumstances, AHR agonists, may be useful as cancer therapeutics.
- Published
- 2018
12. A Novel Capturing Method for Quantification of Extra-Cellular Nanovesicles
- Author
-
Qu Yin, Jianning Ding, Fabrice Laroche, Run Wang, Xiang Zhou, Bencai Lin, Zunfeng Liu, Ningyi Yuan, Jan Pieter Abrahams, and Ningning Shao
- Subjects
Materials science ,Biomedical Engineering ,Bioengineering ,Nanotechnology ,02 engineering and technology ,Microscopy, Atomic Force ,Extracellular vesicles ,03 medical and health sciences ,Extracellular Vesicles ,0302 clinical medicine ,Microscopy, Electron, Transmission ,Humans ,General Materials Science ,Sample preparation ,Particle Size ,Reproducibility ,Atomic force microscopy ,Reproducibility of Results ,030206 dentistry ,General Chemistry ,Extracellular vesicle ,021001 nanoscience & nanotechnology ,Condensed Matter Physics ,HEK293 Cells ,Potential biomarkers ,Nanoparticles ,Nanometre ,0210 nano-technology ,Inorganic nanoparticles - Abstract
Extracellular vesicles (EVs), secreted by cells and found in body fluids play important roles in intercellular communication. Therefore, EVs are receiving increasing attention as potential biomarkers in the diagnosis and prognosis of various diseases. However, the detection and the quantification of EVs are hampered by the nanometer scale of these particles and by the lack of optimized quantification methods. Atomic force microscopy (AFM) is a powerful technology that can detect small particles. Here we report a 3D capture method for sample preparation of AFM which improves the accuracy, sensitivity and reproducibility for EVs’ detection, compared to conventional sample preparation methods. By shaking a mica plate in EV solution, all the EVs were captured onto the 2D surface. The majority of the captured particles have a size ranging from 10 to 120 nm, which correlates with size data obtained from transmission electron microscopy studies. This novel sample preparation method has high adaptability potential and can also be applied to other organic and inorganic nanoparticles.
- Published
- 2018
13. Zebrafish Models of Human Leukemia: Technological Advances and Mechanistic Insights
- Author
-
Nicholas R. Harrison, Alejandro Gutierrez, Fabrice Laroche, and Hui Feng
- Subjects
0301 basic medicine ,Human leukemia ,Leukemia ,animal structures ,Disease mechanisms ,Danio ,Computational biology ,Biology ,biology.organism_classification ,medicine.disease ,Article ,Hematopoiesis ,Transplantation ,Disease Models, Animal ,03 medical and health sciences ,030104 developmental biology ,Genome editing ,medicine ,Animals ,Humans ,Treatment resistance ,Zebrafish - Abstract
Insights concerning leukemic pathophysiology have been acquired in various animal models and further efforts to understand the mechanisms underlying leukemic treatment resistance and disease relapse promise to improve therapeutic strategies. The zebrafish (Danio rerio) is a vertebrate organism with a conserved hematopoietic program and unique experimental strengths suiting it for the investigation of human leukemia. Recent technological advances in zebrafish research including efficient transgenesis, precise genome editing, and straightforward transplantation techniques have led to the generation of a number of leukemia models. The transparency of the zebrafish when coupled with improved lineage-tracing and imaging techniques has revealed exquisite details of leukemic initiation, progression, and regression. With these advantages, the zebrafish represents a unique experimental system for leukemic research and additionally, advances in zebrafish-based high-throughput drug screening promise to hasten the discovery of novel leukemia therapeutics. To date, investigators have accumulated knowledge of the genetic underpinnings critical to leukemic transformation and treatment resistance and without doubt, zebrafish are rapidly expanding our understanding of disease mechanisms and helping to shape therapeutic strategies for improved outcomes in leukemic patients.
- Published
- 2016
14. Identification of an OPR protein involved in the translation initiation of the PsaB subunit of photosystem I
- Author
-
Jean-David Rochaix, Fabrice Laroche, Michèle Rahire, and Lorenzo Cerutti
- Subjects
0106 biological sciences ,0303 health sciences ,biology ,Protein subunit ,Chlamydomonas ,Mutant ,Translation (biology) ,Cell Biology ,Plant Science ,biology.organism_classification ,Photosystem I ,01 natural sciences ,Molecular biology ,03 medical and health sciences ,Eukaryotic translation ,Biochemistry ,Genetics ,Translation factor ,Peptide sequence ,030304 developmental biology ,010606 plant biology & botany - Abstract
Genetic analysis of mutants deficient in the biosynthesis of the photosystem I complex has revealed several nucleus-encoded factors that act at different post-transcriptional steps of chloroplast gene expression. Here we have identified and characterized the gene affected in the tab 1-F15 mutant, which is specifically deficient in the translation of the photosystem I reaction center protein PsaB as the result of a single nucleotide deletion. This gene encodes Tab 1, a 1287 amino acid protein that contains 10 tandem 38-40 amino acid degenerate repeats of the PPPEW/OPR (octatricopeptide repeat) family, first described for the chloroplast translation factor Tbc2. These repeats are involved in the binding of Tab 1 to the 5'-untranslated region of the psaB mRNA based on gel mobility shift assays. Tab 1 is part of a large family of proteins in Chlamydomonas that are also found in several bacteria and protozoans, but are rare in land plants.
- Published
- 2012
15. Crystal structure of the TLDc domain of oxidation resistance protein 2 from zebrafish
- Author
-
Søren Roi Midtgaard, Herman P. Spaink, Jaslyn E. M. M. Wong, Lotte Schack, Fabrice Laroche, Søren Thirup, Husam M. A. B. Alsarraf, Jens Stougaard, and Mickaël Blaise
- Subjects
chemistry.chemical_classification ,0303 health sciences ,biology ,Structural similarity ,Sequence alignment ,Crystal structure ,biology.organism_classification ,Antiparallel (biochemistry) ,Biochemistry ,Amino acid ,03 medical and health sciences ,Crystallography ,0302 clinical medicine ,Protein structure ,chemistry ,Structural Biology ,Molecular Biology ,Zebrafish ,Peptide sequence ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
The oxidation resistance proteins (OXR) help to protect eukaryotes from reactive oxygen species. The sole C-terminal domain of the OXR, named TLDc is sufficient to perform this function. However, the mechanism by which oxidation resistance occurs is poorly understood. We present here the crystal structure of the TLDc domain of the oxidation resistance protein 2 from zebrafish. The structure was determined by X-ray crystallography to atomic resolution (0.97A) and adopts an overall globular shape. Two antiparallel β-sheets form a central β-sandwich, surrounded by two helices and two one-turn helices. The fold shares low structural similarity to known structures.
- Published
- 2012
16. A Mutant Impaired in SNARE Complex Dissociation Identifies the Plasma Membrane as First Target of Synaptobrevin 2
- Author
-
Sonia Martinez-Arca, Rachel Rudge, Thierry Galli, Fabrice Laroche, and Stefan T. Arold
- Subjects
Synaptobrevin ,Vesicle ,SNAP25 ,Lipid bilayer fusion ,Cell Biology ,Biology ,Biochemistry ,Exocytosis ,Cell biology ,Membrane ,Structural Biology ,Genetics ,SNARE complex ,Molecular Biology ,Dynamin - Abstract
Membrane fusion depends on the formation of a complex of four SNARE motifs, three that bear a central glutamine and are localized in the target membrane (t-SNARE) and one that bears an arginine and is localized in the donor vesicle (v-SNARE). We have characterized the arginine 56 to proline mutant (R56P) of synaptobrevin-2 (Sb). SbR56P was blocked at the plasma membrane in association with the endogenous plasma membrane t-SNARE due to an inhibition of SNARE complex dissociation, suggesting that the plasma membrane is its first target. Cell surface blockade of SbR56P could be rescued by coexpression of synaptophysin, a partner of Sb. Sb was blocked at the plasma membrane but SNARE complexes were unaffected in cells expressing defective dynamin, indicating that the phenotype of SbR56P was not due to an internalization defect. When expressed in neurons, SbR56P localized both to axonal and dendritic plasma membranes, showing that both domains are initial targets of Sb. The R56P mutation affects a highly conserved position in v-SNAREs, and might thus provide a general tool for identifying their first target membranes.
- Published
- 2004
17. Abstract 1180: The TCA cycle transferase DLST is critical for MYC-mediated leukemogenesis
- Author
-
Le Meng, Marc R. Mansour, Daniel Helman, Travis T. Denton, A. Thomas Look, H.L. Peng, Hui Feng, Fabrice Laroche, Dun Li, Anurag K. Singh, Donna Neuberg, Evisa Gjini, Itrat Harrold, Takaomi Sanda, Nicole M. Anderson, and Adam Amsterdam
- Subjects
Cancer Research ,Gene knockdown ,Oncogene ,Biology ,medicine.disease ,medicine.disease_cause ,Citric acid cycle ,Leukemia ,Oncology ,Apoptosis ,RNA interference ,Cell culture ,medicine ,Cancer research ,Carcinogenesis - Abstract
The proto-oncogene MYC has been implicated in the pathogenesis of many human cancers, including hematological and solid malignancies1. In the majority of T-ALL cases, MYC is aberrantly expressed downstream of activated NOTCH1 mutations. Despite the pivotal role of MYC in the pathogenesis of T-cell acute lymphoblastic leukemia (T-ALL) and many other cancers, the mechanisms underlying MYC-mediated tumorigenesis remain inadequately understood2. Here we utilized a well-characterized zebrafish model of Myc-induced T-ALL for genetic studies to identify novel genes contributing to disease onset. We found that heterozygous inactivation of a tricarboxylic acid (TCA) cycle enzyme, dihydrolipoamide S-succinyltransferase (Dlst), significantly delayed tumor onset. Concordant with our zebrafish results, RNAi knockdown of DLST led to decreased cell viability and induction of apoptosis in human T-ALL cell lines. DLST is the E2 transferase of the -ketoglutarate dehydrogenase complex (KGDHC), which converts -ketoglutarate (-KG) to succinyl-CoA in the TCA cycle3. Polar metabolomics profiling revealed that the TCA cycle was disrupted by DLST knockdown in human T-ALL cells, as demonstrated by an accumulation of -KG and its precursor glutamine, as well as a loss of succinyl-CoA. Addition of succinate, the downstream TCA cycle intermediate, to human T-ALL cells was sufficient to rescue defects in cell viability caused by DLST inactivation. Taken together, our studies identify DLST as an important mediator of MYC-driven leukemogenesis and provide compelling evidence for the metabolic dependence of T-ALL cells on the TCA cycle. 1. Nesbit CE, Tersak JM, Prochownik EV. MYC oncogenes and human neoplastic disease. Oncogene 1999 May 13; 18(19): 3004-3016. 2. Sharma VM, Calvo JA, Draheim KM, Cunningham LA, Hermance N, Beverly L, et al. Notch1 contributes to mouse T-cell leukemia by directly inducing the expression of c-myc. Molecular and cellular biology 2006 Nov; 26(21): 8022-8031. 3. Sheu KF, Blass JP. The alpha-ketoglutarate dehydrogenase complex. Ann N Y Acad Sci 1999; 893: 61-78. Citation Format: Nicole Anderson, Dun Li, H.L. Peng, Marc Mansour, Fabrice Laroche, Evisa Gjini, Daniel Helman, Itrat Harrold, Le Meng, Takaomi Sanda, Adam Amsterdam, Donna Neuberg, Travis Denton, Anurag Singh, A Thomas Look, Hui Feng. The TCA cycle transferase DLST is critical for MYC-mediated leukemogenesis. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1180.
- Published
- 2016
18. Ultra-Small Graphene Oxide Functionalized with Polyethylenimine (PEI) for Very Efficient Gene Delivery in Cell and Zebrafish Embryos
- Author
-
Gerda E. M. Lamers, Xiang Zhou, Vincenzo Torraca, Zunfeng Liu, Fuqiang Chu, Tao Lu, Fabrice Laroche, Patrick Voskamp, Herman P. Spaink, and Jan Pieter Abrahams
- Subjects
Materials science ,animal structures ,viruses ,Gene delivery ,law.invention ,chemistry.chemical_compound ,law ,Gene expression ,General Materials Science ,Electrical and Electronic Engineering ,Cytotoxicity ,Zebrafish ,Polyethylenimine ,biology ,Graphene ,fungi ,technology, industry, and agriculture ,Transfection ,Condensed Matter Physics ,biology.organism_classification ,Molecular biology ,Atomic and Molecular Physics, and Optics ,Cell biology ,chemistry ,embryonic structures ,DNA - Abstract
Efficient DNA delivery is essential for introducing new genes into living cells. However, effective virus-based systems carry risks and efficient synthetic systems that are non-toxic remain to be discovered. The bottle-neck in synthetic systems is cytotoxicity, caused by the high concentration of DNA-condensing compounds required for efficient uptake of DNA. Here we report a polyethyleneimine (PEI) grafted ultra-small graphene oxide (PEI-g-USGO) for transfection. By removing the free PEI and ensuring a high PEI density on small sized graphene, we obtained very high transfection efficiencies combined with very low cytotoxicity. Plasmid DNA could be transfected into mammalian cell lines with up to 95% efficiency and 90% viability. Transfection in zebrafish embryos was 90%, with high viability, compared to efficiencies of 30% or lower for established transfection technologies. This result suggests a novel approach to the design of synthetic gene delivery vehicles for research and therapy.
- Published
- 2012
19. Crystal structure of the TLDc domain of oxidation resistance protein 2 from zebrafish
- Author
-
Mickaël, Blaise, Husam M A B, Alsarraf, Jaslyn E M M, Wong, Søren Roi, Midtgaard, Fabrice, Laroche, Lotte, Schack, Herman, Spaink, Jens, Stougaard, and Søren, Thirup
- Subjects
Models, Molecular ,Sequence Homology, Amino Acid ,Molecular Sequence Data ,Nuclear Receptor Coactivators ,Proteins ,Zebrafish Proteins ,Crystallography, X-Ray ,Protein Structure, Tertiary ,Mitochondrial Proteins ,Mice ,Animals ,Humans ,Amino Acid Sequence ,Cysteine ,Oxidation-Reduction ,Sequence Alignment ,Zebrafish - Abstract
The oxidation resistance proteins (OXR) help to protect eukaryotes from reactive oxygen species. The sole C-terminal domain of the OXR, named TLDc is sufficient to perform this function. However, the mechanism by which oxidation resistance occurs is poorly understood. We present here the crystal structure of the TLDc domain of the oxidation resistance protein 2 from zebrafish. The structure was determined by X-ray crystallography to atomic resolution (0.97Å) and adopts an overall globular shape. Two antiparallel β-sheets form a central β-sandwich, surrounded by two helices and two one-turn helices. The fold shares low structural similarity to known structures.
- Published
- 2011
20. Purification, crystallization and preliminary crystallographic studies of the TLDc domain of oxidation resistance protein 2 from zebrafish
- Author
-
Mickaël Blaise, Herman P. Spaink, Fabrice Laroche, Søren Thirup, Husam M. A. B. Alsarraf, Institut Jacques Monod (IJM (UMR_7592)), Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), and Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)
- Subjects
Models, Molecular ,animal structures ,DNA damage ,[SDV]Life Sciences [q-bio] ,Biophysics ,medicine.disease_cause ,Crystallography, X-Ray ,Biochemistry ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,Structural Biology ,law ,Genetics ,medicine ,Animals ,Crystallization ,Zebrafish ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,Reactive oxygen species ,biology ,030302 biochemistry & molecular biology ,fungi ,Zebrafish Proteins ,Condensed Matter Physics ,biology.organism_classification ,Protein Structure, Tertiary ,Crystallography ,Enzyme ,chemistry ,Catalase ,Crystallization Communications ,embryonic structures ,biology.protein ,Carrier Proteins ,DNA ,Oxidative stress - Abstract
Cell metabolic processes are constantly producing reactive oxygen species (ROS), which have deleterious effects by triggering, for example, DNA damage. Numerous enzymes such as catalase, and small compounds such as vitamin C, provide protection against ROS. The TLDc domain of the human oxidation resistance protein has been shown to be able to protect DNA from oxidative stress; however, its mechanism of action is still not understood and no structural information is available on this domain. Structural information on the TLDc domain may therefore help in understanding exactly how it works. Here, the purification, crystallization and preliminary crystallographic studies of the TLDc domain from zebrafish are reported. Crystals belonging to the orthorhombic space group P2(1)2(1)2 were obtained and diffracted to 0.97 Å resolution. Selenomethionine-substituted protein could also be crystallized; these crystals diffracted to 1.1 Å resolution and the structure could be solved by SAD/MAD methods.
- Published
- 2011
21. Polymorphisms of coding trinucleotide repeats of homeogenes in neurodevelopmental psychiatric disorders
- Author
-
Marie-Christine Mouren-Simeoni, Celia Fortin, Michel Simonneau, Thierry Galli, Agnès Mogenet, Sophie Leroy, Fabrice Laroche, Nicolas Ramoz, Laurence Colleaux, Anne Philippe, Philip Gorwood, Bérangère Rousselot-Paillet, Marie-Odile Krebs, Bernard Golse, Laurence Robel, Jean-Louis Bresson, Institut Jacques Monod (IJM (UMR_7592)), Université Paris Diderot - Paris 7 (UPD7)-Centre National de la Recherche Scientifique (CNRS), Institut de psychiatrie et neurosciences de Paris (IPNP - U1266 Inserm - Paris Descartes), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Physiopathologie des Maladies Psychiatriques, Développement et Vulnérabilité, Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), CHU Necker - Enfants Malades [AP-HP], Troubles du comportement alimentaire de l'adolescent (UMR_S 669), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Sud - Paris 11 (UP11), Génétique et épigénétique des maladies métaboliques, neurosensorielles et du développement (Inserm U781), Imagine - Institut des maladies génétiques (IMAGINE - U1163), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), CIC - Biotherapie - GHU Ouest APHP (CIC-BT 502), Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Necker - Enfants Malades [AP-HP], Service de psychopathologie de l'enfant et de l'adolescent, Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Hôpital Robert Debré-Université Paris Diderot - Paris 7 (UPD7), Analyse Phenotypique, Developpementale et Genetique des Comportements Addictifs, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris Diderot - Paris 7 (UPD7), Service de psychiatrie, Assistance publique - Hôpitaux de Paris (AP-HP) (APHP)-Université Paris Descartes - Paris 5 (UPD5)-Hôpital Sainte-Anne, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Necker - Enfants Malades [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Robert Debré-Université Paris Diderot - Paris 7 (UPD7), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris Descartes - Paris 5 (UPD5)-Hôpital Sainte-Anne, Trafic Membranaire et Morphogenèse Neuronale & Epithéliale, Institut National de la Santé et de la Recherche Médicale (INSERM), Fondation de France, Fondation pour la recherche sur le cerveau, Université Paris Diderot - Paris 7 (UPD7)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Jacques Monod ( IJM ), Université Paris Diderot - Paris 7 ( UPD7 ) -Centre National de la Recherche Scientifique ( CNRS ), Université Paris Diderot - Paris 7 ( UPD7 ) -Centre National de la Recherche Scientifique ( CNRS ) -Université Paris Diderot - Paris 7 ( UPD7 ) -Centre National de la Recherche Scientifique ( CNRS ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ), Université Paris Diderot - Paris 7 ( UPD7 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ), Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -Centre National de la Recherche Scientifique ( CNRS ), Troubles du comportement alimentaire de l'adolescent ( UMR_S 669 ), Université Paris-Sud - Paris 11 ( UP11 ) -Université Paris Descartes - Paris 5 ( UPD5 ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ), and Assistance publique - Hôpitaux de Paris (AP-HP)-Hôpital Robert Debré-Université Paris Diderot - Paris 7 ( UPD7 )
- Subjects
Male ,Candidate gene ,MESH : Polymorphism, Genetic ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Coding (therapy) ,MESH : Genes, Homeobox ,0302 clinical medicine ,Trinucleotide Repeats ,MESH : Female ,Genetics (clinical) ,Genetics ,0303 health sciences ,Mental Disorders ,Genes, Homeobox ,FOXP2 ,MESH : Trinucleotide Repeats ,3. Good health ,Psychiatry and Mental health ,Schizophrenia ,[ SDV.NEU.NB ] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Female ,medicine.medical_specialty ,MESH: Trinucleotide Repeats ,Brain development ,MESH : Male ,engrailed genes ,autism ,Biology ,mental retardation ,03 medical and health sciences ,HOXA1 ,MESH: Polymorphism, Genetic ,mental disorders ,medicine ,Humans ,MESH: Mental Disorders ,DLX2 ,MESH : Mental Disorders ,Psychiatry ,Transcription factor ,Gene ,Biological Psychiatry ,030304 developmental biology ,Polymorphism, Genetic ,MESH: Humans ,MESH : Humans ,MESH: Genes, Homeobox ,medicine.disease ,MESH: Male ,schizophrenia ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Autism ,MESH: Female ,030217 neurology & neurosurgery - Abstract
International audience; OBJECTIVES: Autism (MIM#209850) and schizophrenia (MIM#181500) are both neurodevelopmental psychiatric disorders characterized by a highly genetic component. Homeogenes and forkhead genes encode transcription factors, which have been involved in brain development and cell differentiation. Thus, they are relevant candidate genes for psychiatric disorders. Genetic studies have reported an association between autism and DLX2, HOXA1, EN2, ARX, and FOXP2 genes whereas only three studies of EN2, OTX2, and FOXP2 were performed on schizophrenia. Interestingly, most of these candidate genes contain trinucleotide repeats coding for polyamino acid stretch in which instability can be the cause of neurodevelopmental disorders. Our goal was to identify variations of coding trinucleotide repeats in schizophrenia, autism, and idiopathic mental retardation. METHODS: We screened the coding trinucleotide repeats of OTX1, EN1, DLX2, HOXA1, and FOXP2 genes in populations suffering from schizophrenia (247 patients), autism (98 patients), and idiopathic mental retardation (56 patients), and compared them with control populations (112 super controls and 202 healthy controls). RESULTS: Novel deletions and insertions of coding trinucleotide repeats were found in the DLX2, HOXA1, and FOXP2 genes. Most of these variations were detected in controls and no difference in their distribution was observed between patient and control groups. Two different polymorphisms in FOXP2 were, however, found only in autistic patients and the functional consequences of these variations of repeats have to be characterized and correlated to particular clinical features. CONCLUSION: This study did not identify specific disease risk variants of trinucleotide repeats in OTX1, EN1, DLX2, HOXA1, and FOXP2 candidate genes in neurodevelopmental psychiatric disorders.
- Published
- 2008
22. Visualizing the localization of transfection complexes during graphene nanoparticle-based transfection
- Author
-
Yue Zhang, Guoqiang Song, Jan Pieter Abrahams, Shougen Yin, Filip ten Bruggencate, Zunfeng Liu, and Fabrice Laroche
- Subjects
Polyethylenimine ,Materials science ,Graphene ,Confocal ,fungi ,Biomedical Engineering ,Nanoparticle ,Nanotechnology ,General Chemistry ,General Medicine ,Transfection ,Nanomaterials ,law.invention ,chemistry.chemical_compound ,chemistry ,law ,Fluorescence microscope ,General Materials Science ,DNA - Abstract
Recent research has shown that ultra-small graphene oxide (USGO) particles functionalized with polyethylenimine (PEI) can be used to transfect human cell lines with very high efficiency and low toxicity. In this study, confocal fluorescence microscopy is used to provide more insight into the nature of the complexes formed by DNA and USGO_PEI and to track their localization during the transfection process. The results indicate that the DNA enters the nucleus in complex with the USGO_PEI, which remains inside the nucleus. Also, it is observed that USGO_PEI and plasmid DNA form large aggregates in the transfection medium. Both observations raise concerns that this type of nanomaterial might need further improvements for therapeutic use.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.