Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Proc Natl Acad Sci U S A ; 114(48): E10281-E10290, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133392

ABSTRACT

Cavitation-facilitated microbubble-mediated focused ultrasound therapy is a promising method of drug delivery across the blood-brain barrier (BBB) for treating many neurological disorders. Unlike ultrasound thermal therapies, during which magnetic resonance thermometry can serve as a reliable treatment control modality, real-time control of modulated BBB disruption with undetectable vascular damage remains a challenge. Here a closed-loop cavitation controlling paradigm that sustains stable cavitation while suppressing inertial cavitation behavior was designed and validated using a dual-transducer system operating at the clinically relevant ultrasound frequency of 274.3 kHz. Tests in the normal brain and in the F98 glioma model in vivo demonstrated that this controller enables reliable and damage-free delivery of a predetermined amount of the chemotherapeutic drug (liposomal doxorubicin) into the brain. The maximum concentration level of delivered doxorubicin exceeded levels previously shown (using uncontrolled sonication) to induce tumor regression and improve survival in rat glioma. These results confirmed the ability of the controller to modulate the drug delivery dosage within a therapeutically effective range, while improving safety control. It can be readily implemented clinically and potentially applied to other cavitation-enhanced ultrasound therapies.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Blood-Brain Barrier/metabolism , Brain Neoplasms/therapy , Doxorubicin/analogs & derivatives , Drug Delivery Systems/methods , Glioma/therapy , Ultrasonic Therapy/methods , Acoustics/instrumentation , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacokinetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carbocyanines/chemistry , Carbocyanines/pharmacokinetics , Corpus Striatum/diagnostic imaging , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Drug Delivery Systems/instrumentation , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Glioma/diagnostic imaging , Glioma/metabolism , Glioma/pathology , Hippocampus/diagnostic imaging , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Luminescent Proteins/chemistry , Luminescent Proteins/pharmacokinetics , Magnetic Resonance Imaging , Male , Microbubbles , Molecular Targeted Therapy , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology , Rats , Rats, Sprague-Dawley , Transducers , Ultrasonic Waves
2.
Bioconjug Chem ; 30(2): 400-404, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30616339

ABSTRACT

The delivery of entire functional proteins into living cells is a long-sought goal in science. Cyclic cell-penetrating peptides (cCPPs) have proven themselves to be potent delivery vehicles to carry proteins upon conjugation into the cytosol of living cells with immediate bioavailability via a non-endosomal uptake pathway. With this strategy, we pursue the cytosolic delivery of mCherry, a medium-sized fluorescent protein. Afterward, we achieve subcellular delivery of mCherry to different intracellular loci by genetic fusion of targeting peptides to the protein sequence. We show efficient transport into a membrane-bound compartment, the nucleus, as well as targeting of the actin cytoskeleton, marking one of the first ways to label actin fluorescently in genetically unmodified living cells. Furthermore, we demonstrate that only by conjugation of cCPPs via a disulfide bond, is flawless localization to the target area achieved. This finding underlines the importance of using a cCPP-based delivery vehicle that is cleaved inside cells, for the precise intracellular localization of a protein of interest.


Subject(s)
Cell-Penetrating Peptides/metabolism , Drug Carriers/metabolism , Luminescent Proteins/administration & dosage , Peptides, Cyclic/metabolism , Cell-Penetrating Peptides/chemistry , Cytosol/metabolism , Drug Carriers/chemistry , Drug Delivery Systems , HeLa Cells , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/pharmacokinetics , Peptides, Cyclic/chemistry , Red Fluorescent Protein
3.
Bioconjug Chem ; 30(2): 432-442, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30615416

ABSTRACT

Proteins are ideal candidates for disease treatment because of their high specificity and potency. Despite this potential, delivery of proteins remains a significant challenge due to the intrinsic size, charge, and stability of proteins. Attempts to overcome these challenges have most commonly relied on direct conjugation of polymers and peptides to proteins via reactive groups on naturally occurring residues. While such approaches have shown some success, they allow limited control of the spacing and number of moieties coupled to proteins, which can hinder bioactivity and delivery capabilities of the therapeutic. Here, we describe a strategy to site-specifically conjugate delivery moieties to therapeutic proteins through unnatural amino acid (UAA) incorporation, in order to explore the effect of epidermal growth factor receptor (EGFR)-targeted ligand valency and spacing on internalization of proteins in EGFR-overexpressing inflammatory breast cancer (IBC) cells. Our results demonstrate the ability to enhance targeted protein delivery by tuning a small number of EGFR ligands per protein and clustering these ligands to promote multivalent ligand-receptor interactions. Furthermore, the tailorability of this simple approach was demonstrated through IBC-targeted cell death via the delivery of yeast cytosine deaminase (yCD), a prodrug converting enzyme.


Subject(s)
Amino Acids/metabolism , Cytosine Deaminase/administration & dosage , Luminescent Proteins/administration & dosage , Amino Acids/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Click Chemistry , Cytosine Deaminase/chemistry , Cytosine Deaminase/pharmacokinetics , Drug Delivery Systems , ErbB Receptors/metabolism , Female , Humans , Ligands , Luminescent Proteins/chemistry , Luminescent Proteins/pharmacokinetics , Models, Molecular , Protein Binding , Yeasts/enzymology , Red Fluorescent Protein
4.
Acta Radiol ; 60(3): 315-326, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29890843

ABSTRACT

BACKGROUND: Mouse models of human-malignant-melanoma (MM) are important tools to study tumor dynamics. The enhanced green fluorescent protein (EGFP) is widely used in molecular imaging approaches, together with optical scanners, and fluorescence imaging. PURPOSE: Currently, there are no data available as to whether other fluorescent proteins are more suitable. The goal of this preclinical study was to analyze two fluorescent proteins of the GFP superfamily under real-time in vivo conditions using fluorescence reflectance imaging (FRI). MATERIAL AND METHODS: The human melanoma cell line MeWo was stable transfected with one plasmid: pEGFP-C1 or pDsRed1-N1. We investigated two severe combined immunodeficiency (SCID)-mice groups: A (solid xenografts) and B (xenografts as metastases). After three weeks, the animals were weekly imaged by FRI. Afterwards the mice were euthanized and metastases were imaged in situ: to quantify the cutis-dependent reduction of emitted light, we compared signal intensities obtained by metastases in vivo with signal intensities obtained by in situ liver parenchyma preparations. RESULTS: More than 90% of cells were stable transfected. EGFP-/DsRed-xenograft tumors had identical growth kinetics. In vivo the emitted light by DsRed tumors/metastases was much brighter than by EGFP. DsRed metastases were earlier (3 vs. 5 weeks) and much more sensitive detectable than EGFP metastases. Cutis-dependent reduction of emitted light was greater in EGFP than in DsRed mice (tenfold). Autofluorescence of DsRed was lower than of EGFP. CONCLUSION: We established an in vivo xenograft mouse model (DsRed-MeWo) that is reliable, reproducible, and superior to the EGFP model as a preclinical tool to study innovative therapies by FRI under real-time in vivo conditions.


Subject(s)
Green Fluorescent Proteins/pharmacokinetics , Melanoma/diagnostic imaging , Animals , Cell Line, Tumor , Disease Models, Animal , Heterografts , Humans , Luminescent Proteins/pharmacokinetics , Male , Mice , Mice, SCID , Microscopy, Fluorescence , Random Allocation , Transfection , Tumor Burden
5.
Biochem Biophys Res Commun ; 480(4): 534-538, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27769857

ABSTRACT

Insulin-stimulated GLUT4 translocation from GLUT4 storage vesicles (GSVs) to the plasma membrane (PM) constitutes a key process for blood glucose control. Therefore, compounds that could promote GLUT4 translocation into the PM represent potential drugs for the treatment of diabetes. In this research, we screened for agonists that induce GLUT4 translocation by using a novel pH-sensitive fluorescent probe, insulin-regulated aminopeptidase (IRAP)-mOrange2. We identified as well as validated one agonist, staurosporine, from a 64,000 compound library. Staurosporine promotes GSVs translocation into the PM and increases glucose uptake through the AMP-activated protein kinase (AMPK) pathway, serving as an effective insulin additive analogue in L6 cells. Our work highlights the convenience and efficiency of this novel pH-sensitive fluorescent probe and reveals the new biological activity of staurosporine as an agonist for GLUT4 translocation and as an effective insulin additive analogue.


Subject(s)
Cell Membrane/metabolism , Glucose Transporter Type 4/metabolism , Glucose/pharmacokinetics , Interleukin 1 Receptor Antagonist Protein/pharmacokinetics , Luminescent Proteins/pharmacokinetics , Staurosporine/pharmacology , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Fluorescent Dyes/pharmacokinetics , Hydrogen-Ion Concentration , Insulin/administration & dosage , Insulin/analogs & derivatives , Microscopy, Fluorescence , Protein Transport/drug effects , Staurosporine/analysis
6.
Zh Evol Biokhim Fiziol ; 52(2): 221-225, 2016 03.
Article in Russian | MEDLINE | ID: mdl-30695502

ABSTRACT

Protein reabsorption in the proximal tubules (PT) of the frog kidney was studied by the methods of immunohistochemistry, fluorescent and confocal microscopy. Yellow fluorescent protein (YFP) was introduced in combination with other proteins. Reabsorption of YFP introduced simultaneously with ly- sozyme or green fluorescent protein (GFP) did not differ from the result of YFP injection only. Previous lysozyme injection did not change YFP absorption in contrast to YFP uptake reduced after GFP pretreat- ment. Lysozyme loading for 4 days resulted in a significant reduction in YFP absorption. The results show that receptor-mediated endocytosis in the frog kidney depends on the molecular nature of absorbable ligands, conditions of their competitive absorption and lysosomal accumulation in epithelial PT cells.


Subject(s)
Bacterial Proteins/pharmacokinetics , Green Fluorescent Proteins/pharmacokinetics , Kidney Tubules, Proximal/metabolism , Luminescent Proteins/pharmacokinetics , Muramidase/pharmacokinetics , Animals , Rana temporaria
7.
FASEB J ; 25(6): 1865-73, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21350116

ABSTRACT

Relatively weak tumor affinities and short retention time in vivo hinder the application of targeting peptides in tumor molecular imaging. Multivalent strategies based on various scaffolds have been utilized to improve the ability of peptide-receptor binding or extend the clearance time of peptide-based probes. Here, we use a tetrameric far-red fluorescent protein (tfRFP) as a scaffold to create a self-assembled octavalent peptide fluorescent nanoprobe (Octa-FNP) using a genetic engineering approach. The multiligand connecting, fluorophore labeling and nanostructure formation of Octa-FNP were performed in one step. In vitro studies showed Octa-FNP is a 10-nm fluorescent probe with excellent serum stability. Cellular uptake of Octa-FNP by human nasopharyngeal cancer 5-8F cells is 15-fold of tetravalent probe, ∼80-fold of monovalent probe and ∼600-fold of nulvalent tfRFP. In vivo enhanced tumor targeting and intracellular uptake of Octa-FNP were confirmed using optical imaging and Western blot analysis. It achieved extremely high contrast of Octa-FNP signal between tumor tissue and normal organs, especially seldom Octa-FNP detected in liver and spleen. Owing to easy preparation, precise structural and functional control, and multivalent effect, Octa-FNP provides a powerful tool for tumor optical molecular imaging and evaluating the targeting ability of numerous peptides in vivo.


Subject(s)
Drug Delivery Systems , Luminescent Agents/chemistry , Luminescent Agents/pharmacokinetics , Nanostructures , Animals , Antineoplastic Agents , Cell Line, Tumor , Genetic Engineering/methods , Humans , Luminescent Agents/metabolism , Luminescent Proteins/chemistry , Luminescent Proteins/metabolism , Luminescent Proteins/pharmacokinetics , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Nude , Models, Molecular , Neoplasms, Experimental/pathology , Protein Conformation , Red Fluorescent Protein
8.
Med Sci (Paris) ; 28(11): 958-62, 2012 Nov.
Article in French | MEDLINE | ID: mdl-23171899

ABSTRACT

The origin of hepatocytes and cholangiocytes in embryonic and adult liver, as well as the source of stellate and sinusoidal cells, have received much attention recently, due to the availability of biological tools enabling to trace the fate of cell lineages. A model is now proposed which defines the differentiation potential of hepatoblasts, ductal plate cells and adult progenitor cells, in normal conditions and in regenerating liver. The mesodermal origin of stellate and sinusoidal cells is also established.


Subject(s)
Bile Ducts/cytology , Cell Lineage , Hepatocytes/cytology , Liver Regeneration/physiology , Animals , Bacterial Proteins/analysis , Bacterial Proteins/pharmacokinetics , Bile Ducts/embryology , Endoderm/cytology , Gene Expression Regulation, Developmental , Hepatic Stellate Cells/cytology , Homeostasis , Liver/embryology , Liver/growth & development , Luminescent Proteins/analysis , Luminescent Proteins/pharmacokinetics , Mesoderm/cytology , Mice , Morphogenesis , SOX9 Transcription Factor/physiology , Zebrafish
9.
Nat Cell Biol ; 3(8): 723-9, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11483957

ABSTRACT

Rho-GTPase stabilizes microtubules that are oriented towards the leading edge in serum-starved 3T3 fibroblasts through an unknown mechanism. We used a Rho-effector domain screen to identify mDia as a downstream Rho effector involved in microtubule stabilization. Constitutively active mDia or activation of endogenous mDia with the mDia-autoinhibitory domain stimulated the formation of stable microtubules that were capped and oriented towards the wound edge. mDia co-localized with stable microtubules when overexpressed and associated with microtubules in vitro. Rho kinase was not necessary for the formation of stable microtubules. Our results show that mDia is sufficient to generate and orient stable microtubules, and indicate that Dia-related formins are part of a conserved pathway that regulates the dynamics of microtubule ends.


Subject(s)
3T3 Cells/enzymology , Cell Polarity/genetics , Microtubules/genetics , rho GTP-Binding Proteins/genetics , 3T3 Cells/cytology , Animals , Culture Media, Serum-Free/pharmacology , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Green Fluorescent Proteins , Indicators and Reagents/pharmacokinetics , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/pharmacokinetics , Mice , Microtubules/metabolism , Mutation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary/genetics , Transfection , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases
10.
Mol Brain ; 14(1): 151, 2021 09 28.
Article in English | MEDLINE | ID: mdl-34583741

ABSTRACT

The atypical chemokine receptor 3, ACKR3, is a G protein-coupled receptor, which does not couple to G proteins but recruits ßarrestins. At present, ACKR3 is considered a target for cancer and cardiovascular disorders, but less is known about the potential of ACKR3 as a target for brain disease. Further, mouse lines have been created to identify cells expressing the receptor, but there is no tool to visualize and study the receptor itself under physiological conditions. Here, we engineered a knock-in (KI) mouse expressing a functional ACKR3-Venus fusion protein to directly detect the receptor, particularly in the adult brain. In HEK-293 cells, native and fused receptors showed similar membrane expression, ligand induced trafficking and signaling profiles, indicating that the Venus fusion does not alter receptor signaling. We also found that ACKR3-Venus enables direct real-time monitoring of receptor trafficking using resonance energy transfer. In ACKR3-Venus knock-in mice, we found normal ACKR3 mRNA levels in the brain, suggesting intact gene transcription. We fully mapped receptor expression across 14 peripheral organs and 112 brain areas and found that ACKR3 is primarily localized to the vasculature in these tissues. In the periphery, receptor distribution aligns with previous reports. In the brain there is notable ACKR3 expression in endothelial vascular cells, hippocampal GABAergic interneurons and neuroblast neighboring cells. In conclusion, we have generated Ackr3-Venus knock-in mice with a traceable ACKR3 receptor, which will be a useful tool to the research community for interrogations about ACKR3 biology and related diseases.


Subject(s)
Bacterial Proteins/genetics , Brain/blood supply , Gene Knock-In Techniques , Genes, Reporter , Luminescent Proteins/genetics , Receptors, CXCR/genetics , Animals , Bacterial Proteins/analysis , Bacterial Proteins/pharmacokinetics , Biomarkers , Computer Systems , Endothelial Cells/chemistry , Endothelial Cells/cytology , GABAergic Neurons/chemistry , GABAergic Neurons/cytology , HEK293 Cells , Humans , Interneurons/chemistry , Interneurons/cytology , Ligands , Luminescent Proteins/analysis , Luminescent Proteins/pharmacokinetics , Mice , Organ Specificity , Receptors, CXCR/analysis , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacokinetics , Tissue Distribution , beta-Arrestin 1/metabolism
11.
Int J Biol Macromol ; 153: 100-106, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32105698

ABSTRACT

Nanoparticles are excellent imaging agents for cancer, but variability in chemical structure, racemic mixtures, and addition of heavy metals hinders FDA approval in the United States. We developed a small ultra-red fluorescent protein, named smURFP, to have optical properties similar to the small-molecule Cy5, a heptamethine subclass of cyanine dyes (Ex/Em = 642/670 nm). smURFP has a fluorescence quantum yield of 18% and expresses so well in E. coli, that gram quantities of fluorescent protein are purified from cultures in the laboratory. In this research, the fluorescent protein smURFP was combined with bovine serum albumin into fluorescent protein nanoparticles. These nanoparticles are fluorescent with a quantum yield of 17% and 12-14 nm in diameter. The far-red fluorescent protein nanoparticles noninvasively image tumors in living mice via the enhanced permeation and retention (EPR) mechanism. This manuscript describes the use of a new fluorescent protein nanoparticle for in vivo fluorescent imaging. This protein nanoparticle core should prove useful as a biomacromolecular scaffold, which could bear extended chemical modifications for studies, such as the in vivo imaging of fluorescent protein nanoparticles targeted to primary and metastatic cancer, theranostic treatment, and/or dual-modality imaging with positron emission tomography for entire human imaging.


Subject(s)
Fluorescent Dyes , Luminescent Proteins , Lung Neoplasms , Nanoparticles/chemistry , Optical Imaging , A549 Cells , Animals , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacokinetics , Fluorescent Dyes/pharmacology , Heterografts , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/pharmacokinetics , Luminescent Proteins/pharmacology , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Red Fluorescent Protein
12.
J Cell Biol ; 145(3): 437-45, 1999 May 03.
Article in English | MEDLINE | ID: mdl-10225946

ABSTRACT

Phytochrome is a ubiquitous photoreceptor of plants and is encoded by a small multigene family. We have shown recently that a functional nuclear localization signal may reside within the COOH-terminal region of a major member of the family, phytochrome B (phyB) (Sakamoto, K., and A. Nagatani. 1996. Plant J. 10:859-868). In the present study, a fusion protein consisting of full-length phyB and the green fluorescent protein (GFP) was overexpressed in the phyB mutant of Arabidopsis to examine subcellular localization of phyB in intact tissues. The resulting transgenic lines exhibited pleiotropic phenotypes reported previously for phyB overexpressing plants, suggesting that the fusion protein is biologically active. Immunoblot analysis with anti-phyB and anti-GFP monoclonal antibodies confirmed that the fusion protein accumulated to high levels in these lines. Fluorescence microscopy of the seedlings revealed that the phyB-GFP fusion protein was localized to the nucleus in light grown tissues. Interestingly, the fusion protein formed speckles in the nucleus. Analysis of confocal optical sections confirmed that the speckles were distributed within the nucleus. In contrast, phyB-GFP fluorescence was observed throughout the cell in dark-grown seedlings. Therefore, phyB translocates to specific sites within the nucleus upon photoreceptor activation.


Subject(s)
Arabidopsis/genetics , Cell Nucleus/metabolism , Indicators and Reagents/pharmacokinetics , Luminescent Proteins/pharmacokinetics , Nuclear Localization Signals , Photoreceptor Cells , Phytochrome/pharmacokinetics , Transcription Factors , Arabidopsis/metabolism , Arabidopsis Proteins , Color , Darkness , Green Fluorescent Proteins , Hypocotyl/physiology , Immunoblotting , Lighting , Luminescent Proteins/analysis , Microscopy, Confocal , Nuclear Proteins/metabolism , Photic Stimulation , Phytochrome/analysis , Phytochrome B , Plant Proteins/metabolism , Plants, Genetically Modified , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/pharmacokinetics
13.
J Cell Biol ; 137(5): 965-74, 1997 Jun 02.
Article in English | MEDLINE | ID: mdl-9166399

ABSTRACT

The perinucleolar compartment (PNC) is a unique nuclear structure preferentially localized at the periphery of the nucleolus. Several small RNAs transcribed by RNA polymerase III (e.g., the Y RNAs, MRP RNA, and RNase P H1 RNA) and the polypyrimidine tract binding protein (PTB; hnRNP I) have thus far been identified in the PNC (Ghetti, A., S. PinolRoma, W.M. Michael, C. Morandi, and G. Dreyfuss. 1992. Nucleic Acids Res. 20:3671-3678; Matera, A.G., M.R. Frey, K. Margelot, and S.L. Wolin. 1995. J. Cell Biol. 129:1181-1193; Lee, B., A.G. Matera, D.C. Ward, and J. Craft. 1996. Proc. Natl. Acad. Sci. USA. 93: 11471-11476). In this report, we have further characterized this structure in both fixed and living cells. Detection of the PNC in a large number of human cancer and normal cells showed that PNCs are much more prevalent in cancer cells. Analysis through the cell cycle using immunolabeling with a monoclonal antibody, SH54, specifically recognizing PTB, demonstrated that the PNC dissociates at the beginning of mitosis and reforms at late telophase in the daughter nuclei. To visualize the PNC in living cells, a fusion protein between PTB and green fluorescent protein (GFP) was generated. Time lapse studies revealed that the size and shape of the PNC is dynamic over time. In addition, electron microscopic examination in optimally fixed cells revealed that the PNC is composed of multiple strands, each measuring approximately 80-180 nm diam. Some of the strands are in direct contact with the surface of the nucleolus. Furthermore, analysis of the sequence requirement for targeting PTB to the PNC using a series of deletion mutants of the GFP-PTB fusion protein showed that at least three RRMs at either the COOH or NH2 terminus are required for the fusion protein to be targeted to the PNC. This finding suggests that RNA binding may be necessary for PTB to be localized in the PNC.


Subject(s)
Cell Compartmentation/physiology , Cell Nucleolus/physiology , Adenocarcinoma , Amino Acid Sequence , Biological Transport/physiology , Breast Neoplasms , Carcinoma, Ductal, Breast , Cell Cycle/physiology , Cell Line, Transformed , Cell Nucleolus/ultrastructure , Colonic Neoplasms , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacokinetics , Female , Fibroblasts/cytology , Fibroblasts/physiology , Fibroblasts/ultrastructure , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/pharmacokinetics , Lung/cytology , Microscopy, Electron , Mutagenesis/physiology , Phenotype , Polypyrimidine Tract-Binding Protein , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , RNA-Binding Proteins/pharmacokinetics , Recombinant Fusion Proteins/pharmacokinetics , Skin/cytology
14.
J Cell Biol ; 154(3): 535-47, 2001 Aug 06.
Article in English | MEDLINE | ID: mdl-11481344

ABSTRACT

Sphingolipids (SLs) are plasma membrane constituents in eukaryotic cells which play important roles in a wide variety of cellular functions. However, little is known about the mechanisms of their internalization from the plasma membrane or subsequent intracellular targeting. We have begun to study these issues in human skin fibroblasts using fluorescent SL analogues. Using selective endocytic inhibitors and dominant negative constructs of dynamin and epidermal growth factor receptor pathway substrate clone 15, we found that analogues of lactosylceramide and globoside were internalized almost exclusively by a clathrin-independent ("caveolar-like") mechanism, whereas an analogue of sphingomyelin was taken up approximately equally by clathrin-dependent and -independent pathways. We also showed that the Golgi targeting of SL analogues internalized via the caveolar-like pathway was selectively perturbed by elevated intracellular cholesterol, demonstrating the existence of two discrete Golgi targeting pathways. Studies using SL-binding toxins internalized via clathrin-dependent or -independent mechanisms confirmed that endogenous SLs follow the same two pathways. These findings (a) provide a direct demonstration of differential SLs sorting into early endosomes in living cells, (b) provide a "vital marker" for endosomes derived from caveolar-like endocytosis, and (c) identify two independent pathways for lipid transport from the plasma membrane to the Golgi apparatus in human skin fibroblasts.


Subject(s)
Antigens, CD , Cell Membrane/metabolism , Clathrin/metabolism , Globosides/pharmacokinetics , Golgi Apparatus/metabolism , Lactosylceramides/pharmacokinetics , Adaptor Proteins, Signal Transducing , Boron Compounds/pharmacokinetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caveolin 1 , Caveolins/metabolism , Cells, Cultured , Dynamins , Endocytosis/physiology , Endosomes/metabolism , Fibroblasts/cytology , Fluorescent Dyes/pharmacokinetics , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gangliosidoses/metabolism , Green Fluorescent Proteins , Humans , Indicators and Reagents/pharmacokinetics , Intracellular Signaling Peptides and Proteins , Luminescent Proteins/pharmacokinetics , Mutagenesis/physiology , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Transport/physiology , Skin/cytology
15.
Nat Commun ; 10(1): 3452, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31388002

ABSTRACT

Bacteria have been extensively utilized for bioimaging, diagnosis and therapy given their unique characteristics including genetic manipulation, rapid proliferation and disease site targeting specificity. However, clinical translation of bacteria for these applications has been largely restricted by their unavoidable side effects and low treatment efficacies. Engineered bacteria for biomedical applications ideally need to generate only a low inflammatory response, show slow elimination by macrophages, low accumulation in normal organs, and almost unchanged inherent bioactivities. Here we describe a set of stealth bacteria, cell membrane coated bacteria (CMCB), meeting these requirement. Our findings are supported by evaluation in multiple mice models and ultimately demonstrate the potential of CMCB to serve as efficient tumor imaging agents. Stealth bacteria wrapped up with cell membranes have the potential for a myriad of bacterial-mediated biomedical applications.


Subject(s)
Bacteria/genetics , Cell Engineering/methods , Diagnostic Imaging/methods , Drug Delivery Systems/methods , Neoplasms/diagnostic imaging , Animals , Bacteria/immunology , Cell Line, Tumor/transplantation , Cell Membrane/immunology , Disease Models, Animal , Erythrocytes/ultrastructure , Female , Humans , Luminescent Proteins/administration & dosage , Luminescent Proteins/pharmacokinetics , Macrophages , Male , Mice , Primary Cell Culture , Probiotics , Proof of Concept Study , Tissue Distribution
16.
Neuron ; 26(2): 331-43, 2000 May.
Article in English | MEDLINE | ID: mdl-10839353

ABSTRACT

Presynaptic terminals contain highly organized subcellular structures to facilitate neurotransmitter release. In C. elegans, the typical presynaptic terminal has an electron-dense active zone surrounded by synaptic vesicles. Loss-of-function mutations in the rpm-1 gene result in abnormally structured presynaptic terminals in GABAergic neuromuscular junctions (NMJs), most often manifested as a single presynaptic terminal containing multiple active zones. The RPM-1 protein has an RCC1-like guanine nucleotide exchange factor (GEF) domain and a RING-H2 finger. RPM-1 is most similar to the Drosophila presynaptic protein Highwire (HIW) and the mammalian Myc binding protein Pam. RPM-1 is localized to the presynaptic region independent of synaptic vesicles and functions cell autonomously. The temperature-sensitive period of rpm-1 coincides with the time of synaptogenesis. rpm-1 may regulate the spatial arrangement, or restrict the formation, of presynaptic structures.


Subject(s)
Adaptor Proteins, Signal Transducing , Caenorhabditis elegans Proteins , Caenorhabditis elegans/metabolism , Drosophila Proteins , Guanine Nucleotide Exchange Factors/physiology , Mixed Function Oxygenases , Presynaptic Terminals/physiology , Amino Acid Sequence , Animals , Biomarkers , Carrier Proteins/genetics , Drosophila/metabolism , Green Fluorescent Proteins , Guanine Nucleotide Exchange Factors/genetics , Indicators and Reagents/pharmacokinetics , Luminescent Proteins/pharmacokinetics , Molecular Sequence Data , Mutation/genetics , Mutation/physiology , Nerve Tissue Proteins/genetics , Neurons/physiology , Presynaptic Terminals/metabolism , Receptors, GABA/metabolism , Synapses/metabolism , Synapses/physiology , Synaptic Vesicles/metabolism , Synaptic Vesicles/ultrastructure , Tissue Distribution , Ubiquitin-Protein Ligases , Zinc Fingers/genetics
17.
Neuron ; 32(1): 113-22, 2001 Oct 11.
Article in English | MEDLINE | ID: mdl-11604143

ABSTRACT

IP(3) signaling in Purkinje cells is involved in the regulation of cell functions including LTD. We have used a GFP-tagged pleckstrin homology domain to visualize IP(3) dynamics in Purkinje cells. Surprisingly, IP(3) production was observed in response not only to mGluR activation, but also to AMPA receptor activation in Purkinje cells in culture. AMPA-induced IP(3) production was mediated by depolarization-induced Ca(2+) influx because it was mimicked by depolarization and was blocked by inhibition of the P-type Ca(2+) channel. Furthermore, trains of complex spikes, elicited by climbing fiber stimulation (1 Hz), induced IP(3) production in Purkinje cells in cerebellar slices. These results revealed a novel IP(3) signaling pathway in Purkinje cells that can be elicited by synaptic inputs from climbing fibers.


Subject(s)
Calcium Channels/metabolism , Cycloleucine/analogs & derivatives , Glycine/analogs & derivatives , Inositol 1,4,5-Trisphosphate/metabolism , Purkinje Cells/metabolism , Receptors, AMPA/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione , Animals , Benzoates/pharmacology , Blood Proteins/genetics , Blood Proteins/pharmacokinetics , Calcium/metabolism , Cell Membrane/metabolism , Cells, Cultured , Cycloleucine/pharmacology , Cytoplasm/metabolism , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Genetic Vectors , Glutamic Acid/pharmacology , Glycine/pharmacology , Green Fluorescent Proteins , Indicators and Reagents/pharmacokinetics , Inositol 1,4,5-Trisphosphate/biosynthesis , Luminescent Proteins/genetics , Luminescent Proteins/pharmacokinetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred ICR , Neuroprotective Agents/pharmacology , Patch-Clamp Techniques , Phosphoproteins/genetics , Phosphoproteins/pharmacokinetics , Purkinje Cells/cytology , Receptors, Metabotropic Glutamate/metabolism , Sindbis Virus/genetics , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
18.
Nucl Med Biol ; 35(2): 159-69, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18312825

ABSTRACT

A construct for tagging neurospheres and monitoring cell transplantations was developed using a new technology for producing luminescent and radiolabeled probes that have identical structures. The HIV1-Tat basic domain derivatives NAcGRKKRRQRRR(SAACQ)G (SAACQ-1) and [NAcGRKKRRQRRR(Re(CO)3SAACQ)G]+ (ReSAACQ-1) were prepared in excellent yields using the single amino acid chelate-quinoline (SAACQ) ligand and its Re(I) complex and conventional automated peptide synthesis methods. The distribution of the luminescent Re probe, using epifluorescence microscopy, showed that it localized primarily in the cell nucleus with a significant degree of association on the nuclear envelope. A smaller amount was found to be dispersed in the cytoplasm. The 99m Tc analogue was then prepared in 43+/-7% (n=12) yield and very high effective specific activity. Following incubation, average uptake of the probe in neurospheres ranged between 10 and 20 Bq/cell. As determined by colorimetric assays, viability for cells labeled with high effective specific activity 99m TcSAACQ-1 was 97+/-4% at 2 h postlabeling and 85+/-25% at 24 h postlabeling for incubation activities ranging from 245 to 8900 Bq/cell. DNA analysis showed that at these levels, there was no significant difference between the extent of DNA damage in the treated cells versus control cells. A series of preliminary SPECT/CT studies of transplants in mice were performed, which showed that the strategy is convenient and feasible and that it is possible to routinely assess procedures noninvasively and determine the number of cells transplanted.


Subject(s)
Luminescent Proteins/chemical synthesis , Luminescent Proteins/pharmacokinetics , Nervous System , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacokinetics , Stem Cell Transplantation , Stem Cells/cytology , Animals , Cells, Cultured , Chelating Agents/chemistry , Cytoplasm/diagnostic imaging , Cytoplasm/metabolism , Fluorescent Dyes/chemistry , Head/diagnostic imaging , Head/pathology , Isotope Labeling , Mice , Nervous System/cytology , Nervous System/diagnostic imaging , Nuclear Envelope/diagnostic imaging , Nuclear Envelope/metabolism , Quinolines/chemistry , Staining and Labeling/methods , Stem Cells/diagnostic imaging , Technetium , Tomography, Emission-Computed, Single-Photon , Tomography, X-Ray Computed , Whole Body Imaging , tat Gene Products, Human Immunodeficiency Virus/chemistry , tat Gene Products, Human Immunodeficiency Virus/pharmacokinetics
19.
Reprod Fertil Dev ; 20(5): 547-56, 2008.
Article in English | MEDLINE | ID: mdl-18577351

ABSTRACT

In the present study, we have related mitochondrial membrane potential (DeltaPsim) and forward scatter (FSC) to apoptotic-related changes in spermatozoa. Thawed red deer spermatozoa were incubated in synthetic oviductal fluid medium (37 degrees C, 5% CO2), with or without antioxidant (100 microm Trolox). At 0, 3, 6 and 9 h, aliquots were assessed for motility and were stained with a combination of Hoechst 33342, propidium ioide (PI), YO-PRO-1 and Mitotracker Deep Red for flow cytometry. The proportion of spermatozoa YO-PRO-1+ and PI+ (indicating a damaged plasmalemma; DEAD) increased, whereas that of YO-PRO-1- and PI- (INTACT) spermatozoa decreased. The proportion of YO-PRO-1+ and PI- spermatozoa (altered plasmalemma; APOPTOTIC) did not change. Both DEAD and APOPTOTIC spermatozoa had low DeltaPsim. Most high-DeltaPsim spermatozoa were INTACT, and their proportion decreased with time. The FSC signal also differed between different groups of spermatozoa, in the order APOPTOTIC > DEAD > INTACT/low DeltaPsim > INTACT/high DeltaPsim; however, the actual meaning of this difference is not clear. APOPTOTIC spermatozoa seemed motile at 0 h, but lost motility with time. Trolox only slightly improved the percentage of INTACT spermatozoa (P < 0.05). The population of APOPTOTIC spermatozoa in the present study may be dying cells, possibly with activated cell death pathways (loss of DeltaPsim). We propose that the sequence of spermatozoon death here would be: (1) loss of DeltaPsim; (2) membrane changes (YO-PRO-1+ and PI-); and (3) membrane damage (PI+). INTACT spermatozoa with low DeltaPsim or altered FSC may be compromised cells. The present study is the first that directly relates membrane integrity, apoptotic markers and mitochondrial status in spermatozoa. The results of the present study may help us understand the mechanisms leading to loss of spermatozoon viability after thawing.


Subject(s)
Apoptosis/physiology , Cell Membrane/physiology , Mitochondria/physiology , Spermatozoa/pathology , Spermatozoa/physiology , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Membrane/drug effects , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/physiology , Deer , Flow Cytometry , Luminescent Proteins/pharmacokinetics , Male , Membrane Potential, Mitochondrial/physiology , Necrosis , Sperm Count , Sperm Motility , Spermatozoa/drug effects , Red Fluorescent Protein
20.
J Control Release ; 287: 132-141, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30016735

ABSTRACT

Recombinant albumin-drug genetic fusions are an effective technology to prolong the serum half-life of therapeutics that has resulted in marketed products. Indirect evidence suggests albumin fusions' long circulation is controlled by engagement with the cellular recycling neonatal Fc receptor (FcRn) in addition to reduced kidney filtration. In this work, we have used a panel of recombinant fusions, engineered with different human FcRn (hFcRn) affinity, including a novel high binding albumin variant (HBII), to directly define and importantly, control the intracellular mechanism as a half-life extension tuning method. mNeonGreen or mCherry fusion to the N-terminal of the recombinant human albumin (rHA) variants null-binder (rHA NB), wild-type (rHA WT), high-binder I (rHA HBI), and high-binder II (rHA HBII) did not generally interfere with hFcRn interaction determined by Biolayer Interferometry. Co-localisation of the albumins with endosomal, but not lysosomal, markers was shown by confocal microscopy for high, but not low, hFcRn binders in a human microvascular endothelial hFcRn overexpressing cell line (HMEC-1 FcRn) suggestive of endosomal compartmentalisation. Furthermore, a cellular recycling assay revealed increased recycling of albumin fusions for the high binding variants (mNeonGreen WT; ~1, mNeonGreen HBI; 5.26-fold higher, and mNeonGreen HBII; 5.77-fold higher) in the hFcRn overexpressing cell line. In vivo experiments demonstrated a direct in vitro recycling/in vivo half-life correlation with a longer circulation for the mCherry fusions engineered with high hFcRn affinity that was highest with the HBII variant of 30.1 h compared to 18.2 h for the mCherry WT. This work gives the first direct evidence for an FcRn-driven endosomal cellular recycling pathway for recombinant albumin fusions that correlates with half-life extension controlled by the affinity to hFcRn; promoting a versatile method to tune the pharmacokinetics of albumin fusion-based therapeutics not met by current technologies.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Luminescent Proteins/pharmacokinetics , Receptors, Fc/metabolism , Serum Albumin, Human/pharmacokinetics , Animals , Cell Line , Half-Life , Humans , Luminescent Proteins/metabolism , Mice, Inbred BALB C , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacokinetics , Serum Albumin, Human/metabolism , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL