Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 22(5): 1463-1474, 2024 May.
Article in English | MEDLINE | ID: mdl-38266680

ABSTRACT

BACKGROUND: Extracellular vesicles (EVs), in particular those derived from activated platelets, are associated with a risk of future venous thromboembolism. OBJECTIVES: To study the biomolecular profile and function characteristics of EVs from control (unstimulated) and activated platelets. METHODS: Biomolecular profiling of single or very few (1-4) platelet-EVs (control/stimulated) was performed by Raman tweezers microspectroscopy. The effects of such EVs on the coagulation system were comprehensively studied. RESULTS: Raman tweezers microspectroscopy of platelet-EVs followed by biomolecular component analysis revealed for the first time 3 subsets of EVs: (i) protein rich, (ii) protein/lipid rich, and (iii) lipid rich. EVs from control platelets presented a heterogeneous biomolecular profile, with protein-rich EVs being the main subset (58.7% ± 3.5%). Notably, the protein-rich subset may contain a minor contribution from other extracellular particles, including protein aggregates. In contrast, EVs from activated platelets were more homogeneous, dominated by the protein/lipid-rich subset (>85%), and enriched in phospholipids. Functionally, EVs from activated platelets increased thrombin generation by 52.4% and shortened plasma coagulation time by 34.6% ± 10.0% compared with 18.6% ± 13.9% mediated by EVs from control platelets (P = .015). The increased procoagulant activity was predominantly mediated by phosphatidylserine. Detailed investigation showed that EVs from activated platelets increased the activity of the prothrombinase complex (factor Va:FXa:FII) by more than 6-fold. CONCLUSION: Our study reports a novel quantitative biomolecular characterization of platelet-EVs possessing a homogenous and phospholipid-enriched profile in response to platelet activation. Such characteristics are accompanied with an increased phosphatidylserine-dependent procoagulant activity. Further investigation of a possible role of platelet-EVs in the pathogenesis of venous thromboembolism is warranted.


Subject(s)
Blood Coagulation , Blood Platelets , Extracellular Vesicles , Phospholipids , Platelet Activation , Spectrum Analysis, Raman , Humans , Blood Platelets/metabolism , Extracellular Vesicles/metabolism , Phospholipids/metabolism , Thrombin/metabolism , Thromboplastin/metabolism , Enzyme Activation
2.
Front Cell Dev Biol ; 11: 1307502, 2023.
Article in English | MEDLINE | ID: mdl-38046667

ABSTRACT

In our study, we harnessed an original Enhanced Speed Structured Illumination Microscopy (Fast-SIM) imaging setup to explore the dynamics of mitochondrial and inner membrane ultrastructure under specific photo-oxidation stress induced by Chlorin-e6 and light irradiation. Notably, our Fast-SIM system allowed us to observe and quantify a distinct remodeling and shortening of the mitochondrial structure after 60-80 s of irradiation. These changes were accompanied by fusion events of adjacent inner membrane cristae and global swelling of the organelle. Preceding these alterations, a larger sequence was characterized by heightened dynamics within the mitochondrial network, featuring events such as mitochondrial fission, rapid formation of tubular prolongations, and fluctuations in cristae structure. Our findings provide compelling evidence that, among enhanced-resolution microscopy techniques, Fast-SIM emerges as the most suitable approach for non-invasive dynamic studies of mitochondrial structure in living cells. For the first time, this approach allows quantitative and qualitative characterization of successive steps in the photo-induced oxidation process with sufficient spatial and temporal resolution.

3.
Nanomaterials (Basel) ; 10(4)2020 Apr 16.
Article in English | MEDLINE | ID: mdl-32316367

ABSTRACT

Surface-enhanced fluorescence (SEF) requires the absorption/emission band of the fluorophore, the localized surface plasmon resonance (LSPR) of the nanostructure and the excitation wavelength to fall in the same (or very close) spectral range. In this paper, we monitor the SEF intensity and lifetime dependence of riboflavin (vitamin B2) adsorbed on a spacer-modified Ag substrate with respect to the thickness of the spacer. The substrates were formed by silver nanoislands deposited onto magnetron-sputtered polytetrafluoroethylene (ms-PTFE). The spacer was formed by the ms-PTFE layer with the thickness ranging from ~5 to 25 nm. The riboflavin dissolved in dimethylsulfoxide (DMSO) at a 10 µM concentration forms, at the ms-PTFE surface, a homogeneous layer of adsorbed molecules corresponding to a monomolecular layer. The microspectroscopic measurements of the adsorbed layer were performed through a sessile droplet; our study has shown the advantages and limitations of this approach. Time-resolved fluorescence enabled us to determine the enhanced fluorescence quantum yield due to the shortening of the radiative decay in the vicinity of the plasmonic surface. For the 5 nm ms-PTFE layer possessing the largest (estimated 4×) fluorescence enhancement, the quantum yield was increased 2.3×.

4.
Cells ; 9(2)2020 02 04.
Article in English | MEDLINE | ID: mdl-32033136

ABSTRACT

Curcumin, a major active component of turmeric (Curcuma longa, L.), is known to have various effects on both healthy and cancerous tissues. In vitro studies suggest that curcumin inhibits cancer cell growth by activating apoptosis, but the mechanism underlying the anticancer effect of curcumin is still unclear. Since there is a recent consensus about endoplasmic reticulum (ER) stress being involved in the cytotoxicity of natural compounds, we have investigated using Image flow cytometry the mechanistic aspects of curcumin's destabilization of the ER, but also the status of the lysosomal compartment. Curcumin induces ER stress, thereby causing an unfolded protein response and calcium release, which destabilizes the mitochondrial compartment and induce apoptosis. These events are also associated with secondary lysosomal membrane permeabilization that occurs later together with an activation of caspase-8, mediated by cathepsins and calpains that ended in the disruption of mitochondrial homeostasis. These two pathways of different intensities and momentum converge towards an amplification of cell death. In the present study, curcumin-induced autophagy failed to rescue all cells that underwent type II cell death following initial autophagic processes. However, a small number of cells were rescued (successful autophagy) to give rise to a novel proliferation phase.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Curcumin/pharmacology , Calcium/metabolism , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Subcellular Fractions/metabolism , Unfolded Protein Response/drug effects
5.
Cell Death Discov ; 5: 150, 2019.
Article in English | MEDLINE | ID: mdl-31839992

ABSTRACT

Iron overload, notably caused by hereditary hemochromatosis, is an excess storage of iron in various organs that causes tissue damage and may promote tumorigenesis. To manage that disorder, free iron depletion can be induced by iron chelators like deferoxamine that are of increasing interest also in the cancer field since iron stock could be a potent target for managing tumorigenesis. Curcumin, a well-known active substance extracted from the turmeric rhizome, destabilizes endoplasmic reticulum, and secondarily lysosomes, thereby increasing mitophagy/autophagy and subsequent apoptosis. Recent findings show that cells treated with curcumin also exhibit a decrease in ferritin, which is consistent with its chemical structure and iron chelating activity. Here we investigated how curcumin influences the intracellular effects of iron overload via Fe-nitriloacetic acid or ferric ammonium citrate loading in Huh-7 cells and explored the consequences in terms of antioxidant activity, autophagy, and apoptotic signal transduction. In experiments with T51B and RL-34 epithelial cells, we have found evidence that curcumin-iron complexation abolishes both curcumin-induced autophagy and apoptosis, together with the tumorigenic action of iron overload.

6.
Sci Rep ; 8(1): 10336, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29985417

ABSTRACT

To increase our understanding of bacterial biofilm complexity, real- time quantitative analyses of the living community functions are required. To reach this goal, accurate fluorescent reporters are needed. In this paper, we used the classical fluorescent genetic reporters of the GFP family and demonstrated their limits in the context of a living biofilm. We showed that fluorescence signal saturated after only a few hours of growth and related this saturation to the reduction of oxygen concentration induced by bacterial consumption. This behaviour prevents the use of GFP-like fluorescent proteins for quantitative measurement in living biofilms. To overcome this limitation, we propose the use of a recently introduced small protein tag, FAST, which is fluorescent in the presence of an exogenously applied fluorogenic dye, enabling to avoid the oxygen sensitivity issue. We compared the ability of FAST to report on biofilm growth with that of GFP and mCherry, and demonstrated the superiority of the FAST:fluorogen probes for investigating dynamics in the complex environment of a living biofilm.


Subject(s)
Biofilms/growth & development , Escherichia coli/physiology , Green Fluorescent Proteins/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Red Fluorescent Protein
7.
Int J Pharm ; 510(1): 240-9, 2016 Aug 20.
Article in English | MEDLINE | ID: mdl-27282536

ABSTRACT

Recent years have seen a growing interest in Berberine, a phytochemical with multispectrum therapeutic activities, as anti-tumoral agent for photodynamic therapy (PDT). In this context, low density lipoproteins (LDL) play a key role in the delivery of the photosensitizer in tumor cells. We correlate the physicochemical parameters of the berberine association to LDL with the influence of LDL-delivery on its accumulation in a glioma cell line and on its photo-induced activity in view of antitumor PDT. Our results evidence an important binding of 400 berberine molecules per LDL. Changes in berberine and apoprotein fluorescence suggest different fixation types, involving various LDL compartments including the vicinity of the apoprotein. The berberine association to LDL does not affect their recognition by the specific B/E receptors, of which over-expression increases the cellular uptake of LDL-preloaded berberine. Fluorescence microscopy evidences the mitochondrial labeling of the glioma model cells, with no significant modification upon LDL-delivery. Moreover, the cellular delivery of berberine by LDL increases its photocytotoxic effects on such cells. So, this research illustrates the potential of berberine as a photosensitizing agent for PDT, in particular due to their behavior towards LDL as plasma vehicles, and gives insights into its mechanisms of cell uptake.


Subject(s)
Antineoplastic Agents/metabolism , Berberine/metabolism , Lipoproteins, LDL/metabolism , Photochemotherapy/methods , Photosensitizing Agents/metabolism , Antineoplastic Agents/administration & dosage , Berberine/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/physiology , Humans , Photic Stimulation/methods , Photosensitizing Agents/administration & dosage
8.
Int J Pharm ; 495(2): 750-60, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26387620

ABSTRACT

Block-polymer nanoparticles are now well-known candidates for the delivery of various non-soluble drugs to cells. The release of drugs from these nanoparticles is a major concern related to their efficiency as nanovectors and is still not completely deciphered. Various processes have been identified, depending of both the nature of the block-polymer and those of the drugs used. We focused our interest on an amphiphilic photosensitizer studied for photodynamic treatments of cancer, Pheophorbide-a (Pheo). We studied the transfer of Pheo from poly(ethyleneglycol-b-ϵ-caprolactone) nanoparticles (I) to MCF-7 cancer cells and (II) to models of membranes. Altogether, our results suggest that the delivery of the major part of the Pheo by the nanoparticles occurs via a direct transfer of Pheo from the nanoparticles to the membrane, by collision. A minor process may involve the internalization of a small amount of the nanoplatforms by the cells. So, this research illustrates the great care necessary to address the question of the choice of such nanocarriers, in relation with the properties - in particular the relative hydrophobicity - of the drugs encapsulated, and gives elements to predict the mechanism and the efficiency of the delivery.


Subject(s)
Chlorophyll/analogs & derivatives , Drug Delivery Systems , Nanoparticles , Radiation-Sensitizing Agents/administration & dosage , Chemistry, Pharmaceutical/methods , Chlorophyll/administration & dosage , Chlorophyll/chemistry , Chlorophyll/pharmacokinetics , Drug Carriers/chemistry , Drug Liberation , Humans , Hydrophobic and Hydrophilic Interactions , Lactones/chemistry , MCF-7 Cells , Polyethylene Glycols/chemistry , Radiation-Sensitizing Agents/chemistry , Radiation-Sensitizing Agents/pharmacokinetics , Solubility
9.
J Fluoresc ; 25(5): 1245-50, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26179074

ABSTRACT

Time-resolved microspectrofluorimetry and fluorescence microscopy imaging-two complementary fluorescence techniques-provide important information about the intracellular distribution, level of uptake and binding/interactions inside living cell of the labeled molecule of interest. They were employed to monitor the "fate" of AS1411 aptamer labeled by ATTO 425 in human living cells. Confocal microspectrofluorimeter adapted for time-resolved intracellular fluorescence measurements by using a phase-modulation principle with homodyne data acquisition was employed to obtain emission spectra and to determine fluorescence lifetimes in U-87 MG tumor brain cells and Hs68 non-tumor foreskin cells. Acquired spectra from both the intracellular space and the reference solutions were treated to observe the aptamer localization and its interaction with biological structures inside the living cell. The emission spectra and the maximum emission wavelengths coming from the cells are practically identical, however significant lifetime lengthening was observed for tumor cell line in comparison to non-tumor one.


Subject(s)
Aptamers, Nucleotide/metabolism , Intracellular Space/metabolism , Microscopy, Fluorescence/methods , Oligodeoxyribonucleotides/metabolism , Spectrometry, Fluorescence/methods , Base Sequence , Cell Line, Tumor , Humans , Intracellular Space/genetics , Oligodeoxyribonucleotides/genetics , Time Factors
10.
Apoptosis ; 19(12): 1779-92, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25300800

ABSTRACT

In order to explain the contribution of the protein kinase Cα (PKCα) in apoptosis induced by photo-activation of hypericin (Hyp), a small interfering RNA was used for post-transcriptional silencing of pkcα gene expression. We have evaluated the influence of Hyp photo-activation on cell death in non-transfected and transfected (PKCα(-)) human glioma cells (U-87 MG). No significant differences were detected in cell survival between non-transfected and transfected PKCα(-) cells. However, the type of cell death was notably affected by silencing the pkcα gene. Photo-activation of Hyp strongly induced apoptosis in non-transfected cells, but the level of necrotic cells in transfected PKCα(-) cells increased significantly. The differences in cell death after Hyp photo-activation are demonstrated by changes in: (i) reactive oxygen species production, (ii) Bcl-2 phosphorylation on Ser70 (pBcl-2(Ser70)), (iii) cellular distributions of pBcl-2(Ser70) and (iv) cellular distribution of endogenous anti-oxidant glutathione and its co-localization with mitochondria. In summary, we suggest that post-transcriptional silencing of the pkcα gene and the related decrease of PKCα level considerably affects the anti-apoptotic function and the anti-oxidant function of Bcl-2. This implies that PKCα, as Bcl-2 kinase, indirectly protects U-87 MG cells against oxidative stress and subsequent cell death.


Subject(s)
Apoptosis/drug effects , Perylene/analogs & derivatives , Photosensitizing Agents/pharmacology , Protein Kinase C-alpha/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Anthracenes , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression , Humans , Mitochondria/metabolism , Necrosis , Perylene/pharmacology , Phosphorylation , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Reactive Oxygen Species/metabolism , Serine/metabolism
11.
Int J Pharm ; 436(1-2): 463-71, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22814227

ABSTRACT

Low-density lipoproteins (LDL), a natural in vivo carrier of cholesterol in the vascular system, play a key role in the delivery of hydrophobic/amphiphilic photosensitizers to tumor cells in photodynamic therapy of cancer. To make this delivery system even more efficient, we have constructed a nano-delivery system by coating of LDL surface by dextran. Fluorescence spectroscopy, confocal fluorescence imaging, stopped-flow experiments and flow-cytometry were used to characterize redistribution of hypericin (Hyp), a natural occurring potent photosensitizer, loaded in LDL/dextran complex to free LDL molecules as well as to monitor cellular uptake of Hyp by U87-MG cells. It is shown that the redistribution process of Hyp between LDL molecules is significantly suppressed by dextran coating of LDL surface. The modification of LDL molecules by dextran does not inhibit their recognition by cellular LDL receptors and U-87 MG cellular uptake of Hyp loaded in LDL/dextran complex appears to be similar to that one observed for Hyp transported by unmodified LDL particles. Thus, it is proposed that dextran modified LDL molecules could be used as a basis for construction of a drug transport system for targeted delivery of hydrophobic/amphiphilic drugs to cancer cells expressing high level of LDL receptors.


Subject(s)
Dextrans/chemistry , Drug Carriers/chemistry , Lipoproteins, LDL/chemistry , Perylene/analogs & derivatives , Radiation-Sensitizing Agents/chemistry , Anthracenes , Cell Line, Tumor , Dextrans/administration & dosage , Drug Carriers/administration & dosage , Humans , Hydrophobic and Hydrophilic Interactions , Lipoproteins, LDL/administration & dosage , Neoplasms/metabolism , Perylene/administration & dosage , Perylene/chemistry , Radiation-Sensitizing Agents/administration & dosage
12.
Photochem Photobiol Sci ; 11(9): 1428-36, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22729350

ABSTRACT

Hypericin (Hyp) is a hydrophobic natural photosensitizer that is considered to be a promising molecule for photodynamic treatment of tumor cells and photo-diagnosis of early epithelial cancers. Its hydrophobicity is the main driving force that governs its redistribution process. Low-density lipoproteins (LDL), a natural in vivo carrier of cholesterol present in the vascular system, have been used for targeted transport of Hyp to U87 glioma cells. For low Hyp-LDL ratios (≤10 : 1), the cellular uptake of Hyp is characterized by endocytosis of the [Hyp-LDL] complex, while Hyp alone can enter cells by passive diffusion. Photo-induced cell death and the mitochondrial membrane potential, observed for glioma cells after various times of incubation with the [Hyp-LDL] complex or Hyp alone, were monitored by flow-cytometry analysis using Annexin-V-FITC propidium iodide and DiOC(6)(3) staining. Differences of the results are discussed in view of the respective dynamic subcellular distributions of the drugs that were obtained by co-localization experiments using confocal fluorescence microscopy. In order to give clear evidence of specific intracellular localization and to identify possible Hyp aggregation in cellular organelles, fluorescence resonance energy transfer (FRET) between selected fluorescent organelle probes and Hyp was also assessed. It is shown, that the observed photo-induced cell deaths can be correlated with the sub-cellular distribution of the active fluorescent monomer form of Hyp in lysosomes (as determined from steady-state fluorescence experiments), but that possible aggregation of Hyp in some organelles, as determined from FRET experiments, should be taken into account for interpretation of the real dynamics of the subcellular redistribution. Results of the present study underline the fact that photo-induced cell death processes are strongly influences by dynamics of Hyp subcellular redistribution processes involving monomer-aggregate equilibrium. Such an observation should be taken in consideration for further optimization of Hyp in vivo PDT applications.


Subject(s)
Apoptosis/drug effects , Organelles/metabolism , Perylene/analogs & derivatives , Photosensitizing Agents/toxicity , Anthracenes , Cell Line, Tumor , Endocytosis , Fluorescence Resonance Energy Transfer , Glioma/metabolism , Glioma/pathology , Humans , Lipoproteins, LDL/chemistry , Lipoproteins, LDL/metabolism , Membrane Potential, Mitochondrial/drug effects , Microscopy, Confocal , Perylene/chemistry , Perylene/toxicity , Photosensitizing Agents/chemistry
13.
Photochem Photobiol ; 87(1): 56-63, 2011.
Article in English | MEDLINE | ID: mdl-21114669

ABSTRACT

Steady-state and time-resolved fluorescence spectroscopy have been used for the study of the incorporation kinetics of hypericin (Hyp) into low-density lipoproteins (LDL). Biphasic kinetics of Hyp association with LDL was observed when solutions of Hyp and LDL were mixed at various concentration ratios. The rapid phase of Hyp incorporation is completed within seconds, while the slow phase lasts several minutes. The relative contributions of the individual phases show that a higher amount of Hyp molecules (65%) are incorporated into LDL in the second phase. The kinetics of the incorporation of Hyp into LDL particles preloaded with Hyp (Hyp/LDL=25:1) was also investigated. The decreased intensity of Hyp fluorescence is a sign of the formation of Hyp aggregates after penetration of additional Hyp molecules into Hyp/LDL=25:1 complex. The time dependence of Hyp fluorescence was measured after mixing the complex Hyp/LDL =200:1 with appropriate amounts of free LDL molecules. For each final Hyp/LDL ratio, an increase in the intensity and lifetime of Hyp fluorescence was observed, suggesting a monomerization of Hyp aggregates. The half-time of Hyp transfer from Hyp/LDL complex to LDL particles is similar to the half-time of the slow phase of Hyp incorporation into free LDL particles.


Subject(s)
Lipoproteins, LDL/chemistry , Perylene/analogs & derivatives , Anthracenes , Fluorescence , Kinetics , Perylene/chemistry
14.
PLoS One ; 5(2): e9342, 2010 Feb 22.
Article in English | MEDLINE | ID: mdl-20179769

ABSTRACT

BACKGROUND: The pro-apoptotic effector Bid induces mitochondrial apoptosis in synergy with Bax and Bak. In response to death receptors activation, Bid is cleaved by caspase-8 into its active form, tBid (truncated Bid), which then translocates to the mitochondria to trigger cytochrome c release and subsequent apoptosis. Accumulating evidence now indicate that the binding of tBid initiates an ordered sequences of events that prime mitochondria from the action of Bax and Bak: (1) tBid interacts with mitochondria via a specific binding to cardiolipin (CL) and immediately disturbs mitochondrial structure and function idependently of its BH3 domain; (2) Then, tBid activates through its BH3 domain Bax and/or Bak and induces their subsequent oligomerization in mitochondrial membranes. To date, the underlying mechanism responsible for targeting tBid to mitochondria and disrupting mitochondrial bioenergetics has yet be elucidated. PRINCIPAL FINDINGS: The present study investigates the mechanism by which tBid interacts with mitochondria issued from mouse hepatocytes and perturbs mitochondrial function. We show here that the helix alphaH6 is responsible for targeting tBid to mitochondrial CL and disrupting mitochondrial bioenergetics. In particular, alphaH6 interacts with mitochondria through electrostatic interactions involving the lysines 157 and 158 and induces an inhibition of state-3 respiration and an uncoupling of state-4 respiration. These changes may represent a key event that primes mitochondria for the action of Bax and Bak. In addition, we also demonstrate that tBid required its helix alphaH6 to efficiently induce cytochrome c release and apoptosis. CONCLUSIONS: Our findings provide new insights into the mechanism of action of tBid, and particularly emphasize the importance of the interaction of the helix alphaH6 with CL for both mitochondrial targeting and pro-apoptotic activity of tBid. These support the notion that tBid acts as a bifunctional molecule: first, it binds to mitochondrial CL via its helix alphaH6 and destabilizes mitochondrial structure and function, and then it promotes through its BH3 domain the activation and oligomerization of Bax and/or Bak, leading to cytochrome c release and execution of apoptosis. Our findings also imply an active role of the membrane in modulating the interactions between Bcl-2 proteins that has so far been underestimated.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/metabolism , Cardiolipins/metabolism , Mitochondrial Proteins/metabolism , src Homology Domains , Amino Acid Sequence , Animals , Apoptosis , BH3 Interacting Domain Death Agonist Protein/chemistry , BH3 Interacting Domain Death Agonist Protein/genetics , Cardiolipins/genetics , Cells, Cultured , Cytochromes c/metabolism , Cytoplasm/metabolism , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Molecular Sequence Data , Mutation , Protein Binding , Reactive Oxygen Species/metabolism , Transfection
15.
Int J Pharm ; 389(1-2): 32-40, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20083174

ABSTRACT

The natural photosensitizer hypericin exhibits potent properties for tumor diagnosis and photodynamic therapy. Fluorescent properties of hypericin along with various technical approaches have been used for dynamic studies of its interaction with low-density lipoprotein and U87 glioma cells. Evidences for hypericin release from low-density lipoprotein towards cells plasmatic membrane are addressed. Subsequent subcellular bulk flow redistribution leading to non-specific staining of intracellular membranes compartment were observed within cells. It was shown, that monomers of hypericin are the only redistributive forms. Increasing concentration of hypericin leads to the formation of non-fluorescent aggregates within low-density lipoprotein as well as within the U87 cells, and can preclude its photosensitizing activities. However, the aggregation process can only account for a part of the observed emission decrease. As shown by the excited state lifetime measurements, this fluorescence quenching actually results from a combination of aggregation process and energy transfer from monomers to aggregates. In all experiments, hydrophobic character of hypericin appears as the driving force of its redistribution process.


Subject(s)
Lipoproteins, LDL/metabolism , Perylene/analogs & derivatives , Photosensitizing Agents/chemistry , Anthracenes , Cell Line, Tumor , Cell Membrane/metabolism , Fluorescence , Glioma , Humans , Hydrophobic and Hydrophilic Interactions , Perylene/chemistry , Perylene/pharmacokinetics , Photosensitizing Agents/pharmacokinetics
16.
Ann N Y Acad Sci ; 1130: 117-21, 2008.
Article in English | MEDLINE | ID: mdl-18596340

ABSTRACT

Time-resolved confocal microspectrofluorometry and fluorescence microscopy imaging were applied to monitor the cellular uptake of fluorescent-labeled oligonucleotides (ONs) delivered by a porphyrin molecule. The fate of porphyrin-ON complexes inside living cells has also been monitored. Due to intrinsic fluorescence of the porphyrin and sensitivity of its characteristics to microenvironment, multicomponent analysis of time-resolved fluorescence provides unique information about stability of the porphyrin-ON complexes, ON interactions with their target sequences, and ON and porphyrin distributions after delivery inside the cells. Time-resolved confocal microspectrofluorometry indeed delivers additional information compared with fluorescence confocal microscopy imaging widely employed to study ON uptake.


Subject(s)
Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Oligonucleotides/chemistry , Porphyrins/chemistry , 3T3 Cells , Animals , Cations , Cell Nucleus/metabolism , Drug Delivery Systems , Fluorescence , Melanoma, Experimental , Mice , Microscopy, Confocal/instrumentation , Microscopy, Fluorescence/instrumentation , Oligonucleotides, Antisense/chemistry , Sensitivity and Specificity , Time Factors
17.
Photochem Photobiol ; 84(1): 120-7, 2008.
Article in English | MEDLINE | ID: mdl-18173711

ABSTRACT

The dependence of the uptake of hypericin (Hyp) by human glioma U-87 MG cells on the level of expression of low-density lipoprotein (LDL) receptors has been studied in this work. A special role of the LDL receptor-pathway for Hyp delivery to U-87 MG cells in the presence of LDL was revealed by the substantial increase of Hyp uptake in the situation, when the number of LDL receptors on the cell surface was elevated. Moreover, the colocalization experiments showed the lysosomal localization of Hyp following the uptake and that the concentration of Hyp in these organelles was enhanced in the cells with elevated number of LDL receptors when the incubation medium contained LDL. Both these findings suggest that LDL and LDL receptor-pathway play an important role in the delivery and accumulation of Hyp into the cells.


Subject(s)
Cholesterol, LDL/metabolism , Perylene/analogs & derivatives , Receptors, LDL/metabolism , Anthracenes , Cell Line, Tumor , Humans , Lysosomes , Molecular Structure , Perylene/chemistry , Perylene/metabolism
18.
Photochem Photobiol ; 81(6): 1395-403, 2005.
Article in English | MEDLINE | ID: mdl-15960595

ABSTRACT

By means of UV-VIS absorption and fluorescence spectroscopy, we demonstrate that the photosensitizer hypericin (Hyp) interacts nonspecifically with low-density lipoproteins (LDL), most probably with the lipid fraction of LDL. The molar ratio of monomeric Hyp binding to nonoxidized LDL and mildly oxidized LDL is 30:1. Increasing the Hyp concentration further leads to the formation of Hyp aggregates inside the LDL molecule. We also demonstrate that photoactivated Hyp oxidizes LDL in a light dose and excitation wavelength dependent manner. The level of oxidation of LDL depends on the amount of Hyp inside the LDL molecule. The maximum of the photosensitized oxidation of the LDL by Hyp is achieved for a 30:1 molar ratio, which corresponds to the maximum concentration of monomeric form of Hyp in LDL.


Subject(s)
Lipoproteins, LDL/chemistry , Lipoproteins, LDL/radiation effects , Perylene/analogs & derivatives , Photosensitizing Agents/chemistry , Anthracenes , Binding Sites , Dose-Response Relationship, Radiation , Lipoproteins, LDL/metabolism , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Macromolecular Substances/radiation effects , Molecular Structure , Oxidation-Reduction , Perylene/chemistry , Perylene/metabolism , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet
19.
Biopolymers ; 74(1-2): 110-4, 2004.
Article in English | MEDLINE | ID: mdl-15137106

ABSTRACT

Interaction, i.e., cellular uptake and intracellular distribution, of synthetic modified antisense oligonucleotide with the B16 melanoma cell line was studied using cationic polyene antibiotic, amphotericin B 3-dimethylaminopropyl amide, as a carrier vector. The antisense oligonucleotide--dT(15) oligomer analogue containing isopolar, nonisosteric, phosphonate-based internucleotide linkages 3'-O-P-CH(2)-O-5'--was labeled with fluorescent tetramethylrhodamine marker. The oligonucleotide itinerancy across the cell membrane and its distribution inside the cell was visualized using fluorescence microimaging. During the first several hours a strong preference staining of the cell nucleus was found. Fluorescence lifetime measurements from the intracellular environment (confocal laser microspectrofluorimeter, frequency domain phase/modulation technique in 1 to 200 MHz frequency region) yielded two spectral components of 4.9 and 1.4 ns lifetime, respectively. While the former component correlates with the previously characterized effect of the fluorophore binding to biomolecular targets in membranes and/or cytoplasm, the latter component is newly observed and its possible origin is discussed.


Subject(s)
Oligonucleotides/pharmacokinetics , Spectrometry, Fluorescence/methods , Amphotericin B/pharmacology , Animals , Cations , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , Lasers , Melanoma, Experimental , Mice , Microscopy, Confocal , Oligonucleotides, Antisense/pharmacokinetics
20.
Exp Cell Res ; 289(2): 195-210, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-14499621

ABSTRACT

Cytochrome c release is thought to play an important role in the initiation of apoptosis. The nature of the control exerted by Bcl-2 and Bcl-XL on such a pathway is not precisely known. We addressed this issue by square-wave pulse electroloading of exogenous cytochrome c into Jurkat cells. Three hours after cytochrome c loading into the cells, characteristic phenotypes of apoptosis were observed. However, a significant drop in the mitochondrial membrane potential (Deltapsim) was also observed, while cytochrome c was generally considered to act downstream from the mitochondria. Related to the Deltapsim drop, there was a release of proapoptotic proteins such as AIF and Smac from the mitochondria. This release, as well as NAD(P)H and cardiolipids oxidation, are linked to previous caspase activation. Cytochrome c-linked caspase activation also led to potassium efflux out of the cell. Overexpression of Bcl-2 and Bcl-XL or N-acetyl-DEVD-aldehyde treatment not only prevented the mitochondrial membrane potential decrease, but also protected cells from the apoptosis directly induced by cytochrome c electroloading. Bcl-2 and Bcl-XL protection is based on the inhibition of the caspase-dependent retroactive pathway affecting the mitochondrial compartment.


Subject(s)
Apoptosis/physiology , Cytochrome c Group/metabolism , Mitochondria/enzymology , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis/drug effects , Apoptosis Inducing Factor , Apoptosis Regulatory Proteins , Carrier Proteins/metabolism , Caspase 3 , Caspases/metabolism , Cytochrome c Group/pharmacology , Electroporation/methods , Flavoproteins/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Humans , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...