Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
Pharmaceutics ; 15(9)2023 Aug 31.
Article En | MEDLINE | ID: mdl-37765224

Systemically administered chemotherapy reduces the efficiency of the anticancer agent at the target tumor tissue and results in distributed drug to non-target organs, inducing negative side effects commonly associated with chemotherapy and necessitating repeated administration. Injectable hydrogels present themselves as a potential platform for non-invasive local delivery vehicles that can serve as a slow-releasing drug depot that fills tumor vasculature, tissue, or resection cavities. Herein, we have systematically formulated and tested an injectable shear-thinning hydrogel (STH) with a highly manipulable release profile for delivering doxorubicin, a common chemotherapeutic. By detailed characterization of the STH physical properties and degradation and release dynamics, we selected top candidates for testing in cancer models of increasing biomimicry. Two-dimensional cell culture, tumor-on-a-chip, and small animal models were used to demonstrate the high anticancer potential and reduced systemic toxicity of the STH that exhibits long-term (up to 80 days) doxorubicin release profiles for treatment of breast cancer and glioblastoma. The drug-loaded STH injected into tumor tissue was shown to increase overall survival in breast tumor- and glioblastoma-bearing animal models by 50% for 22 days and 25% for 52 days, respectively, showing high potential for localized, less frequent treatment of oncologic disease with reduced dosage requirements.

2.
Int J Mol Sci ; 22(22)2021 Nov 10.
Article En | MEDLINE | ID: mdl-34830041

Chemoattraction is a normal and essential process, but it can also be involved in tumorigenesis. This phenomenon plays a key role in glioblastoma (GBM). The GBM tumor cells are extremely difficult to eradicate, due to their strong capacity to migrate into the brain parenchyma. Consequently, a complete resection of the tumor is rarely a possibility, and recurrence is inevitable. To overcome this problem, we proposed to exploit this behavior by using three chemoattractants: CXCL10, CCL2 and CCL11, released by a biodegradable hydrogel (GlioGel) to produce a migration of tumor cells toward a therapeutic trap. To investigate this hypothesis, the agarose drop assay was used to test the chemoattraction capacity of these three chemokines on murine F98 and human U87MG cell lines. We then studied the potency of this approach in vivo in the well-established syngeneic F98-Fischer glioma-bearing rat model using GlioGel containing different mixtures of the chemoattractants. In vitro assays resulted in an invasive cell rate 2-fold higher when chemokines were present in the environment. In vivo experiments demonstrated the capacity of these specific chemoattractants to strongly attract neoplastic glioblastoma cells. The use of this strong locomotion ability to our end is a promising avenue in the establishment of a new therapeutic approach in the treatment of primary brain tumors.


Brain Neoplasms , Chemokine CCL11/metabolism , Chemokine CCL2/metabolism , Chemokine CXCL10/metabolism , Glioblastoma , Neoplasm Proteins/metabolism , Neuroglia , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Male , Mice , Neuroglia/metabolism , Neuroglia/pathology , Rats , Rats, Inbred F344
3.
Int J Mol Sci ; 21(14)2020 Jul 09.
Article En | MEDLINE | ID: mdl-32659905

A liposomal formulation of gold nanoparticles (GNPs) and carboplatin, named LipoGold, was produced with the staggered herringbone microfluidic method. The radiosensitizing potential of LipoGold and similar concentrations of non-liposomal GNPs, carboplatin and oxaliplatin was evaluated in vitro with the human colorectal cancer cell line HCT116 in a clonogenic assay. Progression of HCT116 tumor implanted subcutaneously in NU/NU mice was monitored after an irradiation of 10 Gy combined with either LipoGold, GNPs or carboplatin injected directly into the tumor by convection-enhanced delivery. Radiosensitization by GNPs alone or carboplatin alone was observed only at high concentrations of these compounds. Furthermore, low doses of carboplatin alone or a combination of carboplatin and GNPs did not engender radiosensitization. However, the same low doses of carboplatin and GNPs administered simultaneously by encapsulation in liposomal nanocarriers (LipoGold) led to radiosensitization and efficient control of cell proliferation. Our study shows that the radiosensitizing effect of a combination of carboplatin and GNPs is remarkably more efficient when both compounds are simultaneously delivered to the tumor cells using a liposomal carrier.


Colorectal Neoplasms/therapy , Gold/administration & dosage , Liposomes/administration & dosage , Metal Nanoparticles/administration & dosage , Organoplatinum Compounds/pharmacology , Radiation-Sensitizing Agents/administration & dosage , Animals , Carboplatin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemoradiotherapy/methods , Drug Carriers/administration & dosage , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Oxaliplatin/pharmacology , Xenograft Model Antitumor Assays/methods
4.
J Nanobiotechnology ; 16(1): 77, 2018 Oct 05.
Article En | MEDLINE | ID: mdl-30290821

BACKGROUND: Effectiveness of chemotherapy for treating glioblastoma (GBM) brain tumors is hampered by the blood-brain barrier which limits the entry into the brain of most drugs from the blood. To bypass this barrier, convection-enhanced delivery (CED) was proposed to directly inject drugs in tumor. However, the benefit of CED may be hampered when drugs diffuse outside the tumor to then induce neurotoxicity. Encapsulation of drugs into liposome aims at increasing tumor cells specificity and reduces neurotoxicity. However, the most appropriate liposomal formulation to inject drugs into brain tumor by CED still remains to be determined. In this study, four liposomal carboplatin formulations were prepared and tested in vitro on F98 glioma cells and in Fischer rats carrying F98 tumor implanted in the brain. Impact of pegylation on liposomal surface and relevance of positive or negative charge were assessed. RESULTS: The cationic non-pegylated (L1) and pegylated (L2) liposomes greatly improved the toxicity of carboplatin in vitro compared to free carboplatin, whereas only a modest improvement and even a reduction of efficiency were measured with the anionic non-pegylated (L3) and the pegylated (L4) liposomes. Conversely, only the L4 liposome significantly increased the median survival time of Fisher rats implanted with the F98 tumor, compared to free carboplatin. Neurotoxicity assays performed with the empty L4' liposome showed that the lipid components of L4 were not toxic. These results suggest that the positive charge on liposomes L1 and L2, which is known to promote binding to cell membrane, facilitates carboplatin accumulation in cancer cells explaining their higher efficacy in vitro. Conversely, negatively charged and pegylated liposome (L4) seems to diffuse over a larger distance in the tumor, and consequently significantly increased the median survival time of the animals. CONCLUSIONS: Selection of the best liposomal formulation based on in vitro studies or animal model can result in contradictory conclusions. The negatively charged and pegylated liposome (L4) which was the less efficient formulation in vitro showed the best therapeutic effect in animal model of GBM. These results support that relevant animal model of GBM must be considered to determine the optimal physicochemical properties of liposomal formulations.


Carboplatin/administration & dosage , Carboplatin/therapeutic use , Convection , Drug Delivery Systems , Glioma/drug therapy , Injections , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Survival , Glioma/pathology , Kaplan-Meier Estimate , Lethal Dose 50 , Liposomes/ultrastructure , Rats, Inbred F344
5.
Radiat Res ; 188(6): 605-614, 2017 12.
Article En | MEDLINE | ID: mdl-28956695

Radiation stimulates the expression of inflammatory mediators known to increase cancer cell invasion. Therefore, it is important to determine whether anti-inflammatory drugs can prevent this adverse effect of radiation. Since cyclooxygenase-2 (COX-2) is a central player in the inflammatory response, we performed studies to determine whether the COX-2 inhibitor NS-398 can reduce the radiation enhancement of cancer cell invasion. Thighs of Balb/c mice treated with NS-398 were irradiated with either daily fractions of 7.5 Gy for five consecutive days or a single 30 Gy dose prior to subcutaneous injection of nonirradiated MC7-L1 mammary cancer cells. Five weeks later, tumor invasion, blood vessel permeability and interstitial volumes were assessed using magnetic resonance imaging (MRI). Matrix metalloproteinase-2 (MMP-2) was measured in tissues by zymography at 21 days postirradiation. Cancer cell invasion in the mouse thighs was increased by 12-fold after fractionated irradiations (5 × 7.5 Gy) and by 17-fold after a single 30 Gy dose of radiation. This stimulation of cancer cell invasion was accompanied by a significant increase in the interstitial volume and a higher level of the protease MMP-2. NS-398 treatment largely prevented the stimulation of cancer cell invasion, which was associated with a reduction in interstitial volume in the irradiated thighs and a complete suppression of MMP-2 stimulation. In conclusion, this animal model using MC7-L1 cells demonstrates that radiation-induced cancer cell invasion can be largely prevented with the COX-2 inhibitor NS-398.


Gamma Rays/adverse effects , Mammary Neoplasms, Experimental/pathology , Neoplasm Invasiveness/prevention & control , Nitrobenzenes/therapeutic use , Sulfonamides/therapeutic use , Animals , Cell Line, Tumor , Cell Movement/radiation effects , Cyclooxygenase 2 Inhibitors/pharmacology , Drug Screening Assays, Antitumor , Inflammation Mediators/metabolism , Mammary Neoplasms, Experimental/blood supply , Matrix Metalloproteinase 2/analysis , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/analysis , Neoplasm Transplantation , Nitrobenzenes/pharmacology , Radiotherapy/adverse effects , Sulfonamides/pharmacology , Thigh , Transplantation, Heterotopic , Tumor Burden/drug effects , Tumor Burden/radiation effects
6.
Int J Radiat Biol ; 92(8): 444-50, 2016 08.
Article En | MEDLINE | ID: mdl-27121902

PURPOSE: Irradiation of brain stimulates the expression of inflammatory mediators, some of which can modify the ability of cancer cells to infiltrate the brain. In the present study, the time window during which this stimulation occurs was determined. MATERIALS AND METHODS: Brain of Fischer rat was irradiated (15 Gy) and expression of pro-inflammatory mediators IL-1ß, IL-6 and TNF-α was measured from 4 h to 20 days post-irradiation. Level of the matrix metalloproteinase 2 (MMP-2) and prostaglandin E2 (PGE2) which can favor cancer cell infiltration were also measured. The F98 glioma cells were implanted either during (4 h post-irradiation) or after (10 days post-irradiation) the pro-inflammatory phase. Infiltration distance of F98 cells in brain parenchyma and the median survival time of the animals were determined. RESULTS: Expression of IL-1ß, IL-6 and TNF-α was significantly increased in the irradiated brains with a peak at 4 h post-irradiation. Implantation of F98 glioma cells 4 h post-irradiation reduced the median survival time of Fischer rats to 18 days, compared to 25 days when the F98 were implanted in non-irradiated brain. Irradiation of the brain increased the distance of infiltration of F98 cells and was associated with increased levels of MMP-2 and PGE2. Conversely, F98 cells implanted 10 days post-irradiation have infiltrated the brain over a shorter distance and the median survival time of rats was increased to 35 days. CONCLUSIONS: Cancer recurrence is frequently observed in GBM patients. A better understanding of the inflammatory response observed in irradiated brain could contribute to develop new therapeutic modalities to further increase the efficiency of radiotherapy.


Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cranial Irradiation/adverse effects , Glioma/pathology , Glioma/radiotherapy , Neoplasms, Radiation-Induced/pathology , Animals , Brain Neoplasms/immunology , Cell Line, Tumor , Dose-Response Relationship, Radiation , Glioma/immunology , Male , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology , Neoplasms, Radiation-Induced/etiology , Neoplasms, Radiation-Induced/immunology , Radiotherapy Dosage , Rats , Rats, Inbred F344 , Survival Rate , Treatment Outcome
7.
Med Phys ; 42(5): 2311-6, 2015 May.
Article En | MEDLINE | ID: mdl-25979025

PURPOSE: Targeted and whole-brain irradiation in humans can result in significant side effects causing decreased patient quality of life. To adequately investigate structural and functional alterations after stereotactic radiosurgery, preclinical studies are needed. The purpose of this work is to establish a robust standardized method of targeted irradiation on small regions of the rat brain. METHODS: Euthanized male Fischer rats were imaged in a stereotactic bed, by computed tomography (CT), to estimate positioning variations relative to the bregma skull reference point. Using a rat brain atlas and the stereotactic bregma coordinates obtained from CT images, different regions of the brain were delimited and a treatment plan was generated. A single isocenter treatment plan delivering ≥ 100 Gy in 100% of the target volume was produced by Leksell GammaPlan using the 4 mm diameter collimator of sectors 4, 5, 7, and 8 of the Gamma Knife unit. Impact of positioning deviations of the rat brain on dose deposition was simulated by GammaPlan and validated with dosimetric measurements. RESULTS: The authors' results showed that 90% of the target volume received 100 ± 8 Gy and the maximum of deposited dose was 125 ± 0.7 Gy, which corresponds to an excellent relative standard deviation of 0.6%. This dose deposition calculated with GammaPlan was validated with dosimetric films resulting in a dose-profile agreement within 5%, both in X- and Z-axes. CONCLUSIONS: The authors' results demonstrate the feasibility of standardizing the irradiation procedure of a small volume in the rat brain using a Gamma Knife.


Brain/surgery , Radiometry/methods , Radiosurgery/methods , Animals , Atlases as Topic , Brain/diagnostic imaging , Brain/radiation effects , Calibration , Feasibility Studies , Male , Models, Animal , Patient Positioning , Radiotherapy Dosage , Radiotherapy Planning, Computer-Assisted/methods , Rats, Inbred F344 , Tomography, X-Ray Computed
8.
Int J Radiat Biol ; 91(8): 624-33, 2015 Aug.
Article En | MEDLINE | ID: mdl-25912457

PURPOSE: Radiation induces a neuro-inflammation that is characterized by the expression of genes known to increase the invasion of cancer cells. In Fischer rats, brain irradiation increases the infiltration of cancer cells and reduced the median survival of the animals. In this study, we have determined whether these adverse effects of radiation can be prevented with the cyclooxygenase-2 (COX-2) inhibitor meloxicam. MATERIALS AND METHODS: Brain of Fischer rats treated or not with meloxicam were irradiated (15 Gy) and then implanted with the F98 glioma cells. The median survival of the animals, the infiltration of F98 cells, and the expression of inflammatory cytokines and pro-migration molecules were measured. RESULTS: Meloxicam reduced by 75% the production of prostaglandin E2 (bioproduct of COX-2) in irradiated brains validating its anti-inflammatory effect. Median survival was increased to control levels by the treatment of meloxicam following brain irradiation. This protective effect was associated with a reduction of the infiltration of F98 cells in the brain, a complete inhibition of radiation-enhancement of matrix metalloproteinase-2, and a significant reduction of tumor necrosis factor α (TNF-α) and tumor growth factor ß1 (TGF-ß1) expression. Using invasion chambers, interleukin-1ß (IL-1ß) stimulated by 5-fold the invasiveness of F98 cells, but this stimulation was completely inhibited by meloxicam. This suggests that a cooperation between IL-1ß and COX-2 are involved in radiation-enhancement of F98 cell invasion. CONCLUSIONS: Our results indicate the importance of reducing the inflammatory response of normal brain tissue following irradiation in an effort to extend median survival in F98 tumor-bearing rats.


Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cranial Irradiation/methods , Glioma/pathology , Glioma/therapy , Thiazines/administration & dosage , Thiazoles/administration & dosage , Animals , Cell Line, Tumor , Combined Modality Therapy , Cyclooxygenase 2 Inhibitors/administration & dosage , Male , Meloxicam , Mice , Neoplasm Invasiveness , Radiation Tolerance/drug effects , Rats , Rats, Inbred F344 , Treatment Outcome
9.
J Neurooncol ; 115(3): 365-73, 2013 Dec.
Article En | MEDLINE | ID: mdl-24026531

Treatment of glioblastoma with platinum compounds modestly improves progression-free survival and may cause toxic effects which prevent use at higher dose that would otherwise improve the antineoplastic effect. To reduce toxicity, we propose to encapsulate the platinum drug in a liposome. We have also tested three methods of drug administration (intra-venous, intra-arterial and intra-arterial combined with blood brain barrier disruption) to determine which one optimizes the tumor cell uptake, limits the toxicity and delivers the best concomitance effect with radiotherapy. Cisplatin, oxaliplatin, their respective liposomal formulations, Lipoplatin™ and Lipoxal™, and carboplatin were assessed in F98 glioma, orthotopically implanted in Fischer rats. We found that the modest accumulation of drugs in tumor cells after intra-venous injection was significantly improved when the intra-arterial route was used and further increased after the transient opening of the blood brain barrier with mannitol. The liposomal formulations have largely reduced the toxicity and have allowed a better exploitation of the anti-cancer activity of platinum agent. Although the liposomes Lipoplatin™ and Lipoxal™ have shown a similar ability to that of carboplatin, to accumulate in brain tumors, the highest additive effect with radiotherapy was obtained with carboplatin. We conclude that the intra-arterial infusion of carboplatin or Lipoxal™ in concomitance with radiation therapy leads to the best tumor control as measured by an increase of mean survival time in Fischer rats implanted with the F98 glioma with a benefit in survival time of 13.4 and 6.5 days respectively compared to intra-venous.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/therapy , Chemoradiotherapy , Glioblastoma/therapy , Animals , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Carboplatin/administration & dosage , Cisplatin/administration & dosage , Drug Administration Routes , Glioblastoma/mortality , Glioblastoma/pathology , Liposomes , Male , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Rats , Rats, Inbred F344 , Survival Rate
10.
Int J Radiat Oncol Biol Phys ; 84(1): 244-9, 2012 Sep 01.
Article En | MEDLINE | ID: mdl-22284691

PURPOSE: Treatments of glioblastoma with cisplatin or oxaliplatin only marginally improve the overall survival of patients and cause important side effects. To prevent adverse effects, improve delivery, and optimize the tumor response to treatment in combination with radiotherapy, a potential approach consists of incorporating the platinum agent in a liposome. METHODS AND MATERIALS: In this study, cisplatin, oxaliplatin, carboplatin, Lipoplatin (the liposomal formulation of cisplatin), and Lipoxal (the liposomal formulation of oxaliplatin) were tested on F98 glioma orthotopically implanted in Fischer rats. The platinum compounds were administered by intracarotid infusion and were assessed for the ability to reduce toxicity, improve cancer cell uptake, and increase survival of animals when combined or not combined with radiotherapy. RESULTS: The tumor uptake was 2.4-fold more important for Lipoxal than the liposome-free oxaliplatin. Lipoxal also improved the specificity of oxaliplatin as shown by a higher ratio of tumor to right hemisphere uptake. Surprisingly, Lipoplatin led to lower tumor uptake compared with cisplatin. However, Lipoplatin had the advantage of largely reducing the toxicity of cisplatin and allowed us to capitalize on the anticancer activity of this agent. CONCLUSION: Among the five platinum compounds tested, carboplatin showed the best increase in survival when combined with radiation for treatment of glioma implanted in Fischer rats.


Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Brain Neoplasms/metabolism , Brain Neoplasms/surgery , Carboplatin/administration & dosage , Carboplatin/adverse effects , Carboplatin/pharmacokinetics , Carotid Arteries , Cisplatin/administration & dosage , Cisplatin/adverse effects , Cisplatin/pharmacokinetics , Combined Modality Therapy/methods , Glioblastoma/metabolism , Glioblastoma/surgery , Infusions, Intra-Arterial/methods , Liposomes , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/pharmacokinetics , Oxaliplatin , Radiosurgery/methods , Rats , Rats, Inbred F344
11.
J Neurooncol ; 97(2): 187-93, 2010 Apr.
Article En | MEDLINE | ID: mdl-19760366

Despite significant advances, the radiotherapy and chemotherapy protocols marginally improve the overall survival of patients with glioblastoma. Lipoplatin(TM), and Lipoxal(TM), the liposomal formulations of cisplatin and oxaliplatin respectively, were tested on the F98 glioma cells for their ability to improve the cell uptake and increase the synergic effect when combined with ionizing radiation. The cytotoxicity and synergic effect of platinum compounds were assessed by colony formation assay, while the cellular uptake was measured by Inductively Coupled Plasma Mass Spectrometer (ICP-MS). After 4 h exposure with platinum compounds, cells were irradiated (1.5-6.6 Gy) with a (60)Co source. The liposomal formulations were compared to their liposome-free analogs and to carboplatin. The concomitant treatment of F98 cells with carboplatin and radiation produced the highest radiosensitizing effect (30-fold increase). Among the platinum compounds tested, Lipoplatin(TM) produced the most promising results. This liposomal formulation of cisplatin improved the cell uptake by 3-fold, and its radiosensitizing potential was enhanced by 14-fold. Although Lipoxal(TM) can potentially reduce the adverse effect of oxaliplatin, a synergic effect with radiation was measured only when incubated at a concentration higher than its IC50. Conversely, concomitant treatment with cisplatin did not result in a synergic effect, as in fact a radioprotective effect was measured on the F98 cells. In conclusion, among the five platinum compounds tested, carboplatin and Lipoplatin(TM) showed the best radiosensitizing effect. Lipoplatin(TM) seems the most promising since it led to the best cellular incorporation and has already been reported to be less neurotoxic than other platinum compounds.


Cisplatin/pharmacology , Glioma/drug therapy , Glioma/radiotherapy , Radiation-Sensitizing Agents/administration & dosage , Radiotherapy , Animals , Antineoplastic Agents/administration & dosage , Carboplatin/administration & dosage , Cell Line, Tumor , Combined Modality Therapy , Inhibitory Concentration 50 , Liposomes , Radiation, Ionizing , Rats
12.
Acta Neurochir (Wien) ; 151(6): 677-83; discussion 683-4, 2009 Jun.
Article En | MEDLINE | ID: mdl-19377849

PURPOSE: Accurate targeting is crucial for the irradiation of a small-volume animal brain. We propose an original method, based on a polymer gel, to determine the accuracy and reproducibility of irradiation using a new stereotactic frame. METHODS: An in-house designed rat stereotactic frame, compatible with the Gamma Knife, was constructed. The rat brain was then removed through a small burr hole and the intracranial cavity was washed and filled with a polymer gel. This "gel brain" was irradiated by Gamma Knife and the irradiated volumes and coordinates were measured after the irradiation. RESULTS: The position of the polymerized areas revealed that the stereotactic frame was able to accurately reproduce the same position of irradiation in each animal. The small standard deviation demonstrated the high reproducibility. CONCLUSION: The polymer gel confirmed the ability of the rat stereotactic frame to accurately and reproducibly position a small animal for precise radiosurgery procedures.


Brain Neoplasms/surgery , Gels , Glioma/surgery , Radiosurgery/methods , Skull/surgery , Stereotaxic Techniques/instrumentation , Animals , Brain Tissue Transplantation/methods , Cell Line, Tumor/transplantation , Disease Models, Animal , Gels/radiation effects , Preoperative Care/instrumentation , Preoperative Care/methods , Radiosurgery/instrumentation , Rats , Rats, Inbred F344 , Reproducibility of Results , Skull/anatomy & histology , Surgical Instruments/trends
13.
Biopolymers ; 81(3): 202-14, 2006 Feb 15.
Article En | MEDLINE | ID: mdl-16245262

We present a minimalist approach for the modeling of the three-dimensional structure of multistranded alpha-helical coiled coils. The approach is based on empirical principles introduced by F. H. C. Crick (F. H. C. Crick, Acta Crystallogr, 1953, Vol. 6, pp. 689-697). Crick hypothesized that keeping the distance between the residues at the interacting interface of alpha-helices constant would lead to supercoiling or the formation of a coiled coil through the knobs-into-holes mode of packing. We have implemented the latter hypothesis in a simulating annealing protocol in the simple form of interhelical distance restraints (two per heptad) between Calpha at the interfacial positions and. To demonstrate the authenticity of Crick's hypothesis and the precision and accuracy of our approach, we have modeled the crystal structures of six synthetic coiled coils in dimeric, trimeric, and tetrameric states. The mean root mean square deviations (RMSDs) between the backbone atoms of the ensemble of structures calculated and those of the corresponding geometric averages is always below 0.76 A, indicating that our protocol has an excellent degree of convergence and precision. The RMSDs between the backbone atoms of each of the six geometric average structures and the backbone of the corresponding crystal structures all range between 0.43 and 0.95 A, indicative of excellent accuracy and proving the authenticity of Crick's hypothesis. Moreover, without specifying any dihedral angles, we found that in 81% of the occurrences, the most populated conformer of the side chains at positions and in the ensembles calculated were identical to those observed in the crystal structures. This shows that our simple approach, which is the simplest reported so far, can generate accurate results for the backbone and side chains. Finally, as a test case for a wider application of our approach in the field of structural proteomics, we describe the successful modeling of the overall structure of SNARE and the organization of its interfacial ionic layer known to play an important functional role.


Protein Structure, Secondary , SNARE Proteins/chemistry , Amino Acid Sequence , Biopolymers/chemistry , Models, Molecular , Molecular Sequence Data , Protein Conformation
14.
J Biol Chem ; 279(22): 23463-71, 2004 May 28.
Article En | MEDLINE | ID: mdl-15033973

Hsp27 is expressed at high levels after mild heat shock and contributes to making cells extremely resistant to subsequent treatments. The activity of the protein is regulated at the transcriptional level, but also by phosphorylation, which occurs rapidly during stress and is responsible for causing the dissociation of large 700-kDa Hsp27 oligomers into dimers. We investigated the mechanism by which phosphorylation and oligomerization modulate the protective activity of Chinese hamster Hsp27. In contrast to oligomer dissociation, which only required Ser90 phosphorylation, activation of Hsp27 thermoprotective activity required the phosphorylation of both Ser90 and Ser15. Replacement of Ser90 by Ala90, which prevented the dissociation of the oligomer upon stress, did cause a severe defect in the protective activity. Dissociation was, however, not a sufficient condition to activate the protein because replacement of Ser15 by Ala15, which caused little effect in the oligomeric organization of the protein, also yielded an inactive protein. Analyzes of mutants with short deletions in the NH2 terminus identified the Hsp27 WD/EPF or PF-rich domain as essential for protection, maintenance of the oligomeric structure, and in vitro chaperone activity of the protein. In light of a three-dimensional model of Hsp27 based on the crystallographic structure of wheat Hsp16.9, we propose that the conserved WD/EPF motif of mammalian Hsp27 mediates important intramolecular interactions with hydrophic surfaces of the alpha-crystallin domain of the protein. These interactions are destabilized by Ser90 phosphorylation, making the motif free to interact with heterologous molecular targets upon the additional phosphorylation of the nearby Ser15.


Heat-Shock Proteins/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Cricetinae , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Phosphorylation , Sequence Alignment , Structure-Activity Relationship , Temperature
15.
Biochem J ; 379(Pt 3): 757-63, 2004 May 01.
Article En | MEDLINE | ID: mdl-14741044

The pro-region of the subtilisin-like convertase furin acts early in the biosynthetic pathway as an intramolecular chaperone to enable proper folding of the zymogen, and later on as an inhibitor to constrain the activity of the enzyme until it reaches the trans -Golgi network. To identify residues that are important for pro-region function, we initially identified amino acids that are conserved among the pro-regions of various mammalian convertases. Site-directed mutagenesis of 17 selected amino acids within the 89-residue pro-region and biosynthetic labelling revealed that I60A-furin and H66A-furin were rapidly degraded in a proteasome-dependent manner, while W34A-furin and F67A-furin did not show any autocatalytic activation. Intriguingly, the latter mutants proteolytically cleaved pro-von Willebrand factor precursor to the mature polypeptide, suggesting that the mutations permitted proper folding, but did not allow the pro-region to exercise its role in inhibiting the enzyme. Homology modelling of furin's pro-region revealed that residues Ile-60 and His-66 might be crucial in forming the binding interface with the catalytic domain, while residues Trp-34 and Phe-67 might be involved in maintaining a hydrophobic core within the pro-region itself. These results provide structural insights into the dual role of furin's pro-region.


Furin/chemistry , Furin/metabolism , Protein Folding , Amino Acid Sequence , Catalytic Domain , Cell Line , Enzyme Activation , Enzyme Precursors/chemistry , Enzyme Precursors/genetics , Enzyme Precursors/metabolism , Furin/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Protein Precursors/chemistry , Protein Precursors/metabolism , Sequence Alignment , Structural Homology, Protein , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism
16.
FEBS Lett ; 527(1-3): 309-14, 2002 Sep 11.
Article En | MEDLINE | ID: mdl-12220680

Furin, a member of the subtilisin-like pro-protein convertase family, is a type I membrane protein that undergoes ectodomain shedding. Metabolic labeling of cells stably expressing furin demonstrated that the shed form of furin is detected after 30 min. Moreover, sequence analysis revealed that specific residues of the cysteine-rich region of furin aligned with those of tumor necrosis factor receptor, which is also shed. Introduction within furin's cysteine-rich region of mutations that impair TNFR1 shedding also abolished furin shedding. Our results show that shedding of furin occurs rapidly and further suggest that specific cysteine residues may impart a conformation to the enzyme, thereby affecting its susceptibility to proteolysis.


Cysteine/chemistry , Subtilisins/chemistry , Subtilisins/metabolism , Amino Acid Sequence , Antigens, CD/chemistry , Base Sequence , Cells, Cultured , Furin , Humans , Kinetics , Molecular Sequence Data , Point Mutation , Protein Conformation , Protein Structure, Tertiary , Receptors, Tumor Necrosis Factor/chemistry , Receptors, Tumor Necrosis Factor, Type I , Sequence Alignment , Subtilisins/genetics
...