Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 7(12): e1504727, 2018.
Article in English | MEDLINE | ID: mdl-30524902

ABSTRACT

Pancreatic adenocarcinoma (PAC) has a poor prognosis. One treatment approach, investigated here, is to reinforce antitumor immunity. Dendritic cells (DCs) are essential for the development and regulation of adaptive host immune responses against tumors. A major role for DCs may be as innate tumoricidal effector cells. We explored the efficacy of vaccination with immature (i)DCs, after selecting optimal conditions for generating immunostimulatory iDCs. We used two models, C57BL/6Jrj mice with ectopic tumors induced by the PAC cell line, Panc02, and genetically engineered (KIC) mice developing PAC. Therapeutic iDC-vaccination resulted in a significant reduction in tumor growth in C57BL/6Jrj mice and prolonged survival in KIC mice. Prophylactic iDC-vaccination prevented subcutaneous tumor development. These protective effects were long-lasting in Panc02-induced tumor development, but not in melanoma. iDC-vaccination impacted the immune status of the hosts by greatly increasing the percentage of CD8+ T-cells, and natural killer (NK)1.1+ cells, that express granzyme B associated with Lamp-1 and IFN-γ. Efficacy of iDC-vaccination was CD8+ T-cell-dependent but NK1.1+ cell-independent. We demonstrated the ability of DCs to produce peroxynitrites and to kill tumor cells; this killing activity involved peroxynitrites. Altogether, these findings make killer DCs the pivotal actors in the beneficial clinical outcome that accompanies antitumor immune responses. We asked whether efficacy can be improved by combining DC-vaccination with the FOLFIRINOX regimen. Combined treatment significantly increased the lifespan of KIC mice with PAC. Prolonged treatment with FOLFIRINOX clearly augmented this beneficial effect. Combining iDC-vaccination with FOLFIRINOX may therefore represent a promising therapeutic option for patients with PAC.

2.
Oncotarget ; 9(15): 12513-12533, 2018 Feb 23.
Article in English | MEDLINE | ID: mdl-29552330

ABSTRACT

Pancreatic adenocarcinomas and diabetes mellitus are responsible for the deaths of around two million people each year worldwide. Patients with chronic pancreatitis do not die directly of this disease, except where the pathology is hereditary. Much current literature supports the involvement of bile salt-dependent lipase (BSDL), also known as carboxyl ester lipase (CEL), in the pathophysiology of these pancreatic diseases. The purpose of this review is to shed light on connections between chronic pancreatitis, diabetes, and pancreatic adenocarcinomas by gaining an insight into BSDL and its variants. This enzyme is normally secreted by the exocrine pancreas, and is diverted within the intestinal lumen to participate in the hydrolysis of dietary lipids. However, BSDL is also expressed by other cells and tissues, where it participates in lipid homeostasis. Variants of BSDL resulting from germline and/or somatic mutations (nucleotide insertion/deletion or nonallelic homologous recombination) are expressed in the pancreas of patients with pancreatic pathologies such as chronic pancreatitis, MODY-8, and pancreatic adenocarcinomas. We discuss the possible link between the expression of BSDL variants and these dramatic pancreatic pathologies, putting forward the suggestion that BSDL and its variants are implicated in the cell lipid metabolism/reprogramming that leads to the dyslipidemia observed in chronic pancreatitis, MODY-8, and pancreatic adenocarcinomas. We also propose potential strategies for translation to therapeutic applications.

3.
Oncotarget ; 6(27): 23462-79, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26405163

ABSTRACT

Oncofetal fucose-rich glycovariants of the pathological bile salt-dependent lipase (pBSDL) appear during human pancreatic oncogenesis and are detected by themonoclonal antibody J28 (mAbJ28). We aimed to identify murine counterparts onpancreatic ductal adenocarcinoma (PDAC) cells and tissue and investigate the potential of dendritic cells (DC) loaded with this unique pancreatic tumor antigen to promote immunotherapy in preclinical trials. Pathological BSDLs purified from pancreatic juices of patients with PDAC were cleaved to generate glycosylated C-terminal moieties (C-ter) containing mAbJ28-reactive glycoepitopes. Immunoreactivity of the murine PDAC line Panc02 and tumor tissue to mAbJ28 was detected by immunohistochemistry and flow cytometry. C-ter-J28+ immunization promoted Th1-dominated immune responses. In vitro C-ter-J28+-loaded DCskewed CD3+ T-cells toward Th1 polarization. C-ter-J28+-DC-vaccinations selectively enhanced cell immunoreactivity to Panc02, as demonstrated by CD4+- and CD8+-T-cell activation, increased percentages of CD4+- and CD8+-T-cells and NK1.1+ cells expressing granzyme B, and T-cell cytotoxicity. Prophylactic and therapeutic C-ter-J28+-DC-vaccinations reduced ectopic Panc02-tumor growth, provided long-lasting protection from Panc02-tumor development in 100% of micebut not from melanoma, and attenuated progression of orthotopic tumors as revealed by MRI. Thusmurine DC loaded with pancreatic tumor-specific glycoepitope C-ter-J28+ induce efficient anticancer adaptive immunity and represent a potential adjuvant therapy for patients afflicted with PDAC.


Subject(s)
Cancer Vaccines/genetics , Carcinoma, Pancreatic Ductal/metabolism , Dendritic Cells/metabolism , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Pancreatic Neoplasms/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Biomarkers, Tumor/metabolism , CD3 Complex/metabolism , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Dendritic Cells/cytology , Epitopes/chemistry , Flow Cytometry , Gene Expression Profiling , Glycosylation , Granzymes/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Immunotherapy , Lymphocyte Activation/immunology , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Protein Structure, Tertiary , T-Lymphocytes, Cytotoxic/immunology
4.
J Vis Exp ; (91): 51829, 2014 Sep 21.
Article in English | MEDLINE | ID: mdl-25285979

ABSTRACT

Studies of gene expression on the RNA and protein levels have long been used to explore biological processes underlying disease. More recently, genomics and proteomics have been complemented by comprehensive quantitative analysis of the metabolite pool present in biological systems. This strategy, termed metabolomics, strives to provide a global characterization of the small-molecule complement involved in metabolism. While the genome and the proteome define the tasks cells can perform, the metabolome is part of the actual phenotype. Among the methods currently used in metabolomics, spectroscopic techniques are of special interest because they allow one to simultaneously analyze a large number of metabolites without prior selection for specific biochemical pathways, thus enabling a broad unbiased approach. Here, an optimized experimental protocol for metabolomic analysis by high-resolution NMR spectroscopy is presented, which is the method of choice for efficient quantification of tissue metabolites. Important strengths of this method are (i) the use of crude extracts, without the need to purify the sample and/or separate metabolites; (ii) the intrinsically quantitative nature of NMR, permitting quantitation of all metabolites represented by an NMR spectrum with one reference compound only; and (iii) the nondestructive nature of NMR enabling repeated use of the same sample for multiple measurements. The dynamic range of metabolite concentrations that can be covered is considerable due to the linear response of NMR signals, although metabolites occurring at extremely low concentrations may be difficult to detect. For the least abundant compounds, the highly sensitive mass spectrometry method may be advantageous although this technique requires more intricate sample preparation and quantification procedures than NMR spectroscopy. We present here an NMR protocol adjusted to rat brain analysis; however, the same protocol can be applied to other tissues with minor modifications.


Subject(s)
Brain/metabolism , Metabolomics/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Tissue Extracts/chemistry , Animals , Brain Chemistry , Female , Rats , Rats, Inbred Lew , Tissue Extracts/analysis
5.
PLoS One ; 8(2): e56101, 2013.
Article in English | MEDLINE | ID: mdl-23457507

ABSTRACT

Many diseases, including brain disorders, are associated with perturbations of tissue metabolism. However, an often overlooked issue is the impact that inflammations outside the brain may have on brain metabolism. Our main goal was to study similarities and differences between brain metabolite profiles of animals suffering from experimental autoimmune encephalomyelitis (EAE) and adjuvant arthritis (AA) in Lewis rat models. Our principal objective was the determination of molecular protagonists involved in the metabolism underlying these diseases. EAE was induced by intraplantar injection of complete Freund's adjuvant (CFA) and spinal-cord homogenate (SC-H), whereas AA was induced by CFA only. Naive rats served as controls (n = 9 for each group). Two weeks after inoculation, animals were sacrificed, and brains were removed and processed for metabolomic analysis by NMR spectroscopy or for immunohistochemistry. Interestingly, both inflammatory diseases caused similar, though not identical, changes in metabolites involved in regulation of brain cell size and membrane production: among the osmolytes, taurine and the neuronal marker, N-acetylaspartate, were decreased, and the astrocyte marker, myo-inositol, slightly increased in both inoculated groups compared with controls. Also ethanolamine-containing phospholipids, sources of inflammatory agents, and several glycolytic metabolites were increased in both inoculated groups. By contrast, the amino acids, aspartate and isoleucine, were less concentrated in CFA/SC-H and control vs. CFA rats. Our results suggest that inflammatory brain metabolite profiles may indicate the existence of either cerebral (EAE) or extra-cerebral (AA) inflammation. These inflammatory processes may act through distinct pathways that converge toward similar brain metabolic profiles. Our findings open new avenues for future studies aimed at demonstrating whether brain metabolic effects provoked by AA are pain/stress-mediated and/or due to the presence of systemic proinflammatory molecules. Regardless of the nature of these mechanisms, our findings may be of interest for future clinical studies, e.g. by in-vivo magnetic resonance spectroscopy.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Brain/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Metabolic Networks and Pathways , Animals , Arthritis, Experimental/chemically induced , Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Freund's Adjuvant , Metabolomics , Phospholipids/metabolism , Rats , Rats, Inbred Lew , Spinal Cord/metabolism , Spinal Cord/pathology , Water/metabolism
6.
PLoS One ; 7(10): e47480, 2012.
Article in English | MEDLINE | ID: mdl-23094054

ABSTRACT

Exosomes are of increasing interest as alternative mode of cell-to-cell communication. We previously reported that exosomes secreted by human SOJ-6 pancreatic tumor cells induce (glyco)protein ligand-independent cell death and inhibit Notch-1 pathway, this latter being particularly active during carcinogenesis and in cancer stem cells. Therefore, we asked whether exosomal lipids were key-elements for cell death and hypothesized that cholesterol-rich membrane microdomains were privileged sites of exosome interactions with tumor cells. To address these questions and based on the lipid composition of exosomes from SOJ-6 cells (Ristorcelli et al. (2008) FASEB J. 22; 3358-3369) enriched in cholesterol and sphingomyelin (lipids forming liquid-ordered phase, Lo) and depleted in phospholipids (lipids forming liquid-disordered phase, Ld), we designed Synthetic Exosome-Like Nanoparticles (SELN) with ratios Lo/Ld from 3.0 to 6.0 framing that of SOJ-6 cell exosomes. SELN decreased tumor cell survival, the higher the Lo/Ld ratio, the lower the cell survival. This decreased survival was due to activation of cell death with inhibition of Notch pathway. FRET analyses indicated fusions/exchanges of SELN with cell membranes. Fluorescent SELN co-localized with the ganglioside GM1 then with Rab5A, markers of lipid microdomains and of early endosomes, respectively. These interactions occurred at lipid microdomains of plasma and/or endosome membranes where the Notch-1 pathway matures. We thus demonstrated a major role for lipids in interactions between SELN and tumor cells, and in the ensued cell death. To our knowledge this is the first report on such effects of lipidic nanoparticles on tumor cell behavior. This may have implications in tumor progression.


Subject(s)
Biomimetic Materials/pharmacology , Exosomes/metabolism , Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , Receptor, Notch1/metabolism , Biological Transport , Biomimetic Materials/chemistry , Cell Communication , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cholesterol/chemistry , Cholesterol/pharmacology , Exosomes/chemistry , G(M1) Ganglioside/chemistry , Gene Expression , Humans , Membrane Microdomains/drug effects , Microscopy, Fluorescence , Nanoparticles/ultrastructure , Pancreatic Neoplasms/ultrastructure , Receptor, Notch1/genetics , Signal Transduction , Sphingomyelins/chemistry , Sphingomyelins/pharmacology , rab5 GTP-Binding Proteins/metabolism
7.
J Immunol ; 189(7): 3386-96, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22956586

ABSTRACT

The mAb16D10 was raised against a pathological onco-glycoform of bile salt-dependent lipase isolated from the pancreatic juice of a patient suffering from a pancreatic adenocarcinoma. We previously showed that mAb16D10 specifically discriminates human pancreatic tumor tissues from other cancer and nontumor tissues. In this study, we report that mAb16D10 inhibited the proliferation of only human pancreatic tumor cells expressing 16D10 plasma membrane Ag. Interaction of mAb16D10 with its cognate surface Ag on pancreatic cells promoted cell death by activation of the p53- and caspase-dependent apoptotic pathway, and silencing of p53 decreased cell death. The decreased proliferation was also partly due to cell cycle arrest in G1/S phase, mAb16D10 triggering of glycogen synthase kinase-3ß (GSK-3ß) activation, degradation of ß-catenin, and decreased expression of cyclin D1. GSK-3ß positively affected p53 expression in pancreatic tumor cells after mAb16D10 binding. Inhibition of GSK-3ß activity reversed the effects induced by mAb16D10 in SOJ-6 cells, supporting the pivotal role of GSK-3ß signaling in the mechanisms of action induced by mAb16D10. Also, mAb16D10 cell treatment led to membrane overexpression of E-cadherin. Both E-cadherin and tumor Ag were localized in membrane lipid cholesterol-rich microdomains and are thought to belong to signaling platforms involved in the induction of cell cycle arrest and cell death. Overall, this study reveals that mAb16D10 holds great potential to prevent pancreatic tumor proliferation by apoptotic cell death, thus promising therapeutic prospects for treatment of pancreatic adenocarcinoma, a highly lethal disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/metabolism , Membrane Glycoproteins/metabolism , Molecular Targeted Therapy/methods , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Animals , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Apoptosis/immunology , Cell Death/immunology , Cell Line, Tumor , Humans , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/immunology , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Transplantation, Heterologous
8.
Oncoimmunology ; 1(1): 56-61, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22720212

ABSTRACT

Glycoproteins, as valuable targets for dendritic cell (DC)-vaccination in cancers, remain an open question. Glycosylated structures, which are aberrantly modified during cancerisation, impact positively or negatively on glycoprotein immunogenicity. Here is presented an oncofetal glycovariant of bile-salt-dependent-lipase, expressed on human tumoral pancreas and efficiently processed by DC's, inducing T-lymphocyte activation.

9.
Inflamm Allergy Drug Targets ; 10(5): 322-42, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21745182

ABSTRACT

Traditional healthcare systems in China, India, Greece and the Middle East have for centuries exploited venomous creatures as a resource for medicines. This review focuses on one class of pharmacologically active compounds from venom, namely peptide toxins that target ion channels. We highlight their therapeutic potential and the specific channels they target. The field of therapeutic application is vast, including pain, inflammation, cancer, neurological disorders, cardioprotection, and autoimmune diseases. One of these peptides is in clinical use, and many others are in various stages of pre-clinical and clinical development.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Autoimmune Diseases/drug therapy , Cardiotonic Agents/therapeutic use , Neoplasms/drug therapy , Nervous System Diseases/drug therapy , Spider Venoms/therapeutic use , Toxins, Biological/therapeutic use , Analgesics/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cardiotonic Agents/pharmacology , Clinical Trials as Topic , Humans , Ion Channels/antagonists & inhibitors , Molecular Targeted Therapy , Spider Venoms/pharmacology , Toxins, Biological/pharmacology
10.
J Immunol ; 186(7): 4067-77, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21346236

ABSTRACT

Aberrant glycosylation or overexpression of cell-surface glycosylated tumor-associated Ags (TAA) distinguish neoplastic from normal cells. Interactions of TAA MUC1 and HER2/neu with dendritic cells (DC) preclude efficient processing, which impairs immune responses. It is thus important to define the mechanisms of interactions between DC and glycosylated TAA and their trafficking and processing for further T cell activation. In this work, we study interactions between DC and the oncofetal fucose-rich glycovariants of bile salt-dependent lipase (BSDL), expressed in pancreatic cancer tissues and referred to as pathological BSDL carrying the fucosylated J28 glycotope (pBSDL-J28) because it is characterized by the mAb J28. The expression of pBSDL-J28 was assessed by immunohistochemistry and quantified by confocal microscopy. Nontumoral pancreatic tissues and cells do not express pBSDL-J28. Using multidisciplinary approaches and functional studies, we provide the first evidence, to our knowledge, that this tumoral glycoprotein is rapidly internalized by human DC through macropinocytosis and endocytosis via mannose receptors and then transported to late endosomes for processing. Interestingly, pBSDL-J28 per se induced DC maturation with increased expression of costimulatory and CD83 molecules associated with cytokine secretion (IL-8 and IL-6). Surprisingly, DC retained their full ability to internalize Ags, making this maturation atypical. Finally, the allogeneic pBSDL-J28-treated DC stimulated lymphocyte proliferation. Besides, pulsing DC with pBSDL-J28 C-terminal glycopolypeptide and maturation with CD40L triggered CD4(+) and CD8(+) T cell proliferation. Therefore, interactions of pBSDL-J28, expressed on tumoral pancreatic tissue, with DC may lead to adequate Ag trafficking and processing and result in T cell activation.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Cell Differentiation/immunology , Dendritic Cells/immunology , Endocytosis/immunology , Pancreatic Neoplasms/immunology , Sterol Esterase/metabolism , Antigen Presentation/immunology , Antigens, Neoplasm/physiology , Biomarkers, Tumor/physiology , Coculture Techniques , Dendritic Cells/metabolism , Dendritic Cells/pathology , HEK293 Cells , Humans , Lectins, C-Type/metabolism , Lymphocyte Activation/immunology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Transport/immunology , Receptors, Cell Surface/metabolism , Sterol Esterase/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
11.
Int J Cancer ; 125(5): 1016-26, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19405120

ABSTRACT

We previously reported that exosomal nanoparticles secreted by human pancreatic tumoral cell lines decrease tumoral cell proliferation through the mitochondria-dependent apoptotic pathway, because of activation of pro-apoptotic phosphatase and tensin homolog deleted on chromosome 10 (PTEN) and of glucose synthase kinase-3beta (GSK-3beta). Interactions between exosomal nanoparticles and cells are thought to involve membrane lipid rafts. However, the underlying mechanism is unknown. Here, we report that the interaction of exosomal nanoparticles with pancreatic cancer cells led to decreased expression of hairy and enhancer-of-split homolog-1 (Hes-1), the intranuclear target of Notch-1 signaling pathway, and to activation of the apoptotic pathway after a cell cycle arrest in G(0)G(1) phase. Strikingly, the expression level of Notch-1 pathway components was critical, because exosomal nanoparticles decreased the proliferation of cells in which these partners are either weakly represented, in differentiated adenocarcinoma cells, or inhibited, in poorly differentiated carcinoma cells, by blocking presenilin in the gamma-secretase complex that regulates the Notch-1 pathway. Overexpression of Notch-1 intracellular domain resulted in the reversion of the cell proliferation inhibition promoted by exosomal nanoparticles. Blocking presenilin unexpectedly resulted in activation of PTEN and GSK-3beta. Conversely, inhibiting either PTEN or GSK-3beta increased Hes-1 expression and partially counteracted the inhibition of proliferation promoted by exosomal nanoparticles, highlighting reciprocal regulations between Notch signaling and PTEN/GSK-3beta. We concluded that interactions of exosomal nanoparticles with target cells, at lipid rafts where Notch-1 pathway partners are localized, hampered the functioning of the Notch-1 survival pathway and activated the apoptotic pathway, which determines tumoral cell fate.


Subject(s)
Adenocarcinoma/pathology , Apoptosis , Exosomes/metabolism , Nanoparticles , Pancreatic Neoplasms/pathology , Receptors, Notch/physiology , Adenocarcinoma/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Carbamates/pharmacology , Caspase 3/metabolism , Cell Differentiation , Cell Proliferation , Dipeptides/pharmacology , Flow Cytometry , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mitochondria/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor HES-1 , Tumor Cells, Cultured
12.
FASEB J ; 22(9): 3358-69, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18511551

ABSTRACT

Exosomes are vesicles secreted by most hematopoietic cells on fusion of multivesicular endosomes with the plasma membrane. Many studies have reported that exosomes may also be released by tumor cells. Exosomes are believed to play an antitumor role through immune cells. We asked whether tumor exosomes have biological activities on tumor cells. We report that human pancreatic tumor nanoparticles, exosome-like as characterized by proteomic analyses and rich in lipid rafts, decreased tumor cell proliferation. Nanoparticles increased Bax and decreased Bcl-2 expressions. Caspase-3 and -9 but not caspase-8 inhibitors impaired apoptosis, which implicates the mitochondria apoptotic pathway. The ceramide-sphingomyelin apoptotic pathway was inoperative. Moreover, nanoparticles induced phosphatase and tensin homolog (PTEN) and glycogen synthase kinase (GSK) -3beta activation and decreased pyruvate dehydrogenase activity. In nanoparticle-treated cells, PTEN formed complexes with actin, beta-catenin, and GSK-3beta. Thus, beta-catenin may no longer be available to activate the survival pathway. Nanoparticles triggered the down-regulation of cyclin D1 and poly(ADP-ribose) polymerase. Hence, nanoparticles counteracted the constitutively activated phosphatidylinositol 3-kinase/Akt survival pathway to drive tumor cells toward apoptosis. Our study provides the first evidence of an apoptotic function of tumor-derived nanoparticles on tumor cells. We propose a new role for nanoparticles, i.e., as signal carriers for interaction between cells, which may have implications in physiopathological situations.


Subject(s)
Apoptosis/drug effects , Membrane Microdomains , Nanoparticles , Pancreatic Neoplasms/pathology , Caspase Inhibitors , Cell Line, Tumor , Ceramides/physiology , Endosomes/physiology , Glycogen Synthase Kinase 3/physiology , Glycogen Synthase Kinase 3 beta , Humans , Lipids/analysis , Membrane Microdomains/physiology , Neoplasm Proteins/analysis , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/physiopathology , Phosphatidylinositol 3-Kinases/physiology , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Pyruvate Dehydrogenase Complex/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , bcl-2-Associated X Protein/biosynthesis
13.
Ann Neurol ; 60(5): 586-596, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17044011

ABSTRACT

OBJECTIVE: We asked whether blockade of voltage-gated K+ channel Kv1.1, whose altered axonal localization during myelin insult and remyelination may disturb nerve conduction, treats experimental autoimmune encephalomyelitis (EAE). METHODS: Electrophysiological, cell proliferation, cytokine secretion, immunohistochemical, clinical, brain magnetic resonance imaging, and spectroscopy studies assessed the effects of a selective blocker of Kv1.1, BgK-F6A, on neurons and immune cells in vitro and on EAE-induced neurological deficits and brain lesions in Lewis rats. RESULTS: BgK-F6A increased the frequency of miniature excitatory postsynaptic currents in neurons and did not affect T-cell activation. EAE was characterized by ventriculomegaly, decreased apparent diffusion coefficient, and decreased (phosphocreatine + beta-adenosine triphosphate)/inorganic phosphate ratio. Reduced apparent diffusion coefficient and impaired energy metabolism indicate astrocytic edema. Intracerebroventricularly BgK-F6A-treated rats showed attenuated clinical EAE with unexpectedly reduced ventriculomegaly and preserved apparent diffusion coefficient values and (phosphocreatine + beta-adenosine triphosphate)/inorganic phosphate ratio. Thus, under BgK-F6A treatment, brain damage was dramatically reduced and energy metabolism maintained. INTERPRETATION: Kv1.1 blockade may target neurons and astrocytes, and modulate neuronal activity and neural cell volume, which may partly account for the attenuation of the neurological deficits. We propose that Kv1.1 blockade has a broad therapeutic potential in neuroinflammatory diseases (multiple sclerosis, stroke, and trauma).


Subject(s)
Cnidarian Venoms/pharmacology , Cnidarian Venoms/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Shaker Superfamily of Potassium Channels/antagonists & inhibitors , Shaker Superfamily of Potassium Channels/metabolism , Animals , Astrocytes/drug effects , Astrocytes/pathology , Blood-Brain Barrier/drug effects , Brain Edema/etiology , Brain Edema/metabolism , Brain Edema/pathology , Cell Proliferation/drug effects , Encephalomyelitis, Autoimmune, Experimental/complications , Magnetic Resonance Spectroscopy/methods , Rats , Rats, Inbred Lew
14.
J Biol Chem ; 278(33): 31095-104, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12783861

ABSTRACT

Maurotoxin (MTX) is a 34-residue toxin that has been isolated initially from the venom of the scorpion Scorpio maurus palmatus. It presents a large number of pharmacological targets, including small conductance Ca2+-activated and voltage-gated K+ channels. Contrary to other toxins of the alpha-KTx6 family (Pi1, Pi4, Pi7, and HsTx1), MTX exhibits a unique disulfide bridge organization of the type C1-C5, C2-C6, C3-C4, and C7-C8 (instead of the conventional C1-C5, C2-C6, C3-C7, and C4-C8, herein referred to as Pi1-like) that does not prevent its folding along the classic alpha/beta scaffold of scorpion toxins. Here, we developed an innovative strategy of chemical peptide synthesis to produce an MTX variant (MTXPi1) with a conventional pattern of disulfide bridging without any alteration of the toxin chemical structure. This strategy was used solely to address the impact of half-cystine pairings on MTX structural properties and pharmacology. The data indicate that MTXPi1 displays some marked changes in affinities toward the target K+ channels. Computed docking analyses using molecular models of both MTXPi1 and the various voltage-gated K+ channel subtypes (Shaker B, Kv1.2, and Kv1.3) were found to correlate with MTXPi1 pharmacology. A functional map detailing the interaction between MTXPi1 and Shaker B channel was generated in line with docking experiments.


Subject(s)
Disulfides/chemistry , Scorpion Venoms/chemistry , Scorpion Venoms/toxicity , Scorpions/chemistry , Amino Acid Sequence , Animals , Apamin/metabolism , Apamin/pharmacology , Binding Sites , Binding, Competitive , Iodine Radioisotopes , Membrane Potentials/drug effects , Molecular Sequence Data , Oocytes/physiology , Potassium Channels/chemistry , Potassium Channels/metabolism , Protein Structure, Tertiary , Rats , Scorpion Venoms/metabolism , Sequence Analysis, Protein , Shaker Superfamily of Potassium Channels , Synaptosomes/drug effects , Xenopus
15.
Neuroreport ; 14(3): 317-20, 2003 Mar 03.
Article in English | MEDLINE | ID: mdl-12634475

ABSTRACT

The ability of myelin basic protein (MBP)-reactive T cells to induce conduction failure was investigated and. With the model, somatosensory evoked potentials (SEP) were recorded before and during adoptively transferred experimental autoimmune encephalomyelitis (EAE) in Lewis rats. Maximum amplitude SEP were reached within 15 min of anesthesia. During EAE, the SEP decreased considerably and their onset was delayed. However, the compound action potentials (CAPs) recorded from Lewis rat optic nerves incubated with encephalitogenic T cells were not affected, emphasizing the importance of environmental factors. This study shows that the model described here is an useful means of investigating the neurological disorders associated with EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/physiopathology , Myelin Basic Protein/immunology , Neural Conduction/physiology , T-Lymphocytes/physiology , Action Potentials/physiology , Animals , Cell Line , Evoked Potentials, Somatosensory/physiology , Guinea Pigs , In Vitro Techniques , Optic Nerve/physiology , Rats , Rats, Inbred Lew
16.
FASEB J ; 17(8): 905-7, 2003 May.
Article in English | MEDLINE | ID: mdl-12626426

ABSTRACT

Neuroinflammatory diseases, such as multiple sclerosis (MS), result from aberrant leukocyte traffic into the central nervous system (CNS). To breach the specialized blood-brain barrier, activated leukocytes interact with CNS endothelial cells (EC) and activate a CD54-mediated signaling pathway controlling the Rho GTPase. To function correctly Rho requires posttranslational prenylation, and this can be inhibited by depleting the supply of isoprenoids through inhibition of the cholesterol synthesis pathway with 3-hydroxy-3-methylglutaryl CoA reductase (HMG-CoA reductase) inhibitors (statins). Here we show that treatment of brain EC in vitro with lovastatin inhibits Rho-mediated transendothelial T cell migration. This effect can be reversed by supplementation with mevalonolactone, the downstream product of HMG-CoA reductase, or by ectopic expression of myristoylated Rho, which remains active in the absence of prenylation. In a relapsing-remitting mouse model of MS, lovastatin treatment inhibited leukocyte migration into the CNS and significantly attenuated the development of both acute and relapsing clinical disease. These studies demonstrate that the indirect pharmacological inhibition of Rho proteins in brain EC by statins can inhibit a key stage in the pathogenesis of neuroinflammation, namely leukocyte migration across the blood-brain barrier. These studies demonstrate a novel effect of statins in modulating the immune response in neuroinflammtory diseases and may provide additional rationale for their use in the treatment of MS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Endothelium, Vascular/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Lymphocytes/drug effects , rho GTP-Binding Proteins/metabolism , Animals , Brain/blood supply , Cell Movement/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Lymphocytes/cytology , Mice , Remission Induction , Secondary Prevention
17.
FASEB J ; 16(10): 1185-94, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12153986

ABSTRACT

We have previously shown that the engagement of ICAM-1 on brain endothelial cells (EC) results in the propagation of EC signaling pathways that are necessary for efficient lymphocyte migration across the tight vascular barriers of the brain. Signaling via this receptor alone, however, is unlikely to explain the differential recruitment of leukocytes at different vascular beds. In this study, we investigated the role of EC heterotrimeric G-protein-mediated signaling in supporting transendothelial migration of T lymphocytes. Treatment of brain EC monolayers with pertussis toxin (PTX) resulted in ADP-ribosylation of G-protein alpha subunits and inhibition (>80%) of lymphocyte migration without affecting lymphocyte adhesion. Aortic and high endothelial venule EC treated identically resulted in only partial inhibition of lymphocyte migration (<40%). Expression of ribosylation-resistant (PTX-insensitive) G-protein alpha subunits in brain EC restored their ability to support lymphocyte migration after pretreatment with PTX. Treatment of brain EC with PTX did not inhibit ICAM-1-stimulated tyrosine phosphorylation of focal adhesion kinase, suggesting the effects of PTX in inhibiting EC facilitation of lymphocyte migration are distinct from activation of EC through ICAM-1. We conclude that a heterotrimeric G-protein-mediated signaling pathway in brain EC is essential for efficient transendothelial migration of T lymphocytes into the brain.


Subject(s)
Blood-Brain Barrier/immunology , Cell Movement , Endothelium, Vascular/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , T-Lymphocytes/immunology , ADP-Ribosylation Factors/pharmacology , Animals , Aorta/cytology , Brain/blood supply , Cell Line , Cell Movement/drug effects , Cells, Cultured , Cortactin , Endothelium, Vascular/drug effects , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Heterotrimeric GTP-Binding Proteins/physiology , Humans , Intercellular Adhesion Molecule-1/physiology , Microfilament Proteins/metabolism , Mutation , Pertussis Toxin , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Inbred Lew , Signal Transduction , Virulence Factors, Bordetella/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...