16 results on '"Susanna Carboni"'
Search Results
2. STING Agonist Combined to a Protein-Based Cancer Vaccine Potentiates Peripheral and Intra-Tumoral T Cell Immunity
- Author
-
Matteo Rossi, Susanna Carboni, Wilma Di Berardino-Besson, Erika Riva, Marie-Laure Santiago-Raber, Elodie Belnoue, and Madiha Derouazi
- Subjects
STING agonist ,protein cancer vaccine ,combination immunotherapy ,CD8 T cells functionality ,Th1 CD4 T cells ,tumor microenvironment ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Combining different immunotherapy approaches is currently building the future of immunotherapy, with the view to maximize anti-tumoral efficacy for larger patient population. The KISIMA™ platform allows the development of protein-based cancer vaccines able to induce tumor-specific T cell response resulting in anti-tumoral efficacy in various mouse models. Intra-tumoral administration of stimulator of interferon gene agonists (STINGa) was shown to induce a potent inflammatory response leading to the development of tumor-specific immunity. Here, we explored the efficacy and mechanisms of action of subcutaneous STINGa treatment combined with therapeutic vaccination in various mouse tumor models. This combinatory treatment highly enhanced frequency and effector function of both peripheral and intra-tumoral antigen-specific CD8 T cells, promoting potent IFNγ and TNFα production along with increased cytotoxicity. Moreover, combination therapy favorably modulated the tumor microenvironment by dampening immune-suppressive cells and increasing CD4 T cell infiltration together with their polarization toward Th1 phenotype. Combination with STINGa treatment improved the effect of therapeutic vaccination, resulting in a prolonged control and slower growth of B16-OVA and TC-1 tumors. Altogether, the results presented here highlight the potential of combining STINGa with a therapeutic protein vaccine for cancer treatment.
- Published
- 2021
- Full Text
- View/download PDF
3. 452 Combination treatment using KISIMATM protein-based cancer vaccine and systemic STING agonist results in profound modulation of tumor microenvironment and improved tumor control
- Author
-
Matteo Rossi, Elodie Belnoue, Susanna Carboni, Wilma Besson-Di Berardino, Erika Riva, Marie-Laure Santiago-Raber, and Madiha Derouazi
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2020
- Full Text
- View/download PDF
4. Identification of VHY/Dusp15 as a regulator of oligodendrocyte differentiation through a systematic genomics approach.
- Author
-
Fanny Schmidt, Monique van den Eijnden, Rosanna Pescini Gobert, Gabriela P Saborio, Susanna Carboni, Chantal Alliod, Sandrine Pouly, Susan M Staugaitis, Ranjan Dutta, Bruce Trapp, and Rob Hooft van Huijsduijnen
- Subjects
Medicine ,Science - Abstract
Multiple sclerosis (MS) is a neuroinflammatory disease characterized by a progressive loss of myelin and a failure of oligodendrocyte (OL)-mediated remyelination, particularly in the progressive phases of the disease. An improved understanding of the signaling mechanisms that control differentiation of OL precursors may lead to the identification of new therapeutic targets for remyelination in MS. About 100 mammalian Protein Tyrosine Phosphatases (PTPs) are known, many of which are involved in signaling both in health and disease. We have undertaken a systematic genomic approach to evaluate PTP gene activity in multiple sclerosis autopsies and in related in vivo and in vitro models of the disease. This effort led to the identification of Dusp15/VHY, a PTP previously believed to be expressed only in testis, as being transcriptionally regulated during OL differentiation and in MS lesions. Subsequent RNA interference studies revealed that Dusp15/VHY is a key regulator of OL differentiation. Finally, we identified PDGFR-beta and SNX6 as novel and specific Dusp15 substrates, providing an indication as to how this PTP might exert control over OL differentiation.
- Published
- 2012
- Full Text
- View/download PDF
5. Heterologous Prime-Boost Vaccination with a Peptide-Based Vaccine and Viral Vector Reshapes Dendritic Cell, CD4+ and CD8+ T Cell Phenotypes to Improve the Antitumor Therapeutic Effect
- Author
-
Tamara Hofer, Matteo Rossi, Susanna Carboni, Wilma Di Berardino Besson, Dorothee von Laer, Guido Wollmann, Madiha Derouazi, and Marie-Laure Santiago-Raber
- Subjects
multi-epitope vaccine ,therapeutic prime-boost vaccine ,t-helper 1 cells ,cross-presenting dendritic cells ,cold and hot tumor models ,antigen competition ,Cancer Research ,Oncology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Article - Abstract
Simple Summary Developing new therapeutic cancer vaccines is of paramount importance to counteract tumor escape observed after conventional therapies in certain types of cancer. We have previously shown that the combination of two different vaccine platforms, targeting tumor-specific antigens, resulted in potent immune responses in preclinical models. Here, we show that the heterologous prime-boost combination with a protein vaccine and a viral vector vesicular stomatitis virus immunologically reshapes the immune-excluded TC-1 tumor model as well as the inflamed MC-38 tumor model, leading to beneficial therapeutic efficacy. Furthermore, the treatment with a multi-epitope vaccine allowed us to appreciate the various repartition among three antigen-specific cytotoxic T-cell responses combined with the viral boost. The combination leads to improved efficacy in all animals and highlights the importance of combining tumor epitopes. Our vaccine strategy could be further extended to prophylactic cancer vaccines and beyond, for infectious diseases. Abstract Heterologous prime-boost settings with a protein vaccine and the viral vector vesicular stomatitis virus, both expressing tumor-associated antigens (KISIMA-TAA and VSV-GP-TAA), have been previously shown to generate potent antitumor immunity. In the cold TC-1 model (HPV antigen) and the immune-infiltrate MC-38 model (Adpgk, Reps1 and Rpl18 neo-antigens), we further investigated pivotal immune cells that educate CD8+ T cells. Heterologous prime-boost vaccination induced a superior antitumor response characterized by the increase in number and functionality of antigen-specific CD8+ T cells, recruitment of cross-presenting dendritic cells, and polarization of CD4+ T cells towards an antitumor Th1 phenotype within the tumor and tumor-draining lymph nodes, turning the cold TC-1 tumor into a hot, inflamed tumor. In the inflamed MC-38 tumor model, treatment combination markedly prolonged the overall survival of mice. Treatment with multi-epitope vaccines also induced high frequencies of multiple antigen specificities in the periphery and in the tumor. Prime-boost treatment reduced tumor-infiltrating regulatory CD4+ T cells whilst increasing cross-presenting dendritic cells in tumor-draining lymph nodes. In conclusion, heterologous prime-boost vaccination possesses the ability to induce a potent anti-tumor response in both immune-excluded and immune-infiltrated mouse tumor models. Additionally, this study highlights the design of a multi-epitope vaccine for cancer immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
6. Novel Protein-Based Vaccine Against Self-Antigen Reduces the Formation of Sporadic Colon Adenomas in Mice
- Author
-
Elodie Belnoue, Kristen N. Harvey, Rodrigo T. Macedo, Susanna Carboni, Margie L. Clapper, Harry S. Cooper, Madiha Derouazi, Kimberly B. Colby, Lisa Vanderveer, Alyssa A. Leystra, and Kerry S. Campbell
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,CD3 ,mouse model ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Medicine ,Ascl2 ,biology ,business.industry ,medicine.disease ,T cell response ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Fusion protein ,Tumor antigen ,TLR2 ,030104 developmental biology ,Oncology ,colon cancer ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Cancer vaccine ,business ,cancer vaccine ,humoral response - Abstract
Novel immunopreventive strategies are emerging that show great promise for conferring long-term protection to individuals at high risk of developing colorectal cancer. The KISIMA vaccine platform utilizes a chimeric protein comprising: 1) a selected tumor antigen, 2) a cell-penetrating peptide to improve antigen delivery and epitope presentation, and 3) a TLR2/4 agonist to serve as a self-adjuvant. This study examines the ability of a KISIMA vaccine against achaete-scute family bHLH transcription factor 2 (Ascl2), an early colon cancer antigen, to reduce colon tumor formation by stimulating an anti-tumor immune response. Vaccine administrations were well-tolerated and led to circulating antibodies and antigen-specific T cells in a mouse model of colorectal cancer. To assess preventive efficacy, the vaccine was administered to mice either alone or in combination with the immune checkpoint inhibitor anti-PD-1. When delivered to animals prior to colon tumor formation, the combination strategy significantly reduced the development of colon microadenomas and adenomas, as compared to vehicle-treated controls. This response was accompanied by an increase in the intraepithelial density of CD3+ T lymphocytes. Together, these data indicate that the KISIMA-Ascl2 vaccine shows great potential to be a safe and potent immunopreventive intervention for individuals at high risk of developing colorectal cancer.
- Published
- 2021
7. 452 Combination treatment using KISIMATM protein-based cancer vaccine and systemic STING agonist results in profound modulation of tumor microenvironment and improved tumor control
- Author
-
Madiha Derouazi, Erika Riva, Marie-Laure Santiago-Raber, Wilma Besson-Di Berardino, Elodie Belnoue, Matteo Rossi, and Susanna Carboni
- Subjects
0301 basic medicine ,Tumor microenvironment ,Combination therapy ,business.industry ,medicine.medical_treatment ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Antigen ,Interferon ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Cytotoxic T cell ,Cancer vaccine ,business ,medicine.drug - Abstract
Background KISIMATM platform allows the development of protein-based cancer vaccines able to induce a potent, tumor-specific CD8 and CD4 T cells response. While the cell penetrating peptide and the Anaxa portions confer, respectively, the cell delivery and self-adjuvanticity properties, the multiantigenic domain allows the targeting of different cancer antigens, resulting in anti-tumoral efficacy in different murine models.1 The first clinical candidate developed from KISIMATM is currently tested, together with anti-PD-1 blockade, in a phase I study in metastatic colorectal cancer patients. Stimulator of interferon genes agonists (STINGa) were shown to induce a potent type I interferon response in preclinical studies. The intratumoral administration of STINGa, to promote tumor inflammation, was shown to result in a protective spontaneous immune response in several murine tumor models. However, the encouraging preclinical results are not supported by recent clinical data, challenging the efficacy of unspecific monotherapy.As it is more and more clear that an effective cancer immunotherapy will require the combination of different treatment strategies, we investigate here the efficacy of combining KISIMATM cancer vaccine with STINGa treatment. Methods Mice were vaccinated with subcutaneous (s.c.) injection of KISIMATM vaccine combined with s.c. administration of STINGa. Safety and immunogenicity were assessed by measuring temperature, serum cytokines and the peripheral antigen-specific response. Anti-tumoral efficacy as well as in depth monitoring of TILs and tumor microenvironment modulation were assessed following therapeutic vaccination in a HPV16 E6 and E7 expressing TC-1 cold tumor model. Results Combination treatment was well tolerated and promoted the development of circulating antigen-specific CD8 T cells. In TC-1 tumor bearing mice, KISIMATM therapeutic vaccination resulted in the infiltration of both antigen-specific CD8 and CD4 T cells within the tumor, as well as a switch of tumor associated macrophages polarization toward the more inflammatory type 1. Combination therapy further increased the tumor microenvironment modulation induced by KISIMATM vaccine, promoting the polarization of inflammatory Thelper 1 CD4 T cells and increasing the effector function of antigen-specific CD8 T cells. The profound modulation of the tumor microenvironment induced by combination therapy enhanced the beneficial effect of KISIMATM vaccination, resulting in a prolonged tumor control. Conclusions Combination of KISIMATM cancer vaccine with systemic STINGa treatment induces the development of a potent, tumor-specific immune response resulting in a profound modulation of the TME. As check-point inhibitor (CPI) therapy is ineffective on poorly infiltrated tumors, combination with therapies able to highly enhance tumor infiltration by T cells could expand CPI indications. Ethics Approval The study was approved by the Canton of Geneva Ethic Board, under the license number GE165/19 Reference Belnoue E, et al. Targeting self and neo-epitopes with a modular self-adjuvanting cancer vaccine. JCI Insight 2019. 4:11.
- Published
- 2020
8. Targeting self- and neoepitopes with a modular self-adjuvanting cancer vaccine
- Author
-
Madiha Derouazi, Stefan Stevanovic, Susanna Carboni, Paul R. Walker, Elodie Belnoue, Annika Nelde, Marie-Laure Santiago-Raber, Jean-François Mayol, Eloise Dupuychaffray, and Wilma Di Berardino Besson
- Subjects
CD4-Positive T-Lymphocytes ,medicine.medical_treatment ,Cell-Penetrating Peptides ,Biology ,CD8-Positive T-Lymphocytes ,Major histocompatibility complex ,Cancer Vaccines ,Major Histocompatibility Complex ,Mice ,Immune system ,Lymphocytes, Tumor-Infiltrating ,Antigen ,Adjuvants, Immunologic ,Antigens, Neoplasm ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Antigen-presenting cell ,ddc:616 ,Antigen Presentation ,Immunity, Cellular ,Immunogenicity ,Toll-Like Receptors ,Histocompatibility Antigens Class II ,General Medicine ,Immunotherapy ,Dendritic Cells ,T-Lymphocytes, Helper-Inducer ,Toll-Like Receptor 2 ,Toll-Like Receptor 4 ,Macaca fascicularis ,HEK293 Cells ,Cancer research ,biology.protein ,Cancer vaccine ,Colorectal Neoplasms ,Immunologic Memory ,Research Article ,T-Lymphocytes, Cytotoxic - Abstract
Induction of a potent CD4 and CD8 T-cell response against tumor-specific and tumor-associated antigen is critical for eliminating tumor cells. Recent vaccination strategies have been hampered by an inefficacious and low amplitude immune response. Here we describe a self-adjuvanted chimeric protein vaccine platform to address these challenges, characterized by a multidomain construction incorporating (i) a cell penetrating peptide (CPP) allowing internalization of several multiantigenic Major Histocompatibility Complex (MHC)-restricted peptides within (ii) the multiantigenic domain (Mad) and (iii) a TLR2/4 agonist domain (TLRag). Functionality of the resulting chimeric protein is based on the combined effect of the above-mentioned three different domains for simultaneous activation of antigen presenting cells and antigen cross-presentation, leading to an efficacious multiantigenic and multiallelic cellular immune response. Helper and cytotoxic T-cell responses were observed against model-, neo- and self-antigens, and were highly potent in several murine tumor models. The safety and the immunogenicity of a human vaccine candidate designed for colorectal cancer treatment was demonstrated in a non-human primate model. This newly engineered therapeutic vaccine approach is promising for the treatment of poorly infiltrated tumors that do not respond to currently marketed immunotherapies.
- Published
- 2019
9. Enhancing Antitumor Immune Responses by Optimized Combinations of Cell-penetrating Peptide-based Vaccines and Adjuvants
- Author
-
Madiha Derouazi, Else-Marit Suso-Inderberg, Hubert François Gaertner, Pierre-Yves Dietrich, Paul R. Walker, Wilma Di Berardino-Besson, Sébastien Wälchli, Oliver Hartley, Elodie Belnoue, Fabrice Cerini, Andres M. Salazar, Stéphane König, Susanna Carboni, and Isabelle Dunand-Sauthier
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,medicine.medical_treatment ,Epitopes, T-Lymphocyte ,Cell-Penetrating Peptides ,CD8-Positive T-Lymphocytes ,ddc:616.07 ,Biology ,Cancer Vaccines ,Epitope ,Mice ,03 medical and health sciences ,Transduction (genetics) ,Cross-Priming ,Immune system ,Adjuvants, Immunologic ,Antigen ,In vivo ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Molecular Biology ,ddc:616 ,Pharmacology ,Circular Dichroism ,Histocompatibility Antigens Class I ,Vaccination ,Histocompatibility Antigens Class II ,Dendritic Cells ,ddc:616.8 ,Disease Models, Animal ,Treatment Outcome ,030104 developmental biology ,Immunology ,Trans-Activators ,Cell-penetrating peptide ,Cancer research ,Molecular Medicine ,Female ,Original Article ,Adjuvant - Abstract
Cell penetrating peptides (CPPs) from the protein ZEBRA are promising candidates to exploit in therapeutic cancer vaccines, since they can transport antigenic cargos into dendritic cells and induce tumor-specific T cells. Employing CPPs for a given cancer indication will require engineering to include relevant tumor-associated epitopes, administration with an appropriate adjuvant, and testing for antitumor immunity. We assessed the importance of structural characteristics, efficiency of in vitro transduction of target cells, and choice of adjuvant in inducing the two key elements in antitumor immunity, CD4 and CD8 T cells, as well as control of tumor growth in vivo. Structural characteristics associated with CPP function varied according to CPP truncations and cargo epitope composition, and correlated with in vitro transduction efficiency. However, subsequent in vivo capacity to induce CD4 and CD8 T cells was not always predicted by in vitro results. We determined that the critical parameter for in vivo efficacy using aggressive mouse tumor models was the choice of adjuvant. Optimal pairing of a particular ZEBRA-CPP sequence and antigenic cargo together with adjuvant induced potent antitumor immunity. Our results highlight the irreplaceable role of in vivo testing of novel vaccine constructs together with adjuvants to select combinations for further development.Molecular Therapy (2016); doi:10.1038/mt.2016.134.
- Published
- 2016
- Full Text
- View/download PDF
10. Identification of VHY/Dusp15 as a regulator of oligodendrocyte differentiation through a systematic genomics approach
- Author
-
Susan M. Staugaitis, Fanny Schmidt, Sandrine Pouly, Bruce D. Trapp, Monique van den Eijnden, Chantal Alliod, Rob Hooft van Huijsduijnen, Gabriela Saborio, Ranjan Dutta, Susanna Carboni, and Rosanna Pescini Gobert
- Subjects
Male ,Cellular differentiation ,Regulator ,Gene Expression ,lcsh:Medicine ,Protein tyrosine phosphatase ,Transcriptomes ,Substrate Specificity ,Myelin ,Mice ,0302 clinical medicine ,Cerebellum ,RNA, Small Interfering ,lcsh:Science ,Sorting Nexins ,Cells, Cultured ,0303 health sciences ,Multidisciplinary ,biology ,Brain ,Cell Differentiation ,Genomics ,Middle Aged ,3. Good health ,Cell biology ,Functional Genomics ,Oligodendroglia ,medicine.anatomical_structure ,Neurology ,Spinal Cord ,Gene Knockdown Techniques ,Medicine ,Dual-Specificity Phosphatases ,Female ,Research Article ,Signal Transduction ,Multiple Sclerosis ,Encephalomyelitis, Autoimmune, Experimental ,Biological Data Management ,Autoimmune Diseases ,Molecular Genetics ,Receptor, Platelet-Derived Growth Factor beta ,03 medical and health sciences ,Genome Analysis Tools ,medicine ,Genetics ,Animals ,Humans ,Gene Regulation ,RNA, Messenger ,Remyelination ,Biology ,030304 developmental biology ,Aged ,Computational Neuroscience ,Regulatory Networks ,lcsh:R ,Oligodendrocyte differentiation ,Computational Biology ,Myelin Basic Protein ,Phosphoproteins ,Molecular biology ,Demyelinating Disorders ,Oligodendrocyte ,Myelin basic protein ,Mice, Inbred C57BL ,biology.protein ,Clinical Immunology ,lcsh:Q ,Gene Function ,Genome Expression Analysis ,Transcriptome ,030217 neurology & neurosurgery - Abstract
Multiple sclerosis (MS) is a neuroinflammatory disease characterized by a progressive loss of myelin and a failure of oligodendrocyte (OL)-mediated remyelination, particularly in the progressive phases of the disease. An improved understanding of the signaling mechanisms that control differentiation of OL precursors may lead to the identification of new therapeutic targets for remyelination in MS. About 100 mammalian Protein Tyrosine Phosphatases (PTPs) are known, many of which are involved in signaling both in health and disease. We have undertaken a systematic genomic approach to evaluate PTP gene activity in multiple sclerosis autopsies and in related in vivo and in vitro models of the disease. This effort led to the identification of Dusp15/VHY, a PTP previously believed to be expressed only in testis, as being transcriptionally regulated during OL differentiation and in MS lesions. Subsequent RNA interference studies revealed that Dusp15/VHY is a key regulator of OL differentiation. Finally, we identified PDGFR-beta and SNX6 as novel and specific Dusp15 substrates, providing an indication as to how this PTP might exert control over OL differentiation.
- Published
- 2012
11. A homogeneous 384-well high-throughput binding assay for a TNF receptor using alphascreen technology
- Author
-
Marie Kosco-Vilbois, Claudia Pena Rossi, Susanna Carboni, Claudio Soto, Dominique Perrin, Christèle Frémaux, Alexander Scheer, and Janet Wilson
- Subjects
0301 basic medicine ,Cell signaling ,CD8 Antigens ,Recombinant Fusion Proteins ,Drug Evaluation, Preclinical ,OX40 Ligand ,Computational biology ,Biology ,01 natural sciences ,Biochemistry ,Sensitivity and Specificity ,Receptors, Tumor Necrosis Factor ,Analytical Chemistry ,Protein–protein interaction ,03 medical and health sciences ,Humans ,Dimethyl Sulfoxide ,education ,Receptor ,education.field_of_study ,Membrane Glycoproteins ,Ligand binding assay ,Receptors, OX40 ,Ligand (biochemistry) ,Small molecule ,Fusion protein ,Molecular biology ,0104 chemical sciences ,OX40 ligand ,010404 medicinal & biomolecular chemistry ,030104 developmental biology ,Immunoglobulin G ,Molecular Medicine ,Reagent Kits, Diagnostic ,Peptides ,Biotechnology - Abstract
To take advantage of the growing knowledge of cellular signaling pathways, modern-daydrug discoveryfaces an increasing challenge to develop assays to screen for compounds that modulate protein-protein interactions. One bottleneck in achieving this goal is a lack of suitable and robust assaytechnologies amenable to a high-throughput format. In this report, we describe how we utilized Alphascreen™ technologyto develop a high-throughput assayto monitor ligand binding to a member of the tumor necrosis factor receptor superfamily. We expressed a fusion protein consisting of the extracellular domain of the OX40 receptor with the constant domains of human IgG. In the presence of OX40 ligand, we determined a binding affinityconstant consistent with reported values and optimized the protocol to develop a simple, homogeneous, and sensitive binding assayin a 384-well format. Finally, we assessed if this system could identify small peptides capable of inhibiting the OX40 receptor and ligand interaction. The results showed that the assaywas able to detect such peptides and could be used to launch a high- throughput screening campaign for small molecules able to prevent OX40 receptor activation. (Journal of Biomolecular Screening 2003:522-532)
- Published
- 2003
12. Profound decrement of mesolimbic dopaminergic neuronal activity during ethanol withdrawal syndrome in rats: electrophysiological and biochemical evidence
- Author
-
Marco Diana, Gian Luigi Gessa, Susanna Carboni, Zvani L. Rossetti, and Marco Pistis
- Subjects
Male ,medicine.medical_specialty ,3,4-Dihydroxyphenylacetic acid ,Time Factors ,Tegmentum Mesencephali ,Dopamine ,Action Potentials ,Mesolimbic pathway ,Nucleus accumbens ,Nucleus Accumbens ,Alcohol Withdrawal Delirium ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Internal medicine ,medicine ,Premovement neuronal activity ,Animals ,Neurons ,Analysis of Variance ,Multidisciplinary ,Ethanol ,Dopaminergic ,Homovanillic acid ,Homovanillic Acid ,Rats ,Electrophysiology ,Endocrinology ,chemistry ,3,4-Dihydroxyphenylacetic Acid ,Alcoholic Intoxication ,medicine.drug ,Research Article - Abstract
Activity of the mesolimbic dopaminergic system was investigated in rats withdrawn from chronic ethanol administration by single-cell extracellular recordings from dopaminergic neurons of the ventrotegmental area, coupled with antidromic identification from the nucleus accumbens, and by microdialysis-technique experiments in the nucleus accumbens. Spontaneous firing rates, spikes per burst, and absolute burst firing but not the number of spontaneously active neurons were found drastically reduced; whereas absolute and relative refractory periods increased in rats withdrawn from chronic ethanol treatment as compared with chronic saline-treated controls. Consistently, dopamine outflow in the nucleus accumbens and its acid metabolites were reduced after abruptly stopping chronic ethanol administration. All these changes, as well as ethanol-withdrawal behavioral signs, were reversed by ethanol administration. This reversal suggests that the abrupt cessation of chronic ethanol administration plays a causal role in the reduction of mesolimbic dopaminergic activity seen in the ethanol-withdrawal syndrome. Results indicate that during the ethanol-withdrawal syndrome the mesolimbic dopaminergic system is tonically reduced in activity, as indexed by electrophysiological and biochemical criteria. Considering the role of the mesolimbic dopaminergic system in the reinforcing properties of ethanol, the depressed activity of this system during the ethanol-withdrawal syndrome may be relevant to the dysphoric state associated with ethanol withdrawal in humans.
- Published
- 1993
13. The non-competitive NMDA-receptor antagonist MK-801 prevents the massive release of glutamate and aspartate from rat striatum induced by 1-methyl-4-phenylpyridinium (MPP+)
- Author
-
Susanna Carboni, Franco Melis, Luca Pani, Zvani L. Rossetti, and Maria Hadjiconstantinou
- Subjects
Male ,1-Methyl-4-phenylpyridinium ,1-Methyl-4-phenylpyridinium ion ,Aspartate ,Dopamine ,Glutamate ,Microdialysis ,MK-801 ,N-Methyl-d-aspartate receptor ,medicine.drug_class ,Glutamic Acid ,Striatum ,Pharmacology ,chemistry.chemical_compound ,Glutamates ,medicine ,Neurotoxin ,Animals ,Neurotransmitter ,Aspartic Acid ,Chemistry ,General Neuroscience ,Antagonist ,Glutamate receptor ,Rats, Inbred Strains ,Receptor antagonist ,Corpus Striatum ,Rats ,Kinetics ,Biochemistry ,NMDA receptor ,Dizocilpine Maleate ,medicine.drug - Abstract
The concentrations of dopamine (DA) and of the excitatory amino acids (EAAs) glutamate (Glu) and aspartate (Asp) were measured in dialysates from the striatum of awake rats in order to study the link between the release of DA and of EAAs induced by the infusion of 1-methyl-4-phenylpyridinium ion (MPP + ). DA and EAAs were detected simultaneously by HPLC-EC. The infusion of MPP + at the concentration of 1 mM elevated DA levels in the perfusates, but did not affect EAA release. However, MPP + at 10 mM maximally stimulated Glu and Asp release to 230- and 68-fold of baseline, respectively. In this condition, pretreatment with the N- methyl- d -aspartate (NMDA) receptor antagonist MK-801 (5 mg/kg, i.p.), prevented the MPP + -induced EAA release. In contrast, MK-801 had no effect on DA release induced either by 1 or 10 mM MPP + . These results suggest that MPP + -induced DA and EAA release are independently regulated processes. In addition, the finding that MK-801 inhibits MPP + -induced EAA release suggests that EAAs may act on NMDA receptors to stimulate their own release through a positive-feedback mechanism.
- Published
- 1990
14. Stress increases noradrenaline release in the rat frontal cortex: Prevention by diazepam
- Author
-
Luca Pani, Gian Luigi Gessa, Chiara Portas, Zvani L. Rossetti, and Susanna Carboni
- Subjects
Male ,medicine.medical_specialty ,Frontal cortex ,Microdialysis ,Norepinephrine ,Internal medicine ,medicine ,Diazepam ,Foot-shock ,Noradrenaline ,Animals ,Acute stress ,Chromatography, High Pressure Liquid ,Pharmacology ,Cerebral Cortex ,Electroshock ,Chemistry ,Rats, Inbred Strains ,Rats ,Endocrinology ,Liberation ,Dialysis ,Stress, Psychological ,medicine.drug - Abstract
Foot-shock produced a more than 2-fold increase in noradrenaline (NA) release from the frontal cortex of freely moving rats. The effect of acute stress was almost completely prevented by the administration of diazepam (5 mg/kg i.p.). Diazepam alone inhibited cortical NA release, the maximal inhibition (-57%) being observed 90 min after the injection. Cortical NA release therefore appears to be a reliable index of central noradrenergic activity in response to stressful conditions.
- Published
- 1990
15. Nimodipine inhibits cocaine-induced dopamine release and motor stimulation
- Author
-
Luca Pani, Gian Luigi Gessa, Susanna Carboni, Zvani L. Rossetti, and Alexander Kusmin
- Subjects
Male ,medicine.medical_specialty ,Microdialysis ,Dopamine ,medicine.medical_treatment ,Striatum ,Motor Activity ,Cocaine ,Internal medicine ,medicine ,Animals ,Nimodipine ,Brain Chemistry ,Pharmacology ,business.industry ,Calcium channel ,Antagonist ,Dihydropyridine ,Homovanillic Acid ,Rats, Inbred Strains ,Corpus Striatum ,Rats ,Stimulant ,Endocrinology ,3,4-Dihydroxyphenylacetic Acid ,business ,Dialysis ,Injections, Intraperitoneal ,medicine.drug - Abstract
Dihydropyridine L-type calcium channel antagonist, nimodipine, prevents both the cocaine-induced DA output in the striatum, as measured by brain microdialysis in freely moving rats, and the motor stimulant effect of the drug
- Published
- 1990
16. A Homogeneous 384-Well High-Throughput Binding Assay for a TNF Receptor Using Alphascreen Technology.
- Author
-
Janet Wilson, Claudia Pena Rossi, Susanna Carboni, Christèle Fremaux, Dominique Perrin, Claudio Soto, Marie Kosco-Vilbois, and Alexander Scheer
- Abstract
To take advantage of the growing knowledge of cellular signaling pathways, modern-day drug discovery faces an increasing challenge to develop assays to screen for compounds that modulate protein-protein interactions. One bottleneck in achieving this goal is a lack of suitable and robust assay technologies amenable to a high-throughput format. In this report, we describe how we utilized Alphascreen™ technology to develop a high-throughput assay to monitor ligand binding to a member of the tumor necrosis factor receptor superfamily. We expressed a fusion protein consisting of the extracellular domain of the OX40 receptor with the constant domains of human IgG. In the presence of OX40 ligand, we determined a binding affinity constant consistent with reported values and optimized the protocol to develop a simple, homogeneous, and sensitive binding assay in a 384-well format. Finally, we assessed if this system could identify small peptides capable of inhibiting the OX40 receptor and ligand interaction. The results showed that the assay was able to detect such peptides and could be used to launch a high-throughput screening campaign for small molecules able to prevent OX40 receptor activation. [ABSTRACT FROM AUTHOR]
- Published
- 2003
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.