Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 4096, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38750019

ABSTRACT

The presence of heterogeneity in responses to oncolytic virotherapy poses a barrier to clinical effectiveness, as resistance to this treatment can occur through the inhibition of viral spread within the tumor, potentially leading to treatment failures. Here we show that 4-octyl itaconate (4-OI), a chemical derivative of the Krebs cycle-derived metabolite itaconate, enhances oncolytic virotherapy with VSVΔ51 in various models including human and murine resistant cancer cell lines, three-dimensional (3D) patient-derived colon tumoroids and organotypic brain tumor slices. Furthermore, 4-OI in combination with VSVΔ51 improves therapeutic outcomes in a resistant murine colon tumor model. Mechanistically, we find that 4-OI suppresses antiviral immunity in cancer cells through the modification of cysteine residues in MAVS and IKKß independently of the NRF2/KEAP1 axis. We propose that the combination of a metabolite-derived drug with an oncolytic virus agent can greatly improve anticancer therapeutic outcomes by direct interference with the type I IFN and NF-κB-mediated antiviral responses.


Subject(s)
Oncolytic Virotherapy , Oncolytic Viruses , Succinates , Animals , Humans , Oncolytic Virotherapy/methods , Succinates/pharmacology , Mice , Cell Line, Tumor , Interferon Type I/metabolism , NF-E2-Related Factor 2/metabolism , Colonic Neoplasms/therapy , Colonic Neoplasms/immunology , Colonic Neoplasms/drug therapy , Antiviral Agents/pharmacology , NF-kappa B/metabolism , I-kappa B Kinase/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Inflammation/drug therapy , Female , Vesicular stomatitis Indiana virus/physiology , Vesicular stomatitis Indiana virus/drug effects , Signal Transduction/drug effects
2.
Methods Mol Biol ; 2589: 379-399, 2023.
Article in English | MEDLINE | ID: mdl-36255638

ABSTRACT

Oncolytic virotherapy represents an efficient immunotherapeutic approach for cancer treatment. Oncolytic viruses (OVs) promote antitumor responses through tumor-selective cell lysis and immune system activation. However, some tumor cell lines and primary tumors display resistance to therapy. Here we describe a protocol to identify novel host factors responsible for tumor resistance to oncolysis using an unbiased genome-wide CRISPR-Cas9 loss-of-function screening. Cas9-expressing tumor cells are transduced with a library of pooled single-guide RNA (sgRNA)-expressing lentiviruses that target all human genes to obtain a cell population where each cell is knocked out for a single gene. Upon OV infection, resistant cells survive, while sensitive cells die. The relative abundance of each genome-integrated sgRNA is measured by next-generation sequencing (NGS) in resistant and control cells. This protocol is amenable to uncover host factors involved in the resistance to different OVs in multiple tumor models.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , CRISPR-Cas Systems/genetics , High-Throughput Nucleotide Sequencing , Neoplasms/genetics , Neoplasms/therapy , Oncolytic Viruses/genetics , RNA, Guide, CRISPR-Cas Systems/genetics
3.
Front Cell Infect Microbiol ; 12: 1079926, 2022.
Article in English | MEDLINE | ID: mdl-36590581

ABSTRACT

Virus-Like Particles (VLPs) are nanostructures that share conformation and self-assembly properties with viruses, but lack a viral genome and therefore the infectious capacity. In this study, we produced VLPs by co-expression of VSV glycoprotein (VSV-G) and HIV structural proteins (Gag, Pol) that incorporated a strong sequence-optimized 5'ppp-RNA RIG-I agonist, termed M8. Treatment of target cells with VLPs-M8 generated an antiviral state that conferred resistance against multiple viruses. Interestingly, treatment with VLPs-M8 also elicited a therapeutic effect by inhibiting ongoing viral replication in previously infected cells. Finally, the expression of SARS-CoV-2 Spike glycoprotein on the VLP surface retargeted VLPs to ACE2 expressing cells, thus selectively blocking viral infection in permissive cells. These results highlight the potential of VLPs-M8 as a therapeutic and prophylactic vaccine platform. Overall, these observations indicate that the modification of VLP surface glycoproteins and the incorporation of nucleic acids or therapeutic drugs, will permit modulation of particle tropism, direct specific innate and adaptive immune responses in target tissues, and boost immunogenicity while minimizing off-target effects.


Subject(s)
COVID-19 , Interferon Type I , Vaccines, Virus-Like Particle , Virus Diseases , Humans , SARS-CoV-2 , CD8-Positive T-Lymphocytes , Vaccines, Virus-Like Particle/genetics
4.
J Virol ; 94(24)2020 11 23.
Article in English | MEDLINE | ID: mdl-32999020

ABSTRACT

Dengue virus (DENV) is a mosquito-borne virus that infects upward of 300 million people annually and has the potential to cause fatal hemorrhagic fever and shock. While the parameters contributing to dengue immunopathogenesis remain unclear, the collapse of redox homeostasis and the damage induced by oxidative stress have been correlated with the development of inflammation and progression toward the more severe forms of disease. In the present study, we demonstrate that the accumulation of reactive oxygen species (ROS) late after DENV infection (>24 hpi) resulted from a disruption in the balance between oxidative stress and the nuclear factor erythroid 2-related factor 2 (Nrf2)-dependent antioxidant response. The DENV NS2B3 protease complex strategically targeted Nrf2 for degradation in a proteolysis-independent manner; NS2B3 licensed Nrf2 for lysosomal degradation. Impairment of the Nrf2 regulator by the NS2B3 complex inhibited the antioxidant gene network and contributed to the progressive increase in ROS levels, along with increased virus replication and inflammatory or apoptotic gene expression. By 24 hpi, when increased levels of ROS and antiviral proteins were observed, it appeared that the proviral effect of ROS overcame the antiviral effects of the interferon (IFN) response. Overall, these studies demonstrate that DENV infection disrupts the regulatory interplay between DENV-induced stress responses, Nrf2 antioxidant signaling, and the host antiviral immune response, thus exacerbating oxidative stress and inflammation in DENV infection.IMPORTANCE Dengue virus (DENV) is a mosquito-borne pathogen that threatens 2.5 billion people in more than 100 countries annually. Dengue infection induces a spectrum of clinical symptoms, ranging from classical dengue fever to severe dengue hemorrhagic fever or dengue shock syndrome; however, the complexities of DENV immunopathogenesis remain controversial. Previous studies have reported the importance of the transcription factor Nrf2 in the control of redox homeostasis and antiviral/inflammatory or death responses to DENV. Importantly, the production of reactive oxygen species and the subsequent stress response have been linked to the development of inflammation and progression toward the more severe forms of the disease. Here, we demonstrate that DENV uses the NS2B3 protease complex to strategically target Nrf2 for degradation, leading to a progressive increase in oxidative stress, inflammation, and cell death in infected cells. This study underlines the pivotal role of the Nrf2 regulatory network in the context of DENV infection.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Virus Replication/drug effects , A549 Cells , Cell Line , Dengue/virology , Dengue Virus/genetics , Gene Expression Regulation, Viral , Gene Knockout Techniques , HEK293 Cells , Heme Oxygenase-1/genetics , Humans , Interferons , NF-E2-Related Factor 2/genetics , Reactive Oxygen Species , Signal Transduction/drug effects
5.
Cytokine Growth Factor Rev ; 56: 141-148, 2020 12.
Article in English | MEDLINE | ID: mdl-32859494

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and highly lethal malignancies. Existing therapeutic interventions have so far been unsuccessful in improving prognosis, and survival remains very poor. Oncolytic virotherapy represents a promising, yet not fully explored, alternative strategy for the treatment of PDAC. Oncolytic viruses (OVs) infect, replicate within and lyse tumor cells specifically and stimulate antitumor immune responses. Multiple challenges have hampered the efficacy of oncolytic virotherapy for PDAC, the most significant being the desmoplastic and immunosuppressive pancreatic tumor microenvironment (TME). The TME limits the access of therapeutic drugs and the infiltration of effector T cells and natural killer (NK) cells into the tumor mass. Additionally, cancer cells promote the secretion of immunosuppressive factors and develop mechanisms to evade the host immune system. Because of their oncolytic and immune-stimulating properties, OVs are the ideal candidates for counteracting the pancreatic immunosuppressive TME and for designing combination therapies that can be clinically exploited in clinical trials that seek to improve the prognosis of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Oncolytic Virotherapy , Oncolytic Viruses , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/therapy , Humans , Tumor Microenvironment
6.
Cytokine Growth Factor Rev ; 56: 94-101, 2020 12.
Article in English | MEDLINE | ID: mdl-32826166

ABSTRACT

Recent advances in cancer immunotherapy have renewed interest in oncolytic viruses (OVs) as a synergistic platform for the development of novel antitumor strategies. Cancer cells adopt multiple mechanisms to evade and suppress antitumor immune responses, essentially establishing a non-immunogenic ('cold') tumor microenvironment (TME), with poor T-cell infiltration and low mutational burden. Limitations to the efficacy of immunotherapy still exist, especially for a variety of solid tumors, where new approaches are necessary to overcome physical barriers in the TME and to mitigate adverse effects associated with current immunotherapeutics. OVs offer an attractive alternative by inducing direct oncolysis, immunogenic cell death, and immune stimulation. These multimodal mechanisms make OVs well suited to reprogram non-immunogenic tumors and TME into inflamed, immunogenic ('hot') tumors; enhanced release of tumor antigens by dying cancer cells is expected to augment T-cell infiltration, thereby eliciting potent antitumor immunity. Advances in virus engineering and understanding of tumor biology have allowed the optimization of OV-tumor selectivity, oncolytic potency, and immune stimulation. However, OV antitumor activity is likely to achieve its greatest potential as part of combinatorial strategies with other immune or cancer therapeutics.


Subject(s)
Immunotherapy , Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Neoplasms/therapy , Tumor Microenvironment
7.
Cancer Res ; 80(19): 4087-4102, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32718996

ABSTRACT

Cancer stem-like cells (CSC) induce aggressive tumor phenotypes such as metastasis formation, which is associated with poor prognosis in triple-negative breast cancer (TNBC). Repurposing of FDA-approved drugs that can eradicate the CSC subcompartment in primary tumors may prevent metastatic disease, thus representing an effective strategy to improve the prognosis of TNBC. Here, we investigated spheroid-forming cells in a metastatic TNBC model. This strategy enabled us to specifically study a population of long-lived tumor cells enriched in CSCs, which show stem-like characteristics and induce metastases. To repurpose FDA-approved drugs potentially toxic for CSCs, we focused on pyrvinium pamoate (PP), an anthelmintic drug with documented anticancer activity in preclinical models. PP induced cytotoxic effects in CSCs and prevented metastasis formation. Mechanistically, the cell killing effects of PP were a result of inhibition of lipid anabolism and, more specifically, the impairment of anabolic flux from glucose to cholesterol and fatty acids. CSCs were strongly dependent upon activation of lipid biosynthetic pathways; activation of these pathways exhibited an unfavorable prognostic value in a cohort of breast cancer patients, where it predicted high probability of metastatic dissemination and tumor relapse. Overall, this work describes a new approach to target aggressive CSCs that may substantially improve clinical outcomes for patients with TNBC, who currently lack effective targeted therapeutic options. SIGNIFICANCE: These findings provide preclinical evidence that a drug repurposing approach to prevent metastatic disease in TNBC exploits lipid anabolism as a metabolic vulnerability against CSCs in primary tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplastic Stem Cells/drug effects , Pyrvinium Compounds/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cholesterol/metabolism , Drug Repositioning , Female , Glucose/metabolism , Humans , Lipid Metabolism/drug effects , Mice, Inbred NOD , Neoplastic Stem Cells/pathology , Triple Negative Breast Neoplasms/metabolism , Xenograft Model Antitumor Assays
8.
Cytokine Growth Factor Rev ; 53: 1-9, 2020 06.
Article in English | MEDLINE | ID: mdl-32487439

ABSTRACT

The coronavirus pandemic has engulfed the nations of the world for the first five months of 2020 and altered the pace, fabric and nature of our lives. In this overview accompanying the Special Issue of Cytokine & Growth Factor Reviews, we examine some of the many social and scientific issues impacted by SARS-CoV2 - personal lives, economy, scientific communication, the environment. International members of Istituto Pasteur in Rome and INITIATE, the Marie Curie Training Network reflect on the lasting global impact of the coronavirus pandemic.


Subject(s)
Communicable Disease Control/methods , Coronavirus Infections/epidemiology , Pandemics/statistics & numerical data , Pneumonia, Viral/epidemiology , Betacoronavirus/isolation & purification , Betacoronavirus/pathogenicity , COVID-19 , Humans , SARS-CoV-2
9.
Cancer Immunol Immunother ; 68(9): 1479-1492, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31463653

ABSTRACT

RIG-I is a cytosolic RNA sensor that recognizes short 5' triphosphate RNA, commonly generated during virus infection. Upon activation, RIG-I initiates antiviral immunity, and in some circumstances, induces cell death. Because of this dual capacity, RIG-I has emerged as a promising target for cancer immunotherapy. Previously, a sequence-optimized RIG-I agonist (termed M8) was generated and shown to stimulate a robust immune response capable of blocking viral infection and to function as an adjuvant in vaccination strategies. Here, we investigated the potential of M8 as an anti-cancer agent by analyzing its ability to induce cell death and activate the immune response. In multiple cancer cell lines, M8 treatment strongly activated caspase 3-dependent apoptosis, that relied on an intrinsic NOXA and PUMA-driven pathway that was dependent on IFN-I signaling. Additionally, cell death induced by M8 was characterized by the expression of markers of immunogenic cell death-related damage-associated molecular patterns (ICD-DAMP)-calreticulin, HMGB1 and ATP-and high levels of ICD-related cytokines CXCL10, IFNß, CCL2 and CXCL1. Moreover, M8 increased the levels of HLA-ABC expression on the tumor cell surface, as well as up-regulation of genes involved in antigen processing and presentation. M8 induction of the RIG-I pathway in cancer cells favored dendritic cell phagocytosis and induction of co-stimulatory molecules CD80 and CD86, together with increased expression of IL12 and CXCL10. Altogether, these results highlight the potential of M8 in cancer immunotherapy, with the capacity to induce ICD-DAMP on tumor cells and activate immunostimulatory signals that synergize with current therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Dendritic Cells/immunology , Melanoma/drug therapy , Nelfinavir/analogs & derivatives , Alarmins/immunology , Antigen Presentation/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Calreticulin/metabolism , Caspase 3/metabolism , Cell Differentiation , Cell Line, Tumor , DEAD Box Protein 58/antagonists & inhibitors , HMGB1 Protein/metabolism , Humans , Immunization , Interferons/metabolism , Molecular Targeted Therapy , Nelfinavir/pharmacology , Nelfinavir/therapeutic use , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Immunologic , Signal Transduction
10.
J Virol ; 93(21)2019 11 01.
Article in English | MEDLINE | ID: mdl-31413127

ABSTRACT

The presence of T cell reservoirs in which human immunodeficiency virus (HIV) establishes latency by integrating into the host genome represents a major obstacle to an HIV cure and has prompted the development of strategies aimed at the eradication of HIV from latently infected cells. The "shock-and-kill" strategy is one of the most pursued approaches to the elimination of viral reservoirs. Although several latency-reversing agents (LRAs) have shown promising reactivation activity, they have failed to eliminate the cellular reservoir. In this study, we evaluated a novel immune system-mediated approach to clearing the HIV reservoir, based on a combination of innate immune stimulation and epigenetic reprogramming. The combination of the STING agonist cGAMP (cyclic GMP-AMP) and the FDA-approved histone deacetylase inhibitor resminostat resulted in a significant increase in HIV proviral reactivation and specific apoptosis in HIV-infected cells in vitro Reductions in the proportion of HIV-harboring cells and the total amount of HIV DNA were also observed in CD4+ central memory T (TCM) cells, a primary cell model of latency, where resminostat alone or together with cGAMP induced high levels of selective cell death. Finally, high levels of cell-associated HIV RNA were detected ex vivo in peripheral blood mononuclear cells (PBMCs) and CD4+ T cells from individuals on suppressive antiretroviral therapy (ART). Although synergism was not detected in PBMCs with the combination, viral RNA expression was significantly increased in CD4+ T cells. Collectively, these results represent a promising step toward HIV eradication by demonstrating the potential of innate immune activation and epigenetic modulation for reducing the viral reservoir and inducing specific death of HIV-infected cells.IMPORTANCE One of the challenges associated with HIV-1 infection is that despite antiretroviral therapies that reduce HIV-1 loads to undetectable levels, proviral DNA remains dormant in a subpopulation of T lymphocytes. Numerous strategies to clear residual virus by reactivating latent virus and eliminating the reservoir of HIV-1 (so-called "shock-and-kill" strategies) have been proposed. In the present study, we use a combination of small molecules that activate the cGAS-STING antiviral innate immune response (the di-cyclic nucleotide cGAMP) and epigenetic modulators (histone deacetylase inhibitors) that induce reactivation and HIV-infected T cell killing in cell lines, primary T lymphocytes, and patient samples. These studies represent a novel strategy for HIV eradication by reducing the viral reservoir and inducing specific death of HIV-infected cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epigenesis, Genetic , HIV Infections/immunology , HIV-1/immunology , Immunity, Innate/immunology , Virus Activation/immunology , Virus Latency/immunology , Gene Expression Regulation, Viral , HIV Infections/genetics , HIV Infections/virology , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Sulfonamides/pharmacology , Virus Replication
11.
J Virol ; 93(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31092575

ABSTRACT

Oncolytic virotherapy represents a promising experimental anticancer strategy, based on the use of genetically modified viruses to selectively infect and kill cancer cells. Vesicular stomatitis virus (VSV) is a prototypic oncolytic virus (OV) that induces cancer cell death through activation of the apoptotic pathway, although intrinsic resistance to oncolysis is found in some cell lines and many primary tumors, as a consequence of residual innate immunity to the virus. In the effort to improve OV therapeutic efficacy, we previously demonstrated that different agents, including histone deacetylase inhibitors (HDIs), functioned as reversible chemical switches to dampen the innate antiviral response and improve the susceptibility of resistant cancer cells to VSV infection. In the present study, we demonstrated that the NAD+-dependent histone deacetylase SIRT1 (silent mating type information regulation 2 homolog 1) plays a key role in the permissivity of prostate cancer PC-3 cells to VSVΔM51 replication and oncolysis. HDI-mediated enhancement of VSVΔM51 infection and cancer cell killing directly correlated with a decrease of SIRT1 expression. Furthermore, pharmacological inhibition as well as silencing of SIRT1 by small interfering RNA (siRNA) was sufficient to sensitize PC-3 cells to VSVΔM51 infection, resulting in augmentation of virus replication and spread. Mechanistically, HDIs such as suberoylanilide hydroxamic acid (SAHA; Vorinostat) and resminostat upregulated the microRNA miR-34a that regulated the level of SIRT1. Taken together, our findings identify SIRT1 as a viral restriction factor that limits VSVΔM51 infection and oncolysis in prostate cancer cells.IMPORTANCE The use of nonpathogenic viruses to target and kill cancer cells is a promising strategy in cancer therapy. However, many types of human cancer are resistant to the oncolytic (cancer-killing) effects of virotherapy. In this study, we identify a host cellular protein, SIRT1, that contributes to the sensitivity of prostate cancer cells to infection by a prototypical oncolytic virus. Knockout of SIRT1 activity increases the sensitivity of prostate cancer cells to virus-mediated killing. At the molecular level, SIRT1 is controlled by a small microRNA termed miR-34a. Altogether, SIRT1 and/or miR-34a levels may serve as predictors of response to oncolytic-virus therapy.


Subject(s)
Host Microbial Interactions , Immunity, Innate , Oncolytic Viruses/growth & development , Sirtuin 1/metabolism , Vesiculovirus/growth & development , Virus Replication , Humans , Male , Oncolytic Viruses/immunology , PC-3 Cells , Vesiculovirus/immunology
12.
Nat Commun ; 9(1): 1080, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540686

ABSTRACT

CD28 superagonistic antibodies (CD28SAb) can preferentially activate and expand immunosuppressive regulatory T cells (Treg) in mice. However, pre-clinical trials assessing CD28SAbs for the therapy of autoimmune diseases reveal severe systemic inflammatory response syndrome in humans, thereby implying the existence of distinct signalling abilities between human and mouse CD28. Here, we show that a single amino acid variant within the C-terminal proline-rich motif of human and mouse CD28 (P212 in human vs. A210 in mouse) regulates CD28-induced NF-κB activation and pro-inflammatory cytokine gene expression. Moreover, this Y209APP212 sequence in humans is crucial for the association of CD28 with the Nck adaptor protein for actin cytoskeleton reorganisation events necessary for CD28 autonomous signalling. This study thus unveils different outcomes between human and mouse CD28 signalling to underscore the importance of species difference when transferring results from preclinical models to the bedside.


Subject(s)
Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , CD28 Antigens/genetics , CD28 Antigens/metabolism , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , Mice , NF-kappa B/metabolism , Oncogene Proteins/metabolism , Protein Binding , Signal Transduction/genetics , T-Lymphocytes, Regulatory/metabolism
13.
DNA Cell Biol ; 35(8): 367-72, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27348136

ABSTRACT

A spotlight has been focused on the mosquito-borne Zika virus (ZIKV) because of its epidemic outbreak in Brazil and Latin America, as well as the severe neurological manifestations of microcephaly and Guillain-Barré syndrome associated with infection. In this review, we discuss the recent literature on ZIKV-host interactions, including new mechanistic insight concerning the basis of ZIKV-induced neuropathogenesis.


Subject(s)
Disease Outbreaks , Guillain-Barre Syndrome/pathology , Microcephaly/pathology , Virion/pathogenicity , Zika Virus Infection/complications , Zika Virus/pathogenicity , Aedes/virology , Animals , Female , Fetus , Guillain-Barre Syndrome/epidemiology , Guillain-Barre Syndrome/etiology , Guillain-Barre Syndrome/virology , Host-Pathogen Interactions , Humans , Latin America/epidemiology , Microcephaly/epidemiology , Microcephaly/etiology , Microcephaly/virology , Mosquito Vectors/virology , Placenta/pathology , Placenta/virology , Pregnancy , Virion/physiology , Zika Virus/physiology , Zika Virus Infection/epidemiology , Zika Virus Infection/transmission , Zika Virus Infection/virology
14.
J Immunol ; 196(4): 1955-63, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26773155

ABSTRACT

Phosphatidylinositol 4,5-biphosphate (PIP2) is critical for T lymphocyte activation serving as a substrate for the generation of second messengers and the remodeling of actin cytoskeleton necessary for the clustering of lipid rafts, TCR, and costimulatory receptors toward the T:APC interface. Spatiotemporal analysis of PIP2 synthesis in T lymphocytes suggested that distinct isoforms of the main PIP2-generating enzyme, phosphatidylinositol 4-phosphate 5-kinase (PIP5K), play a differential role on the basis of their distinct localization. In this study, we analyze the contribution of PIP5Kß to T cell activation and show that CD28 induces the recruitment of PIP5Kß to the immunological synapse, where it regulates filamin A and lipid raft accumulation, as well as T cell activation, in a nonredundant manner. Finally, we found that Vav1 and the C-terminal 83 aa of PIP5Kß are pivotal for the PIP5Kß regulatory functions in response to CD28 stimulation.


Subject(s)
Immunological Synapses/immunology , Lymphocyte Activation/immunology , Membrane Microdomains/immunology , Phosphotransferases (Alcohol Group Acceptor)/immunology , T-Lymphocytes/immunology , CD28 Antigens/immunology , CD28 Antigens/metabolism , Humans , Image Processing, Computer-Assisted , Isoenzymes/immunology , Isoenzymes/metabolism , Microscopy, Confocal , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-vav/immunology , Proto-Oncogene Proteins c-vav/metabolism , Real-Time Polymerase Chain Reaction , T-Lymphocytes/enzymology , Transfection
15.
Cell Mol Life Sci ; 72(23): 4461-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26265181

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PIP2) represents about 1 % of plasma membrane phospholipids and behaves as a pleiotropic regulator of a striking number of fundamental cellular processes. In recent years, an increasing body of literature has highlighted an essential role of PIP2 in multiple aspects of leukocyte biology. In this emerging picture, PIP2 is envisaged as a signalling intermediate itself and as a membrane-bound regulator and a scaffold of proteins with specific PIP2 binding domains. Indeed PIP2 plays a key role in several functions. These include directional migration in neutrophils, integrin-dependent adhesion in T lymphocytes, phagocytosis in macrophages, lysosomes secretion and trafficking at immune synapse in cytolytic effectors and secretory cells, calcium signals and gene transcription in B lymphocytes, natural killer cells and mast cells. The coordination of these different aspects relies on the spatio-temporal organisation of distinct PIP2 pools, generated by the main PIP2 generating enzyme, phosphatidylinositol 4-phosphate 5-kinase (PIP5K). Three different isoforms of PIP5K, named α, ß and γ, and different splice variants have been described in leukocyte populations. The isoform-specific coupling of specific isoforms of PIP5K to different families of activating receptors, including integrins, Fc receptors, toll-like receptors and chemokine receptors, is starting to be reported. Furthermore, PIP2 is turned over by multiple metabolising enzymes including phospholipase C (PLC) γ and phosphatidylinositol 3-kinase (PI3K) which, along with Rho family small G proteins, is widely involved in strategic functions within the immune system. The interplay between PIP2, lipid-modifying enzymes and small G protein-regulated signals is also discussed.


Subject(s)
Leukocytes/immunology , Leukocytes/metabolism , Phosphatidylinositol 4,5-Diphosphate/physiology , Animals , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mast Cells/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinases/metabolism , Signal Transduction
16.
J Immunol ; 194(3): 1323-33, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25539813

ABSTRACT

Phosphatidylinositol 4,5-biphosphate (PIP2) is a cell membrane phosphoinositide crucial for cell signaling and activation. Indeed, PIP2 is a pivotal source for second messenger generation and controlling the activity of several proteins regulating cytoskeleton reorganization. Despite its critical role in T cell activation, the molecular mechanisms regulating PIP2 turnover remain largely unknown. In human primary CD4(+) T lymphocytes, we have recently demonstrated that CD28 costimulatory receptor is crucial for regulating PIP2 turnover by allowing the recruitment and activation of the lipid kinase phosphatidylinositol 4-phosphate 5-kinase (PIP5Kα). We also identified PIP5Kα as a key modulator of CD28 costimulatory signals leading to the efficient T cell activation. In this study, we extend these data by demonstrating that PIP5Kα recruitment and activation is essential for CD28-mediated cytoskeleton rearrangement necessary for organizing a complete signaling compartment leading to downstream signaling functions. We also identified Vav1 as the linker molecule that couples the C-terminal proline-rich motif of CD28 to the recruitment and activation of PIP5Kα, which in turn cooperates with Vav1 in regulating actin polymerization and CD28 signaling functions.


Subject(s)
Actins/metabolism , CD28 Antigens/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD28 Antigens/chemistry , CD28 Antigens/genetics , Cell Communication , Cell Line , Enzyme Activation , Gene Expression , Humans , Mutation , Oncogene Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Proline-Rich Protein Domains , Protein Binding , Protein Interaction Domains and Motifs , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
17.
Immunol Lett ; 158(1-2): 134-42, 2014.
Article in English | MEDLINE | ID: mdl-24412596

ABSTRACT

CD28 is a crucial costimulatory receptor necessary full T cell activation. The role of CD28 in multiple sclerosis (MS) has been evaluated as the source of costimulatory signals integrating those delivered by TCR. However, CD28 is also able to act as a unique signaling receptor and to deliver TCR-independent autonomous signals, which regulate the expression and production of pro-inflammatory cytokines and chemokines. By comparing the cytokine/chemokine profiles of CD4(+) T cells from relapsing-remitting multiple sclerosis (RRMS) patients and healthy donors (HD), we found that CD28 engagement without TCR strongly up-regulates IL-8 and IL-6 expression in RRMS compared to HD. More interestingly, in RRMS but not in HD, CD28 stimulation selectively induces the expression of IL-17A by cooperating with IL-6-mediated signals. By using specific inhibitory drugs, we also identify the phosphatidylinositol 3 kinase (PI3K) as the critical regulator of CD28 proinflammatory functions in MS.


Subject(s)
CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Interleukin-17/metabolism , Multiple Sclerosis, Relapsing-Remitting/immunology , Receptors, Antigen, T-Cell/metabolism , Adult , Animals , B7-2 Antigen/genetics , B7-2 Antigen/metabolism , Chromones/pharmacology , Female , Humans , Inflammation Mediators/metabolism , Interleukin-17/genetics , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , L Cells , Male , Mice , Middle Aged , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Transgenes/genetics , Up-Regulation/drug effects
18.
FEMS Yeast Res ; 13(7): 682-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23875998

ABSTRACT

The tumor suppressor p53 plays a central role in the regulation of cellular growth and apoptosis. In the yeast Saccharomyces cerevisiae, the overexpression of the human p53 leads to growth inhibition and apoptotic cell death on minimal medium. In the present work, we show that p53-expressing cells are more susceptible to cell death after an apoptotic stimulus such as H2O2. The analysis of mutants involved in yeast apoptosis-like death suggests that the observed cell death is Yca1 independent and mainly mediated through Nuc1p.


Subject(s)
Apoptosis , Endonucleases/metabolism , Exonucleases/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Caspases/metabolism , Culture Media/chemistry , Gene Expression , Humans , Hydrogen Peroxide/toxicity , Saccharomyces cerevisiae/genetics
19.
J Immunol ; 190(10): 5279-86, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23589613

ABSTRACT

CD28 is one of the most relevant costimulatory receptors that deliver both TCR-dependent and TCR-independent signals regulating a wide range of signaling pathways crucial for cytokine and chemokine gene expressions, T cell survival, and proliferation. Most of the CD28-dependent signaling functions are initiated by the recruitment and activation of class IA PI3Ks, which catalyze the conversion of phosphatidylinositol 4,5-biphosphate (PIP2) into phosphatidylinositol 3,4,5-triphosphate, thus generating the docking sites for key signaling proteins. Hence, PIP2 is a crucial substrate in driving the PI3K downstream signaling pathways, and PIP2 turnover may be an essential regulatory step to ensure the activation of PI3K following CD28 engagement. Despite some data evidence that CD28 augments TCR-induced turnover of PIP2, its direct role in regulating PIP2 metabolism has never been assessed. In this study, we show that CD28 regulates PIP2 turnover by recruiting and activating phosphatidylinositol 4-phosphate 5-kinases α (PIP5Kα) in human primary CD4(+) T lymphocytes. This event leads to the neosynthesis of PIP2 and to its consumption by CD28-activated PI3K. We also evidenced that PIP5Kα activation is required for both CD28 unique signals regulating IL-8 gene expression as well as for CD28/TCR-induced Ca(2+) mobilization, NF-AT nuclear translocation, and IL-2 gene transcription. Our findings elucidate a novel mechanism that involves PIP5Kα as a key modulator of CD28 costimulatory signals.


Subject(s)
CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Interleukin-2/genetics , Interleukin-8/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Calcium/metabolism , Cells, Cultured , Enzyme Activation , Gene Expression , Humans , Interleukin-8/genetics , Lymphocyte Activation , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 4,5-Diphosphate/chemistry , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction
20.
J Biol Chem ; 286(46): 39693-702, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21953469

ABSTRACT

Stress-induced monoubiquitination of p53 is a crucial event for the nuclear-cytoplasm-mitochondria trafficking and transcription-independent pro-apoptotic functions of p53. Although an intact ubiquitination pathway and a functional nuclear export sequence are required for p53 nuclear export, the role of specific residues within this region in regulating both processes remains largely unknown. Here we characterize the mechanisms accounting for the nuclear accumulation of a new point mutation (Lys-351 to Asn) in the nuclear export sequence of p53 identified in a cisplatin-resistant ovarian carcinoma cell line (A2780 CIS). We found that K351N substitution abrogates the monoubiquitination of p53 induced by both Mdm2 and MSL2 E3-ligases. As a consequence, cells expressing p53 K351N mutant showed defects in cisplatin-induced translocation of p53 to mitochondria, Bax oligomerization, and mitochondrial membrane depolarization. These data identify K351N as a critical mutation of p53 that contributes to the development and maintenance of resistance to cisplatin.


Subject(s)
Drug Resistance, Neoplasm , Mitochondria/metabolism , Mutation, Missense , Ovarian Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitination , Amino Acid Substitution , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Female , Humans , Membrane Potential, Mitochondrial/genetics , Mitochondria/genetics , Nuclear Export Signals/genetics , Ovarian Neoplasms/genetics , Protein Transport/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...