Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 23(23)2022 Dec 03.
Article in English | MEDLINE | ID: mdl-36499601

ABSTRACT

Inherited retinal disorders (IRDs) affect millions of people worldwide and are a major cause of irreversible blindness. Therapies based on drugs, gene augmentation or transplantation approaches have been widely investigated and proposed. Among gene therapies for retinal degenerative diseases, the fast-evolving genome-editing CRISPR/Cas technology has emerged as a new potential treatment. The CRISPR/Cas system has been developed as a powerful genome-editing tool in ophthalmic studies and has been applied not only to gain proof of principle for gene therapies in vivo, but has also been extensively used in basic research to model diseases-in-a-dish. Indeed, the CRISPR/Cas technology has been exploited to genetically modify human induced pluripotent stem cells (iPSCs) to model retinal disorders in vitro, to test in vitro drugs and therapies and to provide a cell source for autologous transplantation. In this review, we will focus on the technological advances in iPSC-based cellular reprogramming and gene editing technologies to create human in vitro models that accurately recapitulate IRD mechanisms towards the development of treatments for retinal degenerative diseases.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Degeneration , Humans , Gene Editing , CRISPR-Cas Systems/genetics , Retinal Degeneration/genetics , Retinal Degeneration/therapy , Genetic Therapy
2.
Int J Mol Sci ; 23(14)2022 Jul 07.
Article in English | MEDLINE | ID: mdl-35886882

ABSTRACT

Sleeping Beauty (SB) is the first DNA transposon employed for efficient transposition in vertebrate cells, opening new applications for genetic engineering and gene therapies. A transposon-based gene delivery system holds the favourable features of non-viral vectors and an attractive safety profile. Here, we employed SB to engineer HEK293 cells for optimizing the production of a chimpanzee Adenovector (chAd) belonging to the Human Mastadenovirus C species. To date, chAd vectors are employed in several clinical settings for infectious diseases, last but not least COVID-19. A robust, efficient and quick viral vector production could advance the clinical application of chAd vectors. To this aim, we firstly swapped the hAd5 E1 with chAd-C E1 gene by using the CRISPR/Cas9 system. We demonstrated that in the absence of human Ad5 E1, chimp Ad-C E1 gene did not support HEK293 survival. To improve chAd-C vector production, we engineered HEK293 cells to stably express the chAd-C precursor terminal protein (ch.pTP), which plays a crucial role in chimpanzee Adenoviral DNA replication. The results indicate that exogenous ch.pTP expression significantly ameliorate the packaging and amplification of recombinant chAd-C vectors thus, the engineered HEK293ch.pTP cells could represent a superior packaging cell line for the production of these vectors.


Subject(s)
COVID-19 , Pan troglodytes , Adenoviridae/genetics , Animals , DNA Transposable Elements/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , HEK293 Cells , Humans , Pan troglodytes/genetics
3.
J Exp Clin Cancer Res ; 40(1): 362, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34782004

ABSTRACT

BACKGROUND: Approaches based on expression signatures of prostate cancer (PCa) have been proposed to predict patient outcomes and response to treatments. The transcription factor NF-Y participates to the progression from benign epithelium to both localized and metastatic PCa and is associated with aggressive transcriptional profile. The gene encoding for NF-YA, the DNA-binding subunit of NF-Y, produces two alternatively spliced transcripts, NF-YAs and NF-YAl. Bioinformatic analyses pointed at NF-YA splicing as a key transcriptional signature to discriminate between different tumor molecular subtypes. In this study, we aimed to determine the pathophysiological role of NF-YA splice variants in PCa and their association with aggressive subtypes. METHODS: Data on the expression of NF-YA isoforms were extracted from the TCGA (The Cancer Genome Atlas) database of tumor prostate tissues and validated in prostate cell lines. Lentiviral transduction and CRISPR-Cas9 technology allowed the modulation of the expression of NF-YA splice variants in PCa cells. We characterized 3D cell cultures through in vitro assays and RNA-seq profilings. We used the rank-rank hypergeometric overlap approach to identify concordant/discordant gene expression signatures of NF-YAs/NF-YAl-overexpressing cells and human PCa patients. We performed in vivo studies in SHO-SCID mice to determine pathological and molecular phenotypes of NF-YAs/NF-YAl xenograft tumors. RESULTS: NF-YA depletion affects the tumorigenic potential of PCa cells in vitro and in vivo. Elevated NF-YAs levels are associated to aggressive PCa specimens, defined by Gleason Score and TNM classification. NF-YAl overexpression increases cell motility, while NF-YAs enhances cell proliferation in PCa 3D spheroids and xenograft tumors. The transcriptome of NF-YAs-spheroids has an extensive overlap with localized and metastatic human PCa signatures. According to PCa PAM50 classification, NF-YAs transcript levels are higher in LumB, characterized by poor prognosis compared to LumA and basal subtypes. A significant decrease in NF-YAs/NF-YAl ratio distinguishes PCa circulating tumor cells from cancer cells in metastatic sites, consistently with pro-migratory function of NF-YAl. Stratification of patients based on NF-YAs expression is predictive of clinical outcome. CONCLUSIONS: Altogether, our results indicate that the modulation of NF-YA isoforms affects prostate pathophysiological processes and contributes to cancer-relevant phenotype, in vitro and in vivo. Evaluation of NF-YA splicing may represent a new molecular strategy for risk assessment of PCa patients.


Subject(s)
Alternative Splicing/genetics , CCAAT-Binding Factor/metabolism , Gene Editing/methods , Prostatic Neoplasms/genetics , Animals , Humans , Male , Mice , Xenograft Model Antitumor Assays
4.
Am J Hum Genet ; 108(2): 295-308, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33508235

ABSTRACT

Retinitis pigmentosa (RP) is a group of progressive retinal degenerations of mostly monogenic inheritance, which cause blindness in about 1:3,500 individuals worldwide. Heterozygous variants in the rhodopsin (RHO) gene are the most common cause of autosomal dominant RP (adRP). Among these, missense variants at C-terminal proline 347, such as p.Pro347Ser, cause severe adRP recurrently in European affected individuals. Here, for the first time, we use CRISPR/Cas9 to selectively target the p.Pro347Ser variant while preserving the wild-type RHO allele in vitro and in a mouse model of adRP. Detailed in vitro, genomic, and biochemical characterization of the rhodopsin C-terminal editing demonstrates a safe downregulation of p.Pro347Ser expression leading to partial recovery of photoreceptor function in a transgenic mouse model treated with adeno-associated viral vectors. This study supports the safety and efficacy of CRISPR/Cas9-mediated allele-specific editing and paves the way for a permanent and precise correction of heterozygous variants in dominantly inherited retinal diseases.


Subject(s)
Gene Editing , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Rhodopsin/genetics , Alleles , Animals , CRISPR-Cas Systems , Cell Line , Dependovirus/genetics , Disease Models, Animal , Electroretinography , Genetic Therapy , Humans , INDEL Mutation , Mice , Mice, Transgenic , Mutation, Missense , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Retina/physiopathology , Rhodopsin/metabolism
5.
J Med Genet ; 57(7): 437-444, 2020 07.
Article in English | MEDLINE | ID: mdl-31857428

ABSTRACT

Retinal diseases (RD) include inherited retinal dystrophy (IRD), for example, retinitis pigmentosa and Leber's congenital amaurosis, or multifactorial forms, for example, age-related macular degeneration (AMD). IRDs are clinically and genetically heterogeneous in nature. To date, more than 200 genes are known to cause IRDs, which perturb the development, function and survival of rod and cone photoreceptors or retinal pigment epithelial cells. Conversely, AMD, the most common cause of blindness in the developed world, is an acquired disease of the macula characterised by progressive visual impairment. To date, available therapeutic approaches for RD include nutritional supplements, neurotrophic factors, antiangiogenic drugs for wet AMD and gene augmentation/interference strategy for IRDs. However, these therapies do not aim at correcting the genetic defect and result in inefficient and expensive treatments. The genome editing technology based on clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein (Cas) and an RNA that guides the Cas protein to a predetermined region of the genome, represents an attractive strategy to tackle IRDs without available cure. Indeed, CRISPR/Cas system can permanently and precisely replace or remove genetic mutations causative of a disease, representing a molecular tool to cure a genetic disorder. In this review, we will introduce the mechanism of CRISPR/Cas system, presenting an updated panel of Cas variants and delivery systems, then we will focus on applications of CRISPR/Cas genome editing in the retina, and, as emerging treatment options, in patient-derived induced pluripotent stem cells followed by transplantation of retinal progenitor cells into the eye.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Transfer Techniques/trends , Macular Degeneration/therapy , Retinal Diseases/therapy , Gene Editing/methods , Humans , Induced Pluripotent Stem Cells/transplantation , Macular Degeneration/genetics , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/therapy , Retina/pathology , Retina/transplantation , Retinal Diseases/genetics , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy
6.
Methods Mol Biol ; 1834: 59-74, 2019.
Article in English | MEDLINE | ID: mdl-30324436

ABSTRACT

CRISPR/Cas9 is an efficient tool to knock down specific genes in various organisms. In this chapter, we describe how to assess knockdown of human rhodopsin (RHO) gene carrying the P23H mutation in vitro, in engineered HeLa cells, and in vivo, in P23H RHO transgenic mice. To this aim, we report two molecular assays: site-specific PCR on P23H RHO cells treated with CRISPR/Cas9 and Western blotting analysis on retinal cells prepared from P23H RHO transgenic mice electroporated with CRISPR/Cas9 and GFP plasmids.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Retina/cytology , Retina/metabolism , Animals , Cloning, Molecular , Computational Biology/methods , Fluorescent Antibody Technique , Gene Expression , Gene Knockdown Techniques , Gene Targeting , HeLa Cells , Humans , Mice , Polymerase Chain Reaction , RNA, Guide, Kinetoplastida , Rhodopsin/genetics
7.
Mol Ther ; 26(11): 2592-2603, 2018 11 07.
Article in English | MEDLINE | ID: mdl-30122422

ABSTRACT

Deficiency of basement membrane heterotrimeric laminin 332 component, coded by LAMA3, LAMB3, and LAMC2 genes, causes junctional epidermolysis bullosa (JEB), a severe skin adhesion defect. Herein, we report the first application of CRISPR/Cas9-mediated homology direct repair (HDR) to in situ restore LAMB3 expression in JEB keratinocytes in vitro and in immunodeficient mice transplanted with genetically corrected skin equivalents. We packaged an adenovector carrying Cas9/guide RNA (gRNA) tailored to the intron 2 of LAMB3 gene and an integration defective lentiviral vector bearing a promoterless quasi-complete LAMB3 cDNA downstream a splice acceptor site and flanked by homology arms. Upon genuine HDR, we exploited the in vitro adhesion advantage of laminin 332 production to positively select LAMB3-expressing keratinocytes. HDR and restored laminin 332 expression were evaluated at single-cell level. Notably, monoallelic-targeted integration of LAMB3 cDNA was sufficient to in vitro recapitulate the adhesive property, the colony formation typical of normal keratinocytes, as well as their cell growth. Grafting of genetically corrected skin equivalents onto immunodeficient mice showed a completely restored dermal-epidermal junction. This study provides evidence for efficient CRISPR/Cas9-mediated in situ restoration of LAMB3 expression, paving the way for ex vivo clinical application of this strategy to laminin 332 deficiency.


Subject(s)
CRISPR-Cas Systems/genetics , Cell Adhesion Molecules/genetics , Epidermolysis Bullosa, Junctional/therapy , Genetic Therapy , Animals , Basement Membrane/pathology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/deficiency , DNA Repair/genetics , DNA, Complementary/genetics , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/pathology , Gene Expression Regulation , Humans , Introns/genetics , Keratinocytes/metabolism , Keratinocytes/pathology , Laminin/genetics , Lentivirus/genetics , Mice , Mutation , RNA Editing/genetics , Kalinin
8.
Sci Immunol ; 3(24)2018 06 08.
Article in English | MEDLINE | ID: mdl-29884618

ABSTRACT

Rare individuals, termed HIV controllers, spontaneously control HIV infection by mounting efficient T cell responses against the virus. Protective CD4+ T cell responses from HIV controllers involve high-affinity public T cell receptors (TCRs) recognizing an immunodominant capsid epitope (Gag293) presented by a remarkably broad array of human leukocyte antigen (HLA) class II molecules. Here, we determine the structures of a prototypical public TCR bound to HLA-DR1, HLA-DR11, and HLA-DR15 molecules presenting the Gag293 epitope. TCR recognition was driven by contacts with the Gag293 epitope, a feature that underpinned the extensive HLA cross-restriction. These high-affinity TCRs promoted mature immunological synapse formation and cytotoxic capacity in both CD4+ and CD8+ T cells. The public TCRs suppressed HIV replication in multiple genetic backgrounds ex vivo, emphasizing the functional advantage conferred by broad HLA class II cross-restriction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , HLA-D Antigens/immunology , Host-Pathogen Interactions/immunology , Animals , Cross Reactions/immunology , Fibroblasts , HEK293 Cells , Healthy Volunteers , Humans , Jurkat Cells , Leukocytes, Mononuclear , Lymphocyte Activation , Mice , Primary Cell Culture , Receptors, Antigen, T-Cell/immunology , Viral Load/immunology , Virus Replication/immunology
9.
J Vis Exp ; (131)2018 01 12.
Article in English | MEDLINE | ID: mdl-29364270

ABSTRACT

The Sleeping Beauty (SB) transposon is a non-viral integrating system with proven efficacy for gene transfer and functional genomics. To optimize the SB transposon machinery, a transcriptionally regulated hyperactive transposase (SB100X) and T2-based transposon are employed. Typically, the transposase and transposon are provided transiently by plasmid transfection and SB100X expression is driven by a constitutive promoter. Here, we describe an efficient method to deliver the SB components to human cells that are resistant to several physical and chemical transfection methods, to control SB100X expression and stably integrate a gene of interest (GOI) through a "cut and paste" SB mechanism. The expression of hyperactive transposase is tightly controlled by the Tet-ON system, widely used to control gene expression since 1992. The gene of interest is flanked by inverted repeats (IR) of the T2 transposon. Both SB components are packaged in integration defective lentiviral vectors transiently produced in HEK293T cells. Human cells, either cell lines or primary cells from human tissue, are in vitro transiently transduced with viral vectors. Upon addition of doxycycline (dox, tetracycline analog) into the culture medium, a fine-tuning of transposase expression is measured and results in a long-lasting integration of the gene of interest in the genome of the treated cells. This method is efficient and applicable to the cell line (e.g., HeLa cells) and primary cells (e.g., human primary keratinocytes), and thus represents a valuable tool for genetic engineering and therapeutic gene transfer.


Subject(s)
DNA Transposable Elements , Genetic Vectors/genetics , Lentivirus/genetics , Transposases/genetics , Animals , HEK293 Cells , HeLa Cells , Humans , Transfection
10.
J Immunol ; 199(10): 3437-3452, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28993513

ABSTRACT

Rare patients who spontaneously control HIV replication provide a useful model to inform HIV vaccine development. HIV controllers develop particularly efficient antiviral CD4+ T cell responses mediated by shared high-affinity TCRs. To determine whether the candidate DNA vaccine ADVAX could induce similar responses, we analyzed Gag-specific primary CD4+ T cells from healthy volunteers who received ADVAX DNA by electroporation. Vaccinated volunteers had an immunodominant response to the Gag293 epitope with a functional avidity intermediate between that of controllers and treated patients. The TCR repertoire of Gag293-specific CD4+ T cells proved highly biased, with a predominant usage of the TCRß variable gene 2 (TRBV2) in vaccinees as well as controllers. TCRα variable gene (TRAV) gene usage was more diverse, with the dominance of TRAV29 over TRAV24 genes in vaccinees, whereas TRAV24 predominated in controllers. Sequence analysis revealed an unexpected degree of overlap between the specific repertoires of vaccinees and controllers, with the sharing of TRAV24 and TRBV2 public motifs (>30%) and of public clonotypes characteristic of high-affinity TCRs. MHC class II tetramer binding revealed a broad HLA-DR cross-restriction, explaining how Gag293-specific public clonotypes could be selected in individuals with diverse genetic backgrounds. TRAV29 clonotypes also proved cross-restricted, but conferred responses of lower functional avidity upon TCR transfer. In conclusion, DNA vaccination by electroporation primed for TCR clonotypes that were associated with HIV control, highlighting the potential of this vaccine delivery method. To our knowledge, this study provides the first proof-of-concept that clonotypic analysis may be used as a tool to monitor the quality of vaccine-induced responses and modulate these toward "controller-like" responses.


Subject(s)
AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV/physiology , Receptors, Antigen, T-Cell, alpha-beta/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology , Clone Cells , Cross Reactions , Electroporation , Enzyme-Linked Immunospot Assay , HLA-DR Antigens/metabolism , Humans , Lymphocyte Activation , Protein Binding , Vaccines, DNA , Virus Replication
11.
Bio Protoc ; 7(6): e2187, 2017 Mar 20.
Article in English | MEDLINE | ID: mdl-34458496

ABSTRACT

Major Histocompatibility Complex (MHC) tetramers have been used for two decades to detect, isolate and characterize T cells specific for various pathogens and tumor antigens. In the context of Human Immunodeficiency Virus (HIV) infection, antigen-specific CD8+ T cells have been extensively studied ex vivo, as they can be readily detected by HIV peptide-loaded MHC class I tetramers. In contrast, the detection of HIV-specific CD4+ T cells has proven more challenging, due to the intrinsically lower clonal expansion rates of CD4+ T cells, and to the preferential depletion of HIV-specific CD4+ T cells in the course of HIV infection. In the following protocol, we describe a simple method that facilitates the identification of CD4+ T cells specific for an HIV-1 capsid epitope using peptide-loaded MHC class II tetramers. Tetramer labeled CD4+ T cells can be analyzed for their cell surface phenotype and/or FACS-sorted for further downstream applications. A key point for successful detection of specific CD4+ T cells ex vivo is the choice of a peptide/MHC II combination that results in high-affinity T Cell Receptor (TCR) binding ( Benati et al., 2016 ). A second key point for reliable detection of MHC II tetramer-positive cells is the systematic use of a control tetramer loaded with an irrelevant peptide, with the sample and control tubes being processed in identical conditions.

12.
Mol Ther Nucleic Acids ; 5(11): e389, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27874856

ABSTRACT

The bacterial CRISPR/Cas system has proven to be an efficient tool for genetic manipulation in various organisms. Here we show the application of CRISPR-Cas9 technology to edit the human Rhodopsin (RHO) gene in a mouse model for autosomal dominant Retinitis Pigmentosa. We designed single or double sgRNAs to knock-down mutant RHO expression by targeting exon 1 of the RHO gene carrying the P23H dominant mutation. By delivering Cas9 and sgRNAs in a single plasmid we induced an efficient gene editing in vitro, in HeLa cells engineered to constitutively express the P23H mutant RHO allele. Similarly, after subretinal electroporation of the CRISPR/Cas9 plasmid expressing two sgRNAs into P23H RHO transgenic mice, we scored specific gene editing as well as significant reduction of the mutant RHO protein. Successful in vivo application of the CRISPR/Cas9 system confirms its efficacy as a genetic engineering tool in photoreceptor cells.

13.
J Clin Invest ; 126(6): 2093-108, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27111229

ABSTRACT

The rare patients who are able to spontaneously control HIV replication in the absence of therapy show signs of a particularly efficient cellular immune response. To identify the molecular determinants that underlie this response, we characterized the T cell receptor (TCR) repertoire directed at Gag293, the most immunoprevalent CD4 epitope in the HIV-1 capsid. HIV controllers from the ANRS CODEX cohort showed a highly skewed TCR repertoire that was characterized by a predominance of TRAV24 and TRBV2 variable genes, shared CDR3 motifs, and a high frequency of public clonotypes. The most prevalent public clonotypes generated TCRs with affinities at the higher end of values reported for naturally occurring TCRs. The high-affinity Gag293-specific TCRs were cross-restricted by up to 5 distinct HLA-DR alleles, accounting for the expression of these TCRs in HIV controllers of diverse genetic backgrounds. Transfer of these TCRs to healthy donor CD4+ T cells conferred high antigen sensitivity and polyfunctionality, thus recapitulating key features of the controller CD4 response. Transfer of a high-affinity Gag293-specific TCR also redirected CD8+ T cells to target HIV-1 capsid via nonconventional MHC II restriction. Together, these findings indicate that TCR clonotypes with superior functions are associated with HIV control. Amplification or transfer of such clonotypes may contribute to immunotherapeutic approaches aiming at a functional HIV cure.


Subject(s)
CD4 Antigens/immunology , HIV Infections/immunology , HIV-1 , Receptors, Antigen, T-Cell/immunology , Adoptive Transfer , Adult , Animals , Antiretroviral Therapy, Highly Active , CD4-Positive T-Lymphocytes/immunology , Genes, T-Cell Receptor alpha , Genes, T-Cell Receptor beta , HIV Infections/drug therapy , HIV Infections/genetics , HIV Long-Term Survivors , HLA-DR Antigens/genetics , Humans , Immunity, Cellular , L Cells , Mice , Middle Aged , gag Gene Products, Human Immunodeficiency Virus/immunology
14.
PLoS One ; 8(1): e55570, 2013.
Article in English | MEDLINE | ID: mdl-23383227

ABSTRACT

Idiopathic CD4 lymphocytopenia (ICL) is a rare immune deficiency characterized by a protracted CD4(+) T cell loss of unknown etiology and by the occurrence of opportunistic infections similar to those seen in AIDS. We investigated whether a defect in responses to cytokines that control CD4(+) T cell homeostasis could play a role in ICL. Immunophenotype and signaling responses to interleukin-7 (IL-7), IL-2, and thymic stromal lymphopoietin (TSLP) were analyzed by flow cytometry in CD4(+) T cells from 15 ICL patients and 15 healthy blood donors. The induction of phospho-STAT5 after IL-7 stimulation was decreased in memory CD4(+) T cells of some ICL patients, which correlated with a decreased expression of the IL-7Rα receptor chain (R = 0.74, p<0.005) and with lower CD4(+) T cell counts (R = 0.69, p<0.005). IL-2 responses were also impaired, both in the Treg and conventional memory subsets. Decreased IL-2 responses correlated with decreased IL-7 responses (R = 0.75, p<0.005), pointing to combined defects that may significantly perturb CD4(+) T cell homeostasis in a subset of ICL patients. Unexpectedly, responses to the IL-7-related cytokine TSLP were increased in ICL patients, while they remained barely detectable in healthy controls. TSLP responses correlated inversely with IL-7 responses (R = -0.41; p<0.05), suggesting a cross-regulation between the two cytokine systems. In conclusion, IL-7 and IL-2 signaling are impaired in ICL, which may account for the loss of CD4(+) T cell homeostasis. Increased TSLP responses point to a compensatory homeostatic mechanism that may mitigate defects in γc cytokine responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Homeostasis/immunology , Lymphopenia/immunology , Adolescent , Adult , Aged , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Cytokines/pharmacology , Female , Humans , Immunophenotyping , Interleukin-2/immunology , Interleukin-2/metabolism , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7/immunology , Interleukin-7/metabolism , Interleukin-7/pharmacology , Interleukin-7 Receptor alpha Subunit/metabolism , Lymphocyte Activation , Lymphopenia/metabolism , Male , Middle Aged , Phosphorylation/drug effects , STAT5 Transcription Factor/metabolism , Young Adult , Thymic Stromal Lymphopoietin
15.
J Virol ; 86(19): 10661-74, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837194

ABSTRACT

HIV controllers are rare individuals who spontaneously control HIV replication in the absence of antiretroviral therapy. To identify parameters of the CD4 response that may contribute to viral control rather than merely reflect a persistently low viremia, we compared the T helper profiles in two groups of patients with more than 10 years of viral suppression: HIV controllers from the Agence Nationale de Recherche sur le SIDA et les Hépatites Virales (ANRS) CO18 cohort (n = 26) and efficiently treated patients (n = 16). Cells specific for immunodominant Gag and cytomegalovirus (CMV) peptides were evaluated for the production of 10 cytokines and cytotoxicity markers and were also directly quantified ex vivo by major histocompatibility complex (MHC) class II tetramer staining. HIV controller CD4(+) T cells were characterized by a higher frequency of gamma interferon (IFN-γ) production, perforin(+)/CD107a(+) expression, and polyfunctionality in response to Gag peptides. While interleukin 4 (IL-4), IL-17, and IL-21 production did not differ between groups, the cells of treated patients produced more IL-10 in response to Gag and CMV peptides, pointing to persistent negative immunoregulation after long-term antiretroviral therapy. Gag293 tetramer-positive cells were detected at a high frequency (0.12%) and correlated positively with IFN-γ-producing CD4(+) T cells in the controller group (R = 0.73; P = 0.003). Tetramer-positive cells were fewer in the highly active antiretroviral therapy (HAART) group (0.04%) and did not correlate with IFN-γ production, supporting the notion of a persistent immune dysfunction in HIV-specific CD4(+) T cells of treated patients. In conclusion, HIV controllers maintained a population of highly efficient Th1 effectors directed against Gag in spite of a persistently low antigenemia, while patients treated in the long term showed a loss of CD4 effector functions.


Subject(s)
HIV Infections/blood , HIV Infections/virology , Th1 Cells/virology , ADP-ribosyl Cyclase 1/biosynthesis , Adult , Aged , Anti-Retroviral Agents/pharmacology , CD4-Positive T-Lymphocytes/cytology , Cohort Studies , Cytokines/metabolism , Flow Cytometry/methods , Gene Products, gag/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-17/metabolism , Interleukin-4/metabolism , Interleukins/metabolism , Leukocytes, Mononuclear/cytology , Lysosomal-Associated Membrane Protein 1/biosynthesis , Middle Aged , Phenotype
16.
PLoS One ; 6(12): e29899, 2011.
Article in English | MEDLINE | ID: mdl-22242145

ABSTRACT

Rai/ShcC is a member of the Shc family of protein adaptors expressed with the highest abundance in the central nervous system, where it exerts a protective function by coupling neurotrophic receptors to the PI3K/Akt survival pathway. Rai is also expressed, albeit at lower levels, in other cell types, including T and B lymphocytes. We have previously reported that in these cells Rai attenuates antigen receptor signaling, thereby impairing not only cell proliferation but also, opposite to neurons, cell survival. Here we have addressed the mechanism underlying the inhibitory activity of Rai on TCR signaling. We show that Rai interferes with the TCR signaling cascade one of the earliest steps--recruitment of the initiating kinase ZAP-70 to the phosphorylated subunit of the TCR/CD3 complex, which results in a generalized dampening of the downstream signaling events. The inhibitory activity of Rai is associated to its inducible recruitment to phosphorylated CD3, which occurs in the physiological signaling context of the immune synapse. Rai is moreover found as a pre-assembled complex with ZAP-70 and also constitutively interacts with the regulatory p85 subunit of PI3K, similar to neuronal cells, notwithstanding the opposite biological outcome, i.e. impairment of PI-3K/Akt activation. The data highlight the ability of Rai to establish interactions with the TCR and key signaling mediators which, either directly (e.g. by inhibiting ZAP-70 recruitment to the TCR or sequestering ZAP-70/PI3K in the cytosol) or indirectly (e.g. by promoting the recruitment of effectors responsible for signal extinction) prevent full triggering of the TCR signaling cascade.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , Shc Signaling Adaptor Proteins/metabolism , Signal Transduction , ZAP-70 Protein-Tyrosine Kinase/antagonists & inhibitors , Animals , CD3 Complex/metabolism , Enzyme Activation , Humans , Immunological Synapses/metabolism , Jurkat Cells , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Binding , Shc Signaling Adaptor Proteins/deficiency , Src Homology 2 Domain-Containing, Transforming Protein 3 , ZAP-70 Protein-Tyrosine Kinase/metabolism
17.
J Leukoc Biol ; 87(3): 433-42, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19892847

ABSTRACT

Besides lowering circulating cholesterol, statins act as immunomodulators. Although the effects of statins on lymphocyte activation and differentiation have been clearly defined, there is no consensus as to effects of these drugs on phagocytes. We have addressed the outcome of simvastatin treatment on the activation and effector function of human macrophages in the pathophysiologically relevant context of challenge with an opportunistic pathogen. We provide evidence that: simvastatin blocks the biological effects rapidly triggered by IgG-opsonized bacteria (phagocytosis and oxidative burst) while enhancing the delayed effects elicited by FcgammaR stimulation (production of proinflammatory mediators); these opposite effects of simvastatin result from enhancement of the JNK pathway and concomitant impairment of other signaling modules activated by FcgammaR engagement; and these activities are dependent on the capacity of simvastatin to block protein prenylation. The results provide novel mechanistic insight into the activities of statins on phagocytes and are of relevance to the assessment of potential side-effects in patients undergoing long-term hypocholesterolemic therapy.


Subject(s)
Inflammation/immunology , Macrophages/immunology , Macrophages/microbiology , Microbial Viability/drug effects , Opsonin Proteins/immunology , Simvastatin/pharmacology , Staphylococcus aureus/immunology , Actins/metabolism , Animals , Cell Line , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Humans , Inflammation/microbiology , Inflammation Mediators/metabolism , Macrophages/drug effects , Macrophages/enzymology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Mice , Phagocytosis/drug effects , Protein Prenylation/drug effects , Receptors, IgG/metabolism , Respiratory Burst/drug effects , Signal Transduction/drug effects , Staphylococcus aureus/cytology , Staphylococcus aureus/drug effects , ras Proteins/metabolism , rho GTP-Binding Proteins/metabolism
18.
J Insect Physiol ; 56(3): 253-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19879278

ABSTRACT

It is unexplained how strepsipteran insects manipulate the physiology of their hosts in order to undergo endoparasitic development without being entrapped by the innate immune defences of the host. Here we present pioneering work that aimed to explore for the first time several components of the cellular and humoral immune response among immature stages of the paper wasp Polistes dominulus, in both unparasitized insects and after infection by the strepsipteran endoparasite Xenos vesparum. We carried out hemocyte counts, phagocytosis assays in vitro and antibacterial response in vivo. On the whole, hemocyte load does not seem to be drastically affected by parasitization: a non-significant increase in hemocyte numbers was observed in parasitized wasps as respect to control, while the two dominant hemocyte types were present with similar proportions in both groups. On the other hand, phagocytosis was significantly reduced in hemocytes from parasitized wasps while the antibacterial response seemed to be less effective in control. These somewhat unexpected results are discussed, along with the implications of a multiple approach in immune response studies.


Subject(s)
Host-Parasite Interactions , Insecta/physiology , Wasps/immunology , Wasps/parasitology , Animals , Hemocytes/immunology , Immunocompetence , Larva/growth & development , Larva/immunology , Larva/parasitology , Phagocytosis , Wasps/growth & development
19.
Eur J Immunol ; 38(10): 2832-44, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18958884

ABSTRACT

Statins block the activity of HMG-CoA reductase, which catalyses the production of mevalonate, an intermediate in cholesterol biosynthesis, which is also a precursor of isoprenoids. In addition to lowering circulating cholesterol, these drugs display anti-inflammatory and immunomodulatory activities in vitro; however, their effects on the development of adaptive immune responses in vivo, as well as the underlying mechanisms, are as yet largely unknown. Here we investigated the outcome of simvastatin treatment on a number of processes, which together orchestrate adaptive immunity to specific antigen. Simvastatin treatment resulted in a marked reduction of T and B cells in spleen, lymph nodes and peripheral blood in mice. This effect could be ascribed principally to an impairment of lymphocyte homing to secondary lymphoid organs. In addition, simvastatin was found to strongly inhibit T-cell responses to the MHCI restricted hen ovalbumin peptide antigen SIINFEKL and to impair ovalbumin uptake and cross-presentation by MHCI. Simvastatin also suppressed antibody responses to immunization with ovalbumin and delayed-type hypersensitivity to allergens. These activities could be largely accounted for by the simvastatin-dependent inhibition of HMG-CoA reductase. The data provide novel mechanistic insight into the activities of simvastatin in the highly complex context of the immune response.


Subject(s)
Antibody Formation/drug effects , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/drug effects , Immunity, Cellular/drug effects , Lymphocyte Activation/drug effects , Simvastatin/pharmacology , Animals , Antigen Presentation/drug effects , B-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mice
20.
Curr Med Chem ; 15(12): 1154-65, 2008.
Article in English | MEDLINE | ID: mdl-18473810

ABSTRACT

The Src family consists of eight non-receptor protein tyrosine kinases characterised by a common structure. Based on their amino acid sequence, Src family kinases are grouped into two subfamilies, which are also characterised by different tissue specificity. Src kinases are involved in signal transduction pathways triggered by a wide variety of surface receptors, including receptor tyrosine kinases, integrins, G-protein-coupled receptors and antigen receptors. Several pieces of evidence implicate Src family kinases in cancer development, as a consequence of changes in protein expression and/or kinase activity, and have prompted the design of potent specific inhibitors, the most common of which are adenine mimetics, as tools of relevant clinical interest for the treatment of both solid tumours and leukaemias. In addition, the finding that some Src kinases expressed in haematopoietic cells play pivotal roles in lymphocyte maturation and activation has fostered the development of safe and effective inhibitors selective for specific Src family members, which are currently being tested in clinical trials as immunosuppressants for the treatment of immunological disorders. Here we shall review the recent literature on the involvement of Src family kinases in human neoplasias and immunological disorders and the goals reached in the search for selective pharmacological inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Immune System Diseases/drug therapy , Neoplasms/drug therapy , src-Family Kinases/drug effects , Animals , Antineoplastic Agents/therapeutic use , Catalysis , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Immune System Diseases/enzymology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors , Neoplasms/enzymology , Receptors, Cell Surface/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...