Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Viruses ; 14(9)2022 08 30.
Article in English | MEDLINE | ID: mdl-36146734

ABSTRACT

In people living with HIV, Mycobacterium tuberculosis (Mtb) is the major cause of death. Due to the increased morbidity/mortality in co-infection, further research is urgently required. A limiting factor to research in HIV and HIV/Mtb co-infection is the lack of accessible in vivo models. Next-generation humanized mice expressing HLA transgenes report improved human immune reconstitution and functionality, which may better recapitulate human disease. This study compares well-established huNRG mice and next-generation HLA I/II-transgenic (huDRAG-A2) mice for immune reconstitution, disease course, and pathology in HIV and TB. HuDRAG-A2 mice have improved engraftment of key immune cell types involved in HIV and TB disease. Upon intravaginal HIV-1 infection, both models developed significant HIV target cell depletion in the blood and tissues. Upon intranasal Mtb infection, both models sustained high bacterial load within the lungs and tissue dissemination. Some huDRAG-A2 granulomas appeared more classically organized, characterized by focal central necrosis, multinucleated giant cells, and foamy macrophages surrounded by a halo of CD4+ T cells. HIV/Mtb co-infection in huNRG mice trended towards worsened TB pathology and showed potential for modeling co-infection. Both huNRG and huDRAG-A2 mice are viable options for investigating HIV and TB, but the huDRAG-A2 model may offer advantages.


Subject(s)
Coinfection , HIV Infections , Mycobacterium tuberculosis , Tuberculosis , Animals , CD4-Positive T-Lymphocytes , Disease Models, Animal , Humans , Mice
2.
BMC Vet Res ; 17(1): 168, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33858420

ABSTRACT

BACKGROUND: Equine infectious anemia (EIA) is a viral disease, caused by the Equine Infectious Anemia virus (EIAV) belonging to the Retroviridae family, genus Lentivirus. Horses (or equids) infected with EIAV are lifelong carriers and they remain contagious for other horses even in the absence of clinical signs. So far, EIAV infection has been reported among horses in North and South America, France, Germany, Italy, Hungary and Romania, with no publication regarding the presence of EIAV in horses in Serbia. To determine the circulation of EIAV among, approximately, the 5000 horses of the Vojvodina region, northern part of Serbia, 316 serum undergone serological testing for EIA. Then, identification and full genome sequencing using next generation sequencing was performed from one EIA positive horse. RESULTS: the 316 sera were tested with 3 different commercial agar gel immunodiffusion (AGID) tests and two different commercial enzyme-linked immunosorbent assay (ELISA). With the three AGID kits, 311 (98.4%) among the 316 tested sera were negative and only five (1.6%) sera were positive for EIA. Some discrepancies were seen for the two ELISA kits tested since one exhibited the same results as AGID test and the second gave 295 sera with negative results, five with a positive result and 16 with doubtful outcome. Phylogenetic analysis performed using the full genome sequence showed that EIAV characterized from a horse in Serbia is different from those identify so fare around the world and form a distinct and separate group together with another EIAV strain. CONCLUSIONS: This study demonstrate for the first time that EIAV is circulating at a low level in the horse population from the Northern part of Serbia. Interestingly, phylogenetic data indicates that this EIAV from the western Balkan region of Europe belongs to a new cluster.


Subject(s)
Equine Infectious Anemia/epidemiology , Infectious Anemia Virus, Equine/genetics , Infectious Anemia Virus, Equine/isolation & purification , Animals , Enzyme-Linked Immunosorbent Assay/veterinary , Equine Infectious Anemia/virology , Genome, Viral , Horses , Infectious Anemia Virus, Equine/classification , Phylogeny , Serbia/epidemiology , Seroepidemiologic Studies
3.
Sci Rep ; 11(1): 3894, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594113

ABSTRACT

The progestin-based hormonal contraceptive Depot Medroxyprogesterone Acetate (DMPA) is widely used in sub-Saharan Africa, where HIV-1 is endemic. Meta-analyses have shown that women using DMPA are 40% more likely than women not using hormonal contraceptives to acquire Human Immunodeficiency Virus (HIV-1). Therefore understanding how DMPA increases susceptibility to HIV-1 is an important public health issue. Using C57BL/6 mice and our previously optimized humanized mouse model (NOD-Rag1tm1Mom Il2rgtm1Wjl transplanted with hCD34-enriched hematopoietic stem cells; Hu-mice) where peripheral blood and tissues are reconstituted by human immune cells, we assessed how DMPA affected mucosal barrier function, HIV-1 susceptibility, viral titres, and target cells compared to mice in the diestrus phase of the estrous cycle, when endogenous progesterone is highest. We found that DMPA enhanced FITC-dextran dye leakage from the vaginal tract into the systemic circulation, enhanced target cells (hCD68+ macrophages, hCD4+ T cells) in the vaginal tract and peripheral blood (hCD45+hCD3+hCD4+hCCR5+ T cells), increased the rate of intravaginal HIV-1 infection, extended the window of vulnerability, and lowered vaginal viral titres following infection. These findings suggest DMPA may enhance susceptibility to HIV-1 in Hu-mice by impairing the vaginal epithelial barrier, increasing vaginal target cells (including macrophages), and extending the period of time during which Hu-mice are susceptible to infection; mechanisms that might also affect HIV-1 susceptibility in women.


Subject(s)
Contraceptive Agents, Hormonal/adverse effects , HIV-1 , Host-Pathogen Interactions/drug effects , Medroxyprogesterone Acetate/adverse effects , Vagina/drug effects , Animals , Cytokines/metabolism , Delayed-Action Preparations , Disease Susceptibility/chemically induced , Female , Humans , Infant, Newborn , Macrophages , Mice , Mice, Inbred C57BL , Vagina/immunology , Vagina/metabolism , Vagina/virology
4.
Dis Model Mech ; 12(10)2019 10 23.
Article in English | MEDLINE | ID: mdl-31537512

ABSTRACT

The hormonal contraceptive medroxyprogesterone acetate (MPA) is associated with increased risk of human immunodeficiency virus (HIV), via incompletely understood mechanisms. Increased diversity in the vaginal microbiota modulates genital inflammation and is associated with increased HIV-1 acquisition. However, the effect of MPA on diversity of the vaginal microbiota is relatively unknown. In a cohort of female Kenyan sex workers, negative for sexually transmitted infections (STIs), with Nugent scores <7 (N=58 of 370 screened), MPA correlated with significantly increased diversity of the vaginal microbiota as assessed by 16S rRNA gene sequencing. MPA was also significantly associated with decreased levels of estrogen in the plasma, and low vaginal glycogen and α-amylase, factors implicated in vaginal colonization by lactobacilli, bacteria that are believed to protect against STIs. In a humanized mouse model, MPA treatment was associated with low serum estrogen, low glycogen and enhanced HIV-1 susceptibility. The mechanism by which the MPA-mediated changes in the vaginal microbiota may contribute to HIV-1 susceptibility in humans appears to be independent of inflammatory cytokines and/or activated T cells. Altogether, these results suggest MPA-induced hypo-estrogenism may alter key metabolic components that are necessary for vaginal colonization by certain bacterial species including lactobacilli, and allow for greater bacterial diversity in the vaginal microbiota.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cellular Microenvironment , HIV-1/physiology , Medroxyprogesterone Acetate/adverse effects , Microbiota/drug effects , Vagina/microbiology , Adult , Animals , Bacteria/drug effects , Biodiversity , Contraception , Cytokines/metabolism , Estrogens/metabolism , Female , Glycogen/metabolism , HIV-1/drug effects , Humans , Inflammation Mediators/metabolism , Kenya , Mice , Models, Biological , Sex Workers , Vagina/drug effects , Vagina/metabolism , Young Adult , alpha-Amylases/metabolism
5.
Virology ; 537: 121-129, 2019 11.
Article in English | MEDLINE | ID: mdl-31493650

ABSTRACT

Equine infectious anemia virus (EIAV) is responsible of acute disease episodes characterized by fever, anemia, thrombocytopenia and anorexia in equids. The high mutation rate in EIAV genome limited the number of full genome sequences availability. In the present study, we used the SureSelect target enrichment system with Illumina Next Generation Sequencing to characterize the proviral DNA of Equine Infectious Anemia Virus (EIAV) from asymptomatic horses. This approach allows a direct sequencing of the EIAV whole genome without cloning or amplification steps and we could obtain for the first time the complete genomic DNA sequences of French EIAV strains. We analyzed their phylogenetic relationship and genetic variability by comparison with 17 whole EIAV genome sequences from different parts of the world. The results obtained provide new insights into the molecular detection of EIAV and genetic diversity of European viral strains.


Subject(s)
Equine Infectious Anemia/virology , Genetic Variation , High-Throughput Nucleotide Sequencing , Infectious Anemia Virus, Equine/classification , Infectious Anemia Virus, Equine/genetics , Animals , Asymptomatic Diseases , France , Horses , Infectious Anemia Virus, Equine/isolation & purification , Phylogeny , Proviruses/classification , Proviruses/genetics , Proviruses/isolation & purification , Whole Genome Sequencing
6.
J Virol ; 93(7)2019 04 01.
Article in English | MEDLINE | ID: mdl-30674627

ABSTRACT

The molecular basis for HIV-1 susceptibility in primary human monocyte-derived macrophages (MDMs) was previously evaluated by comparing the transcriptome of infected and bystander populations. Careful analysis of the data suggested that the ubiquitin ligase MDM2 acted as a positive regulator of HIV-1 replication in MDMs. In this study, MDM2 silencing through transcript-specific small interfering RNAs in MDMs induced a reduction in HIV-1 reverse transcription and integration along with an increase in the expression of p53-induced genes, including CDKN1A Experiments with Nutlin-3, a pharmacological inhibitor of MDM2 p53-binding activity, showed a similar effect on HIV-1 infection, suggesting that the observed restriction in HIV-1 production results from the release/activation of p53 and not the absence of MDM2 per se Knockdown and inhibition of MDM2 also both correlate with a decrease in the Thr592-phosphorylated inactive form of SAMHD1. The expression level of MDM2 and the p53 activation status are therefore important factors in the overall susceptibility of macrophages to HIV-1 infection, bringing a new understanding of signaling events controlling the process of virus replication in this cell type.IMPORTANCE Macrophages, with their long life span in vivo and their resistance to HIV-1-mediated cytopathic effect, might serve as viral reservoirs, contributing to virus persistence in an infected individual. Identification of host factors that increase the overall susceptibility of macrophages to HIV-1 might provide new therapeutic targets for the efficient control of viral replication in these cells and limit the formation of reservoirs in exposed individuals. In this study, we demonstrate the importance of p53 regulation by MDM2, which creates a cellular environment more favorable to the early steps of HIV-1 replication. Moreover, we show that p53 stabilization reduces virus infection in human macrophages, highlighting the important role of p53 in antiviral immunity.


Subject(s)
HIV Infections/genetics , HIV-1/pathogenicity , Macrophages/metabolism , Macrophages/virology , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics , HEK293 Cells , HIV Infections/metabolism , HIV Infections/virology , Humans , Phosphorylation/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Reverse Transcription/genetics , Tumor Suppressor Protein p53/metabolism , Virus Replication/genetics
7.
Glia ; 66(7): 1363-1381, 2018 07.
Article in English | MEDLINE | ID: mdl-29464785

ABSTRACT

The "shock and kill" HIV-1 cure strategy proposes eradication of stable cellular reservoirs by clinical treatment with latency-reversing agents (LRAs). Although resting CD4+ T cells latently infected with HIV-1 constitute the main reservoir that is targeted by these approaches, their consequences on other reservoirs such as the central nervous system are still unknown and should be taken into consideration. We performed experiments aimed at defining the possible role of astrocytes in HIV-1 persistence in the brain and the effect of LRA treatments on this viral sanctuary. We first demonstrate that the diminished HIV-1 production in a proliferating astrocyte culture is due to a reduced proliferative capacity of virus-infected cells compared with uninfected astrocytes. In contrast, infection of non-proliferating astrocytes led to a robust HIV-1 infection that was sustained for over 60 days. To identify astrocytes latently infected with HIV-1, we designed a new dual-color reporter virus called NL4.3 eGFP-IRES-Crimson that is fully infectious and encodes for all viral proteins. Although we detected a small fraction of astrocytes carrying silent HIV-1 proviruses, we did not observe any reactivation using various LRAs and even strong inducers such as tumor necrosis factor, thus suggesting that these proviruses were either not transcriptionally competent or in a state of deep latency. Our findings imply that astrocytes might not constitute a latent reservoir per se but that relentless virus production by this brain cell population could contribute to the neurological disorders seen in HIV-1-infected persons subjected to combination antiretroviral therapy.


Subject(s)
Astrocytes/physiology , Astrocytes/virology , HIV Infections/physiopathology , HIV-1/physiology , Astrocytes/pathology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/virology , Cell Proliferation , Cells, Cultured , Coculture Techniques , HEK293 Cells , HIV-1/genetics , Humans , Virus Latency
8.
Sci Rep ; 7(1): 15263, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127409

ABSTRACT

Approximately 40% of HIV-1 infections occur in the female genital tract (FGT), primarily through heterosexual transmission. FGT factors determining outcome of HIV-1 exposure are incompletely understood, limiting prevention strategies. Here, humanized NOD-Rag1-/- γc-/- mice differentially reconstituted with human CD34+ -enriched hematopoietic stem cells (Hu-mice), were used to assess target cell frequency and viral inoculation dose as determinants of HIV-1 infection following intravaginal (IVAG) challenge. Results revealed a significant correlation between HIV-1 susceptibility and hCD45+ target cells in the blood, which correlated with presence of target cells in the FGT, in the absence of local inflammation. HIV-1 plasma load was associated with viral dose at inoculation and frequency of target cells. Events following IVAG HIV-1 infection; viral dissemination and CD4 depletion, were not affected by these parameters. Following IVAG inoculation, HIV-1 titres peaked, then declined in vaginal lavage while plasma showed a reciprocal pattern. The greatest frequency of HIV-1-infected (p24+) cells were found one week post-infection in the FGT versus blood and spleen, suggesting local viral amplification. Five weeks post-infection, HIV-1 disseminated into systemic tissues, in a dose-dependent manner, followed by depletion of hCD45+ CD3+ CD4+ cells. Results indicate target cell frequency in the Hu-mouse FGT is a key determinant of HIV-1 infection, which might provide a useful target for prophylaxis in women.


Subject(s)
HIV Infections/transmission , HIV-1/metabolism , Leukocyte Common Antigens/metabolism , Vagina/metabolism , Viral Load , Animals , Disease Models, Animal , Female , HIV Infections/immunology , HIV Infections/metabolism , HIV-1/immunology , Humans , Leukocyte Common Antigens/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Vagina/immunology , Vagina/virology
9.
Cell Rep ; 21(1): 141-153, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978468

ABSTRACT

Macrophages are heterogeneous immune cells with distinct origins, phenotypes, functions, and tissue localization. Their susceptibility to HIV-1 is subject to variations from permissiveness to resistance, owing in part to regulatory microRNAs. Here, we used RNA sequencing (RNA-seq) to examine the expression of >400 microRNAs in productively infected and bystander cells of HIV-1-exposed macrophage cultures. Two microRNAs upregulated in bystander macrophages, miR-221 and miR-222, were identified as negative regulators of CD4 expression and CD4-mediated HIV-1 entry. Both microRNAs were enhanced by tumor necrosis factor alpha (TNF-α), an inhibitor of CD4 expression. MiR-221/miR-222 inhibitors recovered HIV-1 entry in TNF-α-treated macrophages by enhancing CD4 expression and increased HIV-1 replication and spread in macrophages by countering TNF-α-enhanced miR-221/miR-222 expression in bystander cells. In line with these findings, HIV-1-resistant intestinal myeloid cells express higher levels of miR-221 than peripheral blood monocytes. Thus, miR-221/miR-222 act as effectors of the antiviral host response activated during macrophage infection that restrict HIV-1 entry.


Subject(s)
CD4 Antigens/genetics , HIV-1/genetics , Host-Pathogen Interactions , Macrophages/virology , MicroRNAs/genetics , Bystander Effect , CD4 Antigens/metabolism , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , HIV-1/growth & development , HIV-1/metabolism , Humans , Macrophages/drug effects , Macrophages/metabolism , MicroRNAs/metabolism , Primary Cell Culture , Sequence Analysis, RNA , Signal Transduction , THP-1 Cells , Tumor Necrosis Factor-alpha/pharmacology , Virus Replication
10.
Sci Rep ; 7(1): 5238, 2017 07 12.
Article in English | MEDLINE | ID: mdl-28701698

ABSTRACT

It has been proposed that macrophages could serve as long-lived compartments for HIV-1 infection under in vivo situations because these cells are resistant to the virus-mediated cytopathic effect, produce progeny virus over extended periods of time and are localized in tissues that are often less accessible by treatment. Comprehensive experimental studies are thus needed to characterize the HIV-1-induced modulation of host genes in these myeloid lineage cells. To shed light on this important issue, we performed comparative analyses of mRNA expression levels of host genes in uninfected bystander and HIV-1-infected human macrophages using an infectious reporter virus construct coupled with a large-scale RNA sequencing approach. We observed a rapid differential expression of several host factors in the productively infected macrophage population including genes regulating DNA replication factors and chromatin remodeling. A siRNA-mediated screening study to functionally identify host determinants involved in HIV-1 biology has provided new information on the virus molecular regulation in macrophages.


Subject(s)
Chromatin/metabolism , DNA/metabolism , Gene Expression Regulation , HIV Infections/virology , HIV-1/physiology , Host-Pathogen Interactions/physiology , Macrophages/metabolism , Transcriptome , Biomarkers/analysis , Chromatin/genetics , DNA/genetics , HIV Infections/genetics , HIV Infections/pathology , Humans , Macrophages/cytology , Macrophages/virology , Virus Replication
11.
J Immunol ; 196(9): 3806-17, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27022194

ABSTRACT

HIV-1 infection is characterized by persistent viral replication, chronic immune activation, and CD4(+) T cell depletion. Moreover, several immune dysfunctions are observed in cells that are not targeted by the virus, such as B cells. Some B cell abnormalities include hypergammaglobulinemia, nonspecific B cell activation, class switching, increased cell turnover, breakage of tolerance, and a loss of the capacity to generate and maintain memory. Several cytokines and growth factors that are increased in the serum of HIV-1-infected individuals have been suggested to directly or indirectly trigger B cell activation, and one of these is BAFF. In this study, we investigate the ability of fully competent (R5-tropic) HIV-1 to induce BAFF production by monocyte-derived macrophages (MDMs). We demonstrate here that HIV-1 drives BAFF production in MDMs in a type-I IFN- and TLR-independent manner. Moreover, we determine that HIV-1 Nef accessory protein is dispensable in BAFF upregulation as a nef-deleted HIV-1 strain is still able to increase BAFF at levels similar to the wild type strain. Finally, we show that the macrophage phenotype status affects HIV-1 replication and BAFF induction, as both were abrogated in MDMs displaying a M1 phenotype. This study provides new useful information about the increased levels of BAFF observed during HIV-1 infection and highlights the importance of macrophages as a source of BAFF, a phenomenon that might contribute to B cell dysfunctions at inflammatory tissue sites in infected individuals.


Subject(s)
B-Cell Activating Factor/metabolism , HIV Infections/immunology , HIV-1/immunology , Macrophages/immunology , nef Gene Products, Human Immunodeficiency Virus/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Endosomes/metabolism , Humans , Interferon Type I/metabolism , Macrophages/virology , Phenotype , RNA, Small Interfering/genetics , Th1 Cells/immunology , Toll-Like Receptors/metabolism , nef Gene Products, Human Immunodeficiency Virus/genetics
12.
Proc Natl Acad Sci U S A ; 109(5): 1506-11, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22307605

ABSTRACT

The organization of cells into epithelium depends on cell interaction with both the extracellular matrix (ECM) and adjacent cells. The role of cell-cell adhesion in the regulation of epithelial topology is well-described. ECM is better known to promote cell migration and provide a structural scaffold for cell anchoring, but its contribution to multicellular morphogenesis is less well-understood. We developed a minimal model system to investigate how ECM affects the spatial organization of intercellular junctions. Fibronectin micropatterns were used to constrain the location of cell-ECM adhesion. We found that ECM affects the degree of stability of intercellular junction positioning and the magnitude of intra- and intercellular forces. Intercellular junctions were permanently displaced, and experienced large perpendicular tensional forces as long as they were positioned close to ECM. They remained stable solely in regions deprived of ECM, where they were submitted to lower tensional forces. The heterogeneity of the spatial organization of ECM induced anisotropic distribution of mechanical constraints in cells, which seemed to adapt their position to minimize both intra- and intercellular forces. These results uncover a morphogenetic role for ECM in the mechanical regulation of cells and intercellular junction positioning.


Subject(s)
Extracellular Matrix/physiology , Intercellular Junctions/physiology , Cell Line, Tumor , Humans
13.
Mol Cell Biochem ; 356(1-2): 11-20, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21755461

ABSTRACT

Protein kinase CK2 participates in the regulation of fundamental cellular processes. Among these processes, cell polarity and cell morphology are controlled by this enzyme probably through the phosphorylation of key proteins. To further study the involvement of CK2 in these processes, we showed that in epithelial cells, the regulatory CK2ß subunit was required for LKB1-dependent polarization and cell adhesion. Moreover, CK2ß silencing in MCF10A mammary epithelial cells triggered changes in their morphology correlated with the acquisition of mesenchymal phenotype, which were reminiscent to TGFß-induced epithelial-to-mesenchymal-transition (EMT). TGFß has emerged as a major inducer of EMT both in vitro and in vivo. We found that among the TGFß isoforms, TGFß2 expression was strongly induced in CK2ß-knockdown cells. However, the EMT phenotype induced in response to CK2ß silencing was not abolished by blocking the TGFß signaling pathway at TGFß receptor level, suggesting that alternative pathways might be involved. Given the importance of CK2 in tumorigenesis, a dysregulation of CK2ß expression might contribute to EMT induction during cancer progression.


Subject(s)
Casein Kinase II/metabolism , Epithelial-Mesenchymal Transition , Animals , Cell Adhesion , Cell Line , Cell Polarity , Cell Shape , Epithelial Cells/cytology , Epithelial Cells/enzymology , Gene Knockdown Techniques , Humans , Mice , NIH 3T3 Cells , Phenotype , RNA, Small Interfering/metabolism , Transforming Growth Factor beta2/metabolism , Up-Regulation
14.
Proc Natl Acad Sci U S A ; 108(28): 11464-9, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21709252

ABSTRACT

The intracellular localization and shape of the nucleus plays a central role in cellular and developmental processes. In fibroblasts, nuclear movement and shape are controlled by a specific perinuclear actin network made of contractile actin filament bundles called transmembrane actin-associated nuclear (TAN) lines that form a structure called the actin cap. The identification of regulatory proteins associated with this specific actin cytoskeletal dynamic is a priority for understanding actin-based changes in nuclear shape and position in normal and pathological situations. Here, we first identify a unique family of actin regulators, the refilin proteins (RefilinA and RefilinB), that stabilize specifically perinuclear actin filament bundles. We next identify the actin-binding filamin A (FLNA) protein as the downstream effector of refilins. Refilins act as molecular switches to convert FLNA from an actin branching protein into one that bundles. In NIH 3T3 fibroblasts, the RefilinB/FLNA complex organizes the perinuclear actin filament bundles forming the actin cap. Finally, we demonstrate that in epithelial normal murine mammary gland (NmuMG) cells, the RefilinB/FLNA complex controls formation of a new perinuclear actin network that accompanies nuclear shape changes during the epithelial-mesenchymal transition (EMT). Our studies open perspectives for further functional analyses of this unique actin-based network and shed light on FLNA function during development and in human syndromes associated with FLNA mutations.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Cell Nucleus/metabolism , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Amino Acid Sequence , Animals , Astrocytoma/metabolism , Astrocytoma/ultrastructure , Base Sequence , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Nucleus/ultrastructure , Dimerization , Epithelial-Mesenchymal Transition , Female , Filamins , Humans , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Microscopy, Electron, Transmission , Molecular Sequence Data , Multiprotein Complexes , NIH 3T3 Cells , Protein Interaction Domains and Motifs , RNA, Small Interfering/genetics , Sequence Deletion
15.
Mol Cell Biochem ; 316(1-2): 107-13, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18587631

ABSTRACT

There is increasing evidence that protein kinase CK2 is involved, among a wide variety of cellular processes, in the maintenance of mammalian cell morphology and cell polarity. Here, we show that in epithelial cells, a fraction of CK2 is associated to the plasma membrane and that this localization is controlled by cell-matrix interactions. In addition, inhibition of CK2 activity in mammary epithelial cells (MCF10A), using either the specific CK2 inhibitor TBB or siRNA-mediated CK2beta knockdown, induced differential phenotypes revealing an important role of this enzyme in epithelial cell morphology.


Subject(s)
Casein Kinase II/metabolism , Cell Polarity , Epithelial Cells/cytology , Epithelial Cells/enzymology , Animals , Casein Kinase II/antagonists & inhibitors , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Polarity/drug effects , Dogs , Epithelial Cells/drug effects , Humans , Mice , Phenotype , Protein Kinase Inhibitors/pharmacology , Protein Subunits/metabolism , Protein Transport/drug effects , Subcellular Fractions/drug effects , Subcellular Fractions/enzymology
16.
Biochem J ; 408(3): 363-73, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17714077

ABSTRACT

X-ray crystallography studies, as well as live-cell fluorescent imaging, have recently challenged the traditional view of protein kinase CK2. Unbalanced expression of catalytic and regulatory CK2 subunits has been observed in a variety of tissues and tumours. Thus the potential intersubunit flexibility suggested by these studies raises the likely prospect that the CK2 holoenzyme complex is subject to disassembly and reassembly. In the present paper, we show evidence for the reversible multimeric organization of the CK2 holoenzyme complex in vitro. We used a combination of site-directed mutagenesis, binding experiments and functional assays to show that, both in vitro and in vivo, only a small set of primary hydrophobic residues of CK2beta which contacts at the centre of the CK2alpha/CK2beta interface dominates affinity. The results indicate that a double mutation in CK2beta of amino acids Tyr188 and Phe190, which are complementary and fill up a hydrophobic pocket of CK2alpha, is the most disruptive to CK2alpha binding both in vitro and in living cells. Further characterization of hotspots in a cluster of hydrophobic amino acids centred around Tyr188-Phe190 led us to the structure-based design of small-peptide inhibitors. One conformationally constrained 11-mer peptide (Pc) represents a unique CK2beta-based small molecule that was particularly efficient (i) to antagonize the interaction between the CK2 subunits, (ii) to inhibit the assembly of the CK2 holoenzyme complex, and (iii) to strongly affect its substrate preference.


Subject(s)
Casein Kinase II/antagonists & inhibitors , Peptides/pharmacology , Protein Kinase Inhibitors/pharmacology , Amino Acid Sequence , Base Sequence , Casein Kinase II/chemistry , Casein Kinase II/genetics , Casein Kinase II/metabolism , Catalysis , Crystallography, X-Ray , DNA Primers , HeLa Cells , Humans , Ligands , Mutagenesis, Site-Directed , Peptides/chemistry , Peptides/metabolism , Protein Conformation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL