Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Biomedicines ; 11(6)2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37371814

ABSTRACT

Actinic keratosis (AK) is a carcinoma in situ precursor of cutaneous squamous cell carcinoma (cSCC), the second most common cancer affecting the Caucasian population. AK is frequently present in the sun-exposed skin of the elderly population, UV radiation being the main cause of this cancer, and other risk factors contributing to AK incidence. The dysregulation of microRNAs (miRNAs) observed in different cancers leads to an improper expression of miRNA targets involved in several cellular pathways. The TaqMan Array Human MicroRNA Card assay for miRNA expression profiling was performed in pooled AK compared to healthy skin scraping samples from the same patients. Forty-three miRNAs were modulated in the AK samples. The expression of miR-19b (p < 0.05), -31, -34a (p < 0.001), -126, -146a (p < 0.01), -193b, and -222 (p < 0.05) was validated by RT-qPCR. The MirPath tool was used for MiRNA target prediction and enriched pathways. The top DIANA-mirPath pathways regulated by the targets of the 43 miRNAs are TGF-beta signaling, Proteoglycans in cancer, Pathways in cancer, and Adherens junction (7.30 × 10-10 < p < 1.84 × 10-8). Selected genes regulating the KEGG pathways, i.e., TP53, MDM2, CDKN1A, CDK6, and CCND1, were analyzed. MiRNAs modulated in AK regulate different pathways involved in tumorigenesis, indicating miRNA regulation as a critical step in keratinocyte cancer.

2.
Front Immunol ; 11: 2129, 2020.
Article in English | MEDLINE | ID: mdl-33072075

ABSTRACT

Macrophages are key targets of human immunodeficiency virus type 1 (HIV-1) infection and main producers of the proinflammatory chemokine CC chemokine ligand 2 (CCL2), whose expression is induced by HIV-1 both in vitro and in vivo. We previously found that CCL2 neutralization in monocyte-derived macrophages (MDMs) strongly inhibited HIV-1 replication affecting post-entry steps of the viral life cycle. Here, we used RNA-sequencing to deeply characterize the cellular factors and pathways modulated by CCL2 blocking in MDMs and involved in HIV-1 replication restriction. We report that exposure to CCL2 neutralizing antibody profoundly affected the MDM transcriptome. Functional annotation clustering of up-regulated genes identified two clusters enriched for antiviral defense and immune response pathways, comprising several interferon-stimulated, and restriction factor coding genes. Transcripts in the clusters were enriched for RELA and NFKB1 targets, suggesting the activation of the canonical nuclear factor κB pathway as part of a regulatory network involving miR-155 up-regulation. Furthermore, while HIV-1 infection caused small changes to the MDM transcriptome, with no evidence of host defense gene expression and type I interferon signature, CCL2 blocking enabled the activation of a strong host innate response in infected macrophage cultures, and potently inhibited viral genes expression. Notably, an inverse correlation was found between levels of viral transcripts and of the restriction factors APOBEC3A (apolipoprotein B mRNA editing enzyme catalytic polypeptide-like 3 A), ISG15, and MX1. These findings highlight an association between activation of innate immune pathways and HIV-1 restriction upon CCL2 blocking and identify this chemokine as an endogenous factor contributing to the defective macrophage response to HIV-1. Therapeutic targeting of CCL2 may thus strengthen host innate immunity and restrict HIV-1 replication.


Subject(s)
Antibodies, Neutralizing/pharmacology , Chemokine CCL2/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Viral/drug effects , HIV-1/genetics , Immunity, Innate , Macrophages/metabolism , Antibodies, Neutralizing/immunology , Antibody Specificity , Cells, Cultured , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/immunology , Cytidine Deaminase/physiology , Datasets as Topic , Humans , Macrophages/drug effects , Macrophages/immunology , Macrophages/virology , MicroRNAs/biosynthesis , MicroRNAs/genetics , Molecular Sequence Annotation , NF-kappa B/metabolism , Proteins/physiology , RNA, Viral/biosynthesis , RNA, Viral/genetics , RNA-Seq , Real-Time Polymerase Chain Reaction , Virus Latency , Virus Replication
3.
Papillomavirus Res ; 9: 100196, 2020 06.
Article in English | MEDLINE | ID: mdl-32222599

ABSTRACT

Actinic keratosis (AK) arises on photo-damaged skin and is considered to be the precursor lesion of cutaneous squamous cell carcinoma (cSCC). Many findings support the involvement of ß human papillomaviruses (HPVs) in cSCC, while very little is known on γ HPV types. The objective of this study was to characterize the spectrum of PV types in healthy skin (HS) and AK samples of the same immunocompetent individuals using next generation sequencing (NGS). Viral DNA of 244 AK and 242 HS specimens were amplified by PCR using two different sets of primers (FAP59/64 and FAPM1). Purified amplicons were pooled and sequenced using NGS. The study resulted in the identification of a large number of known ß and γ PV types. In addition, 27 putative novel ß and 16 γ and 4 unclassified PVs were isolated. HPV types of species γ-1 (e.g. HPV4) appeared to be strongly enriched in AK versus HS. The NGS analysis revealed that a large spectrum of known and novel PVs is present in HS and AK. The evidence that species γ-1 HPV types appears to be enriched in AK in comparison to HS warrants further studies to evaluate their role in development of skin (pre)cancerous lesions.


Subject(s)
Alphapapillomavirus/genetics , High-Throughput Nucleotide Sequencing , Keratosis, Actinic/virology , Papillomavirus Infections/diagnosis , Skin/virology , Aged , Aged, 80 and over , Alphapapillomavirus/classification , Alphapapillomavirus/isolation & purification , DNA, Viral/genetics , Female , Humans , Immunocompetence , Male , Middle Aged , Papillomavirus Infections/virology , Sequence Analysis, DNA , Skin/pathology
4.
Cytokine Growth Factor Rev ; 51: 92-98, 2020 02.
Article in English | MEDLINE | ID: mdl-31973992

ABSTRACT

A small group of mucosal Human Papillomaviruses are the causative agents of cervical cancer and are also associated with other types of cancers. Certain cutaneous Human Papillomaviruses seem to have a role as co-factors in the UV-induced carcinogenesis of the skin. The main mechanism of the tumorigenesis induced by Human Papillomaviruses is linked to the transforming activity of the viral E6 and E7 oncoproteins. However, other mechanisms, such as the gene expression control by specific microRNAs expression and deregulation of immune inflammatory mediators, may be important in the process of transformation. In this context, the release of Extracellular Vesicles with a specific cargo (microRNAs involved in tumorigenesis, mRNAs of viral oncoproteins, cytokines, chemokines) appears to play a key role.


Subject(s)
Alphapapillomavirus/pathogenicity , Carcinogenesis/pathology , Cell Communication , Extracellular Vesicles/physiology , Papillomavirus Infections/complications , Carcinoma, Squamous Cell/virology , Extracellular Vesicles/pathology , Female , Humans , MicroRNAs , RNA, Messenger , Skin/pathology , Skin/virology , Uterine Cervical Neoplasms/virology
5.
Cytokine ; 106: 182-189, 2018 06.
Article in English | MEDLINE | ID: mdl-29137858

ABSTRACT

The connection between chronic inflammation and risk of cancer has been supported by several studies. The development of cancer might be a process driven by the presence of a specific combination of inflammatory mediators, including cytokines, chemokines and enzymes, in the tumor microenvironment. Virus-induced tumors, like HPV-induced Squamous Cell Carcinomas, represent a paradigmatic example of the interplay between inflammation, as integral part of the innate antiviral response, and malignant transformation. Here, the role of inflammatory microenvironment in the HPV-induced carcinogenesis is addressed, with a specific focus on the involvement of the immune molecules as well as their delivery through the microvesicle cargo possibly correlated to the different HPV genotype. The expression of the inflammatory mediators in HPV positive cells has been analyzed in primary human foreskin keratinocytes and keratinocytes transduced by E6 and E7 from mucosal HPV-16 or cutaneous HPV-38 genotypes. HPV E6 and E7 proteins can modulate the expression of immune mediators in HPV-infected cells and can affect the levels of immune molecules, mainly chemokines, in the extracellular milieu. HPV-16 E6 and E7 oncoproteins have been silenced to confirm the specificity of the modulation of the inflammatory microenvironment. Our results suggest that the expression of HPV oncoproteins allows the modification of the tumor milieu through the synthesis and release of specific pro-inflammatory cytokines and chemokines, affecting the efficacy of the immune response. The microenvironment can also be conditioned by an altered mRNA cargo delivered by extracellular vesicles, thereby efficiently affecting the surrounding cells with possible implication for tumorigenesis and tumor diagnosis.


Subject(s)
Cellular Microenvironment , Extracellular Vesicles/metabolism , Inflammation Mediators/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Cell Line , Gene Silencing , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Cytokine ; 89: 235-238, 2017 01.
Article in English | MEDLINE | ID: mdl-26748726

ABSTRACT

Human Papilloma Viruses (HPVs) are the causative agents of cervical cancer although other types of cancers are associated with HPV infection. Type I Interferons can interfere with HPV E6- and/or E7-dependent transformation and can affect microRNA (miRNA) expression. Cancer cells show a specific pattern of miRNA expression and HPVs are able to modulate miRNAs expressed in infected cells. Keratinocytes transduced with E6 and E7 from mucosal HPV-16 or cutaneous HPV-38 (K16 and K38) were studied to analyze the involvement of HPV oncoproteins in the anti-proliferative activity of IFN-ß. In view of our previous data showing senescence induction by the cytokine in K38 cells, we observe that IFN-ß treatment leads to p53-indipendent apoptosis in K16 cells whereas induces senescence in K16 cells if E6 is silenced and p53 expression is restored. The levels of selected miRNAs, deregulated in K16 and K38 cells, can be modulated by IFN-ß when E6 and E7 proteins of HPV-16, but not HPV-38, are expressed.


Subject(s)
Apoptosis/drug effects , Human papillomavirus 16/metabolism , Interferon-beta/pharmacology , Keratinocytes/metabolism , MicroRNAs/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Apoptosis/genetics , Cell Line, Transformed , Human papillomavirus 16/genetics , Humans , Keratinocytes/pathology , Keratinocytes/virology , MicroRNAs/genetics , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins/genetics , Repressor Proteins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
J Cancer Res Clin Oncol ; 142(8): 1751-63, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27300513

ABSTRACT

PURPOSE: Human papillomaviruses (HPVs) are the causative agents of cervical cancer and are also associated with other types of cancers. HPVs can modulate microRNAs (miRNAs) expressed by infected cells. The production of extracellular vesicles is deregulated in cancer, and their cargo delivered to the microenvironment can promote tumorigenesis. The involvement of HPV oncoproteins on miRNA expression in cells and exosomes was analyzed in keratinocytes transduced with E6 and E7 from mucosal HPV-16 or cutaneous HPV-38 (K16 and K38). METHODS: MiRNAs were investigated through the TaqMan Array Human MicroRNA Cards, followed by real-time RT-PCR assay for specific miRNAs. Selected miRNA targets were analyzed by Western blot and correlated to the HPV oncoproteins by specifically silencing E6 and E7 expression. Exosomes, isolated from K16 and K38 supernatants by differential centrifugations, were quantified through the vesicle-associated acetylcholinesterase activity. RESULTS: MiRNAs deregulated in K16 and K38 cells were identified. HPV-16 and/or HPV-38 E6 and E7 single proteins can modify the expression of selected miRNAs involved in the tumorigenesis, in particular miR-18a, -19a, -34a and -590-5p. The analysis of the content of exosomes isolated from HPV-positive cells revealed the presence of E6 and E7 mRNAs and few miRNAs. MiR-222, a key miRNA deregulated in many cancers, was identified in exosomes from K16 cells. CONCLUSIONS: HPV E6 and/or E7 oncoprotein expression can induce the deregulation of some miRNAs. Through the production and function of exosomes, HPV oncogenes as well as HPV-deregulated miRNAs can potentiate the virus oncogenic effects in the tumor cell microenvironment.


Subject(s)
Carcinogenesis , MicroRNAs/genetics , Neoplasms/genetics , Oncogene Proteins, Viral/metabolism , Humans , Neoplasms/virology
8.
Cytokine Growth Factor Rev ; 30: 103-11, 2016 08.
Article in English | MEDLINE | ID: mdl-27021827

ABSTRACT

More than 15% of the global cancer burden is attributable to infectious agents. Pathogens that cause persistent infections are strongly associated with cancer, inflammation being a major component of the chronic infections as revealed by basic, clinical and epidemiological studies. Persistent infection and viral oncoproteins induce specific cellular pathways modifications that promote tumorigenesis. Deregulated and continuous immune response leads to severe tissue and systemic damage, impaired tumor surveillance and consequent carcinogenesis promotion by selecting for metastatic and therapeutically resistant tumor phenotypes. In this review, the role of inflammatory microenvironment in the HPV-induced carcinogenesis is addressed, with a specific focus on the involvement of the immune molecules and microRNAs as well as their delivery through the microvesicle cargo.


Subject(s)
Carcinogenesis/immunology , Inflammation/complications , Papillomaviridae , Papillomavirus Infections/complications , Tumor Microenvironment/immunology , Animals , Dendritic Cells/immunology , Humans , MicroRNAs/immunology
9.
PLoS One ; 10(6): e0130189, 2015.
Article in English | MEDLINE | ID: mdl-26066624

ABSTRACT

BACKGROUND: The potential role of the human immunodeficiency virus-1 (HIV-1) accessory protein Nef in the pathogenesis of neuroAIDS is still poorly understood. Nef is a molecular adapter that influences several cellular signal transduction events and membrane trafficking. In human macrophages, Nef expression induces the production of extracellular factors (e.g. pro-inflammatory chemokines and cytokines) and the recruitment of T cells, thus favoring their infection and its own transfer to uninfected cells via exosomes, cellular protrusions or cell-to-cell contacts. Murine cells are normally not permissive for HIV-1 but, in transgenic mice, Nef is a major disease determinant. Both in human and murine macrophages, myristoylated Nef (myr+Nef) treatment has been shown to activate NF-κB, MAP kinases and interferon responsive factor 3 (IRF-3), thereby inducing tyrosine phosphorylation of signal transducers and activator of transcription (STAT)-1, STAT-2 and STAT-3 through the production of proinflammatory factors. METHODOLOGY/PRINCIPAL FINDINGS: We report that treatment of BV-2 murine microglial cells with myr+Nef leads to STAT-1, -2 and -3 tyrosine phosphorylation and upregulates the expression of inducible nitric oxide synthase (iNOS) with production of nitric oxide. We provide evidence that extracellular Nef regulates iNOS expression through NF-κB activation and, at least in part, interferon-ß (IFNß) release that acts in concert with Nef. All of these effects require both myristoylation and a highly conserved acidic cluster in the viral protein. Finally, we report that Nef induces the release of neurotoxic factors in the supernatants of microglial cells. CONCLUSIONS: These results suggest a potential role of extracellular Nef in promoting neuronal injury in the murine model. They also indicate a possible interplay between Nef and host factors in the pathogenesis of neuroAIDS through the production of reactive nitrogen species in microglial cells.


Subject(s)
Macrophages/pathology , Microglia/pathology , Myristic Acid/metabolism , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Humans , Immunoenzyme Techniques , Interferon-gamma/genetics , Interferon-gamma/metabolism , Macrophages/metabolism , Mice , Microglia/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/genetics , Phosphorylation , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , nef Gene Products, Human Immunodeficiency Virus/genetics
10.
Cytokine Growth Factor Rev ; 26(2): 183-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25466647

ABSTRACT

Numerous microRNAs (miRNAs), small non-coding RNAs encoded in the human genome, have been shown to be involved in cancer pathogenesis and progression. There is evidence that some of these miRNAs possess proapoptotic or proliferation promoting roles in the cell by negatively regulating target mRNAs. Oncogenic viruses are able to produce persistent infection, favoring tumor development by deregulating cell proliferation and inhibiting apoptosis. It has been recently suggested that cellular miRNAs may participate in host-virus interactions, influencing viral replication. Many mammalian viruses counteract this cellular antiviral defense by using viral proteins but also by encoding viral miRNAs involved in virus-induced tumorigenesis. Interferons (IFNs) modulate a number of non-coding RNA genes, especially miRNAs, that may be used by mammalian organisms as a mechanism of IFN system to combat viral infection and related diseases. In particular, IFNs might induce specific cellular miRNAs that target viral transcripts thereby using this strategy as part of their effectiveness against invading viruses. Therefore IFNs, interferon stimulated genes and miRNAs could act synergistically as innate response to virus infection to induce a potent non-permissive cellular environment for virus replication and virus-induced cancer. The relevance of this reviewed research topic is clearly related to the observation that although virus infections are responsible of specific tumors, other unidentified genetic alterations are likely involved in the induction of malignant transformation. The identification of such genetic alterations, i.e. miRNA expression in transformed cells, would be of considerable importance for the analysis of the pathogenesis and for the treatment of cancer induced by specific viruses as well as for the advancement of the current knowledge on the molecular mechanisms underlying virus-host interaction. In this respect, we will review also the important, still little explored, roles of miRNAs acting both as IFN-stimulated anti-viral molecules and as critical regulators of IFNs and IFN-stimulated genes.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/immunology , Interferons/immunology , MicroRNAs/metabolism , Neoplasms/virology , Oncogenic Viruses , Animals , Antiviral Agents/immunology , Antiviral Agents/therapeutic use , Cell Transformation, Viral , Humans , Interferons/genetics , Interferons/metabolism , Interferons/therapeutic use , MicroRNAs/genetics , Neoplasms/therapy , Oncogenic Viruses/genetics , Oncogenic Viruses/immunology , Oncogenic Viruses/pathogenicity , Receptors, Interferon/metabolism , Virus Replication
11.
Curr Med Chem ; 22(8): 958-74, 2015.
Article in English | MEDLINE | ID: mdl-25515511

ABSTRACT

Tumor microenvironment can differ considerably in various types of tumors in terms of cellular and cytokine networks and molecular drivers. The well known link between inflammation and cancer has recently found a number of genetic and molecular confirmations. In this respect, numerous reports have revealed that infection and chronic inflammation can contribute to cancer development, progression and control. Adhesion molecules, chemokines and proinflammatory cytokines, that enroll leukocytes, are persistently present in cancer microenvironment, thus increasing the risk for developing tumors. In this respect, cancer-derived microvescicles, in particular exosomes, exert an important role in the recruitment and reprogramming of components of tumor microenvironment. The relationship between cancer and virus infection has generated, in recent years, a great interest for studies aiming to better understand the role of the immune system in the control of these infections and of the immune cofactors in the promotion of the virus-induced neoplastic transformation. This suggests that virus-induced immune alterations may play a role to create an immunotolerogenic microenvironment during the carcinogenesis process.


Subject(s)
Neoplasms/etiology , Tumor Microenvironment , Tumor Virus Infections/complications , Animals , Humans , Neoplasms/immunology , Oncogenic Viruses
12.
PLoS One ; 7(5): e36909, 2012.
Article in English | MEDLINE | ID: mdl-22615843

ABSTRACT

Interferon (IFN)-ß inhibits cell proliferation and affects cell cycle in keratinocytes transformed by both mucosal high risk Human Papilloma Virus (HPV) and cutaneous HPV E6 and E7 proteins. In particular, upon longer IFN-ß treatments, cutaneous HPV38 expressing cells undergo senescence. IFN-ß appears to induce senescence by upregulating the expression of the tumor suppressor PML, a well known IFN-induced gene. Indeed, experiments in gene silencing via specific siRNAs have shown that PML is essential in the execution of the senescence programme and that both p53 and p21 pathways are involved. IFN-ß treatment leads to a modulation of p53 phosphorylation and acetylation status and a reduction in the expression of the p53 dominant negative ΔNp73. These effects allow the recovery of p53 transactivating activity of target genes involved in the control of cell proliferation. Taken together, these studies suggest that signaling through the IFN pathway might play an important role in cellular senescence. This additional understanding of IFN antitumor action and mechanisms influencing tumor responsiveness or resistance appears useful in aiding further promising development of biomolecular strategies in the IFN therapy of cancer.


Subject(s)
Cell Transformation, Viral , Interferon-beta/metabolism , Keratinocytes/metabolism , Papillomaviridae/physiology , Trans-Activators/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Acetylation , Cell Cycle/genetics , Cell Growth Processes/genetics , Cells, Cultured , Cellular Senescence/genetics , Gene Silencing , Humans , Interferon-beta/genetics , Keratinocytes/virology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Phosphorylation , Promyelocytic Leukemia Protein , Protein Processing, Post-Translational , Trans-Activators/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Up-Regulation
13.
PLoS One ; 6(8): e22982, 2011.
Article in English | MEDLINE | ID: mdl-21886773

ABSTRACT

BACKGROUND: HIV-1 Nef is a virulence factor that plays multiple roles during HIV replication. Recently, it has been described that Nef intersects the CD40 signalling in macrophages, leading to modification in the pattern of secreted factors that appear able to recruit, activate and render T lymphocytes susceptible to HIV infection. The engagement of CD40 by CD40L induces the activation of different signalling cascades that require the recruitment of specific tumor necrosis factor receptor-associated factors (i.e. TRAFs). We hypothesized that TRAFs might be involved in the rapid activation of NF-κB, MAPKs and IRF-3 that were previously described in Nef-treated macrophages to induce the synthesis and secretion of proinflammatory cytokines, chemokines and IFNß to activate STAT1, -2 and -3. METHODOLOGY/PRINCIPAL FINDINGS: Searching for possible TRAF binding sites on Nef, we found a TRAF2 consensus binding site in the AQEEEE sequence encompassing the conserved four-glutamate acidic cluster. Here we show that all the signalling effects we observed in Nef treated macrophages depend on the integrity of the acidic cluster. In addition, Nef was able to interact in vitro with TRAF2, but not TRAF6, and this interaction involved the acidic cluster. Finally silencing experiments in THP-1 monocytic cells indicate that both TRAF2 and, surprisingly, TRAF6 are required for the Nef-induced tyrosine phosphorylation of STAT1 and STAT2. CONCLUSIONS: Results reported here revealed TRAF2 as a new possible cellular interactor of Nef and highlighted that in monocytes/macrophages this viral protein is able to manipulate both the TRAF/NF-κB and TRAF/IRF-3 signalling axes, thereby inducing the synthesis of proinflammatory cytokines and chemokines as well as IFNß.


Subject(s)
HIV-1/metabolism , Inflammation/pathology , Macrophages/pathology , TNF Receptor-Associated Factor 2/metabolism , nef Gene Products, Human Immunodeficiency Virus/chemistry , nef Gene Products, Human Immunodeficiency Virus/metabolism , Amino Acid Sequence , Binding Sites , Chemokines/biosynthesis , Consensus Sequence/genetics , Gene Expression Regulation , Humans , Inflammation/metabolism , Inflammation Mediators/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta/biosynthesis , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/metabolism , Molecular Sequence Data , Mutation/genetics , Myristic Acid/metabolism , NF-kappa B/metabolism , Phosphotyrosine/metabolism , Protein Binding , Protein Structure, Tertiary , STAT Transcription Factors/metabolism , Structure-Activity Relationship , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
14.
Curr Drug Discov Technol ; 4(1): 12-23, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17630924

ABSTRACT

Human immunodeficiency virus (HIV)-1 Nef is a regulatory protein critically involved in AIDS pathogenesis. We previously demonstrated that extracellular Nef is efficiently internalized by human primary monocyte-derived macrophages (MDMs), thereby activating a number of transcription factors including STATs, MAPKs, IRF-3, and NF-kappaB. Such an activation state leads to the release of inflammatory factors whose paracrine effects deserve deep consideration. Here, we demonstrate that quiescent CD4 lymphocytes undergo cell activation when cultivated in supernatants from autologous MDMs treated with extracellular wt Nef but not with its counterpart mutated in the (72)PxxP(75) polyproline domain. Of a pathogenetic relevance, this effect coupled with the sensitization of quiescent CD4 lymphocytes to HIV-1 infection. By microarray assay, we found that the CCL24/eotaxin-2 gene was up-regulated in MDMs treated with wt Nef but not with the (72)AxxA(75) mutant. In addition, the higher transcription activity correlated with a significant increase of the CCL24/Eotaxin-2 release. Finally, we observed that anti-CCL24/eotaxin-2 antibodies efficiently neutralized the stimulatory effect on CD4 lymphocytes of supernatants from MDMs treated with extracellular Nef. Overall, these data support the idea that CCL24/eotaxin-2 is part of the mechanism of CD4 lymphocyte activation paracrinally induced by Nef.


Subject(s)
Chemokines, CC/physiology , Gene Products, nef/physiology , HIV-1/pathogenicity , Adult , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Chemokine CCL24 , Chemokines, CC/biosynthesis , Gene Products, nef/genetics , Gene Products, nef/metabolism , HIV-1/physiology , Humans , Leukocytes, Mononuclear , Lymphocyte Activation , Macrophages/metabolism , Macrophages/virology , Male , Mutation , Oligonucleotide Array Sequence Analysis , Paracrine Communication , Peptides/genetics , Transcription, Genetic , Virus Replication , nef Gene Products, Human Immunodeficiency Virus
15.
J Virol ; 81(6): 2777-91, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17182689

ABSTRACT

The viral protein Nef is a virulence factor that plays multiple roles during the early and late phases of human immunodeficiency virus (HIV) replication. Nef regulates the cell surface expression of critical proteins (including down-regulation of CD4 and major histocompatibility complex class I), T-cell receptor signaling, and apoptosis, inducing proapoptotic effects in uninfected bystander cells and antiapoptotic effects in infected cells. It has been proposed that Nef intersects the CD40 ligand signaling pathway in macrophages, leading to modification in the pattern of secreted factors that appear able to recruit and activate T lymphocytes, rendering them susceptible to HIV infection. There is also increasing evidence that in vitro cell treatment with Nef induces signaling effects. Exogenous Nef treatment is able to induce apoptosis in uninfected T cells, maturation in dendritic cells, and suppression of CD40-dependent immunoglobulin class switching in B cells. Previously, we reported that Nef treatment of primary human monocyte-derived macrophages (MDMs) induces a cycloheximide-independent activation of NF-kappaB and the synthesis and secretion of a set of chemokines/cytokines that activate STAT1 and STAT3. Here, we show that Nef treatment is capable of hijacking cellular signaling pathways, inducing a very rapid regulatory response in MDMs that is characterized by the rapid and transient phosphorylation of the alpha and beta subunits of the IkappaB kinase complex and of JNK, ERK1/2, and p38 mitogen-activated protein kinase family members. In addition, we have observed the activation of interferon regulatory factor 3, leading to the synthesis of beta interferon mRNA and protein, which in turn induces STAT2 phosphorylation. All of these effects require Nef myristoylation.


Subject(s)
Gene Products, nef/pharmacology , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mitogen-Activated Protein Kinases/metabolism , Monocytes/drug effects , Enzyme Activation/drug effects , Gene Products, nef/genetics , Gene Products, nef/physiology , HIV-1/chemistry , HIV-1/genetics , Humans , In Vitro Techniques , Models, Biological , Monocytes/metabolism , Monocytes/virology , Myristic Acid/metabolism , Recombinant Fusion Proteins/pharmacology , nef Gene Products, Human Immunodeficiency Virus
16.
Oncogene ; 24(15): 2536-46, 2005 Apr 07.
Article in English | MEDLINE | ID: mdl-15735750

ABSTRACT

Interferon (IFN)-beta induces S-phase slowing and apoptosis in human papilloma virus (HPV)-positive cervical carcinoma cell line ME-180. Here, we show that apoptosis is a consequence of the S-phase lengthening imposed by IFN-beta, demonstrating the functional correlation between S-phase alteration and apoptosis induction. In ME-180 cells, where p53 function is inhibited by HPV E6 oncoprotein, IFN-beta effects on cell cycle and apoptosis occur independently of p53. The apoptosis due to IFN-beta is mediated by the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in a manner dependent on the S-phase deregulation. IFN-beta appears to increase TRAIL expression both directly at the mRNA level and indirectly by augmenting surface protein levels as a consequence of the induced S-phase cell accumulation. Moreover, the alteration of the S-phase due to IFN-beta promotes TRAIL-dependent apoptosis by potentiating cell sensitivity to TRAIL, possibly through induction of a proapoptotic NF-kappaB activity and TRAIL-R2 receptor expression. Interestingly, IFN-beta-induced TRAIL-dependent apoptotic events strongly differ in the requirement of caspase activity. These results show that IFN-beta may induce an apoptotic response by deregulating cell cycle. Understanding the linkage between these mechanisms appears to be of primary importance in the search for new IFN-based therapeutic strategies to circumvent cancer disease or improve clinical outcome.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma/pathology , Interferon-beta/pharmacology , Membrane Glycoproteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Uterine Cervical Neoplasms/pathology , Apoptosis/physiology , Apoptosis Regulatory Proteins , Carcinoma/virology , Caspases/pharmacology , Female , Gene Expression Profiling , Genes, p53 , Humans , Oligonucleotide Array Sequence Analysis , Papillomaviridae/pathogenicity , RNA, Messenger/analysis , S Phase , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Uterine Cervical Neoplasms/virology
17.
J Leukoc Biol ; 74(5): 821-32, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12960275

ABSTRACT

Increasing evidence indicates that the expression of the human immunodeficiency virus-1 (HIV-1) Nef protein significantly influences the activation state of the host cell. Here we report that Nef specifically activates STAT3 in primary human monocyte-derived macrophages (MDM). This was demonstrated by both single-cycle infection experiments driven by Vesicular Stomatitis virus glycoprotein (VSV-G) pseudotyped HIV-1 and treatment with exogenous recombinant Nef. The analysis of the effects of Nef mutants revealed that domains of the C-terminal flexible loop interacting with the cell endocytotic machinery are involved in the STAT3 activation. In particular, our data suggest that the Nef-dependent STAT3 activation relies on the targeting of Nef to the late endosome/lysosome compartment. In addition, we found that Nef activates STAT3 through a mechanism mediated by the release of soluble factor(s), including MIP-1alpha, that requires de novo protein synthesis but appears independent from the activation of src tyrosine kinases. The results presented here support the idea that the first intervention of Nef in the intracellular signaling of monocyte-macrophages could generate, by means of the release of soluble factor(s), a secondary wave of activation that could be of a potential pathogenetic significance.


Subject(s)
DNA-Binding Proteins/metabolism , Endocytosis/physiology , Gene Products, nef/pharmacology , HIV-1/physiology , Macrophage Activation/drug effects , Macrophages/physiology , Monocytes/physiology , Trans-Activators/metabolism , Acute-Phase Proteins/metabolism , Cells, Cultured , Endocytosis/drug effects , Gene Products, nef/chemistry , Gene Products, nef/genetics , HIV-1/genetics , Humans , Macrophage Activation/physiology , Macrophages/drug effects , Models, Biological , Monocytes/drug effects , Phosphorylation , Recombinant Proteins/pharmacology , STAT3 Transcription Factor , nef Gene Products, Human Immunodeficiency Virus
18.
J Immunol ; 170(4): 1716-27, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12574335

ABSTRACT

It has been recently reported that the endogenous expression of HIV-1 Nef in human monocyte/macrophages induces the release of chemokines and other as yet unidentified soluble factors leading to multiple effects of pathogenic significance, such as the recruitment and activation of quiescent lymphocytes. However, the description of underlying molecular mechanisms remained elusive. We recently demonstrated that human monocyte-derived macrophages (MDM) efficiently internalize soluble rNef, thereby inducing effects largely resembling those observed in cells endogenously expressing Nef. By exploiting the rNef/MDM model, we sought to gain more insights on the molecular mechanisms underlying the response of MDM to Nef. Array analysis for the detection of transcripts from a large number of monokines, chemokines, cytokines, and receptors thereof showed that MDM promptly responded to rNef treatment by increasing the transcription of genes for several inflammatory factors. Analysis of supernatants revealed that rNef treatment induced the release of macrophage inflammatory proteins 1alpha and 1beta, IL-1beta, IL-6, and TNF-alpha. Conversely, rNefs mutated in domains critical for the interaction with the endocytotic machinery (i.e., EE155-156QQ, and DD174-175AA) were ineffective. Interestingly, we found that the Nef-dependent release of inflammatory factors correlated with the activation of the NF-kappaB transcription factor, mainly in its p50/p50 homodimeric form, and in a de novo protein synthesis-independent manner. Our data add new hints supporting the idea that the presence of Nef is per se heavily detrimental for monocyte/macrophages and relative cross-talking cell types.


Subject(s)
Endocytosis/immunology , Gene Products, nef/physiology , HIV-1/physiology , Inflammation Mediators/metabolism , Macrophages/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , Signal Transduction/immunology , Adult , Cells, Cultured , Chemokines/metabolism , Cycloheximide/pharmacology , Cytokines/metabolism , Endocytosis/genetics , Gene Products, nef/genetics , Gene Products, nef/metabolism , HIV-1/genetics , Humans , Macrophages/immunology , Macrophages/virology , Male , Monocytes/immunology , Monocytes/virology , Myristic Acid/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/physiology , Protein Biosynthesis , Protein Structure, Tertiary/genetics , Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology , Signal Transduction/genetics , Solubility , Transcription, Genetic/immunology , Up-Regulation/genetics , Up-Regulation/immunology , nef Gene Products, Human Immunodeficiency Virus
19.
J Interferon Cytokine Res ; 22(1): 39-47, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11846974

ABSTRACT

Numerous evidence has demonstrated the involvement in growth control of interferon (IFN) regulatory factor-1 (IRF-1), which shows tumor suppressor activity. IRF-1 is a well-studied member of the IRF transcription factors that reveals functional diversity in the regulation of cellular response by activating expression of a diverse set of target genes, depending on the cell type and on the specific stimuli. IRF-1 gene rearrangements may be a crucial point in the pathogenesis of some cancer types. Furthermore, different aspects of the tumor suppressor function of IRF-1 may be explained, at least in part, by the observations that IRF-1 is a regulator of cell cycle and apoptosis and that its inactivation accelerates cell transformation. Studies on gene knockout mice contributed greatly to the clarification of these multiple IRF-1 functions. We summarize our current knowledge of the antigrowth effect of IRF-1, focusing also on a more general involvement of IRF-1 in mediating negative regulation of cell growth induced by numerous cytokines and other biologic response modifiers.


Subject(s)
DNA-Binding Proteins/physiology , Phosphoproteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , Base Sequence , Cell Cycle , Cell Division , Cell Transformation, Neoplastic , Cytokines/physiology , DNA-Binding Proteins/genetics , Gene Expression Regulation , Growth Inhibitors/genetics , Growth Inhibitors/physiology , Humans , Interferon Regulatory Factor-1 , Mice , Phosphoproteins/genetics , Response Elements , Transcription Factors/genetics , Transcription Factors/physiology , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...