Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Clin Immunol ; 263: 110233, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38697554

RESUMO

Ataxia-telangiectasia (A-T) is a rare disorder caused by genetic defects of A-T mutated (ATM) kinase, a key regulator of stress response, and characterized by neurodegeneration, immunodeficiency, and high incidence of cancer. Here we investigated NK cells in a mouse model of A-T (Atm-/-) showing that they are strongly impaired at killing tumor cells due to a block of early signaling events. On the other hand, in Atm-/- littermates with thymic lymphoma NK cell cytotoxicity is enhanced as compared with ATM-proficient mice, possibly via tumor-produced TNF-α. Results also suggest that expansion of exhausted NKG2D+ NK cells in Atm-/- mice is driven by low-level expression of stress-inducible NKG2D ligands, whereas development of thymoma expressing the high-affinity MULT1 ligand is associated with NKG2D down-regulation on NK cells. These results expand our understanding of immunodeficiency in A-T and encourage exploring NK cell biology in A-T patients in the attempt to identify cancer predictive biomarkers and novel therapeutic targets.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia , Células Matadoras Naturais , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Animais , Células Matadoras Naturais/imunologia , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Camundongos , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/imunologia , Camundongos Knockout , Camundongos Endogâmicos C57BL , Timoma/imunologia , Timoma/genética , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Citotoxicidade Imunológica , Neoplasias do Timo/imunologia , Neoplasias do Timo/genética , Transdução de Sinais , Proteínas de Membrana , Antígenos de Histocompatibilidade Classe I
2.
Clin Immunol ; 230: 108802, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34298181

RESUMO

Ataxia-telangiectasia (A-T) is a multisystem disorder caused by biallelic pathogenic variants in the gene encoding A-T mutated (ATM) kinase, a master regulator of the DNA damage response (DDR) pathway. Most A-T patients show cellular and/or humoral immunodeficiency that has been associated with cancer risk and reduced survival, but NK cells have not been thoroughly studied. Here we investigated NK cells of A-T patients with a special focus on the NKG2D receptor that triggers cytotoxicity upon engagement by its ligands (NKG2DLs) commonly induced via the DDR pathway on infected, transformed, and variously stressed cells. Using flow cytometry, we examined the phenotype and function of NK cells in 6 A-T patients as compared with healthy individuals. NKG2D expression was evaluated also by western blotting and RT-qPCR; plasma soluble NKG2DLs (sMICA, sMICB, sULBP1, ULBP2) were measured by ELISA. Results showed that A-T NK cells were skewed towards the CD56neg anergic phenotype and displayed decreased expression of NKG2D and perforin. NKG2D was reduced at the protein but not at the mRNA level and resulted in impaired NKG2D-mediated cytotoxicity in 4/6 A-T patients. Moreover, in A-T plasma we found 24-fold and 2-fold increase of sMICA and sULBP1, respectively, both inversely correlated with NKG2D expression. Overall, NK cells are disturbed in A-T patients showing reduced NKG2D expression, possibly caused by persistent engagement of its ligands, that may contribute to susceptibility to cancer and infections and represent novel targets for therapeutic interventions.


Assuntos
Ataxia Telangiectasia/imunologia , Células Matadoras Naturais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Adolescente , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/metabolismo , Estudos de Casos e Controles , Linhagem Celular , Criança , Citotoxicidade Imunológica , Regulação para Baixo , Feminino , Citometria de Fluxo , Proteínas Ligadas por GPI/sangue , Antígenos de Histocompatibilidade Classe I/sangue , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/sangue , Peptídeos e Proteínas de Sinalização Intracelular/sangue , Células K562 , Células Matadoras Naturais/metabolismo , Ligantes , Masculino , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Adulto Jovem
3.
RNA Biol ; 18(sup1): 75-87, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34224323

RESUMO

Adenosine deaminases acting on RNA (ADARs) are enzymes that convert adenosines to inosines in double-stranded RNAs (RNA editing A-to-I). ADAR1 and ADAR2 were previously reported as HIV-1 proviral factors. The aim of this study was to investigate the composition of the ADAR2 ribonucleoprotein complex during HIV-1 expression. By using a dual-tag affinity purification procedure in cells expressing HIV-1 followed by mass spectrometry analysis, we identified 10 non-ribosomal ADAR2-interacting factors. A significant fraction of these proteins was previously found associated to the Long INterspersed Element 1 (LINE1 or L1) ribonucleoparticles and to regulate the life cycle of L1 retrotransposons. Considering that we previously demonstrated that ADAR1 is an inhibitor of LINE-1 retrotransposon activity, we investigated whether also ADAR2 played a similar function. To reach this goal, we performed specific cell culture retrotransposition assays in cells overexpressing or ablated for ADAR2. These experiments unveil a novel function of ADAR2 as suppressor of L1 retrotransposition. Furthermore, we showed that ADAR2 binds the basal L1 RNP complex.Overall, these data support the role of ADAR2 as regulator of L1 life cycle.


Assuntos
Adenosina Desaminase/metabolismo , Elementos Nucleotídeos Longos e Dispersos , Edição de RNA , Proteínas de Ligação a RNA/metabolismo , Adenosina Desaminase/genética , Células HEK293 , Células HeLa , Humanos , Proteínas de Ligação a RNA/genética
4.
Int J Mol Sci ; 22(13)2021 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-34206330

RESUMO

The 'shock-and-kill' strategy to purge the latent HIV reservoir relies on latency-reversing agents (LRAs) to reactivate the provirus and subsequent immune-mediated killing of HIV-expressing cells. Yet, clinical trials employing histone deacetylase inhibitors (HDACis; Vorinostat, Romidepsin, Panobinostat) as LRAs failed to reduce the HIV reservoir size, stressing the need for more effective latency reversal strategies, such as 2-LRA combinations, and enhancement of the immune responses. Interestingly, several LRAs are employed to treat cancer because they up-modulate ligands for the NKG2D NK-cell activating receptor on tumor cells. Therefore, using in vitro T cell models of HIV latency and NK cells, we investigated the capacity of HDACis, either alone or combined with a distinct LRA, to potentiate the NKG2D/NKG2D ligands axis. While Bortezomib proteasome inhibitor was toxic for both T and NK cells, the GS-9620 TLR-7 agonist antagonized HIV reactivation and NKG2D ligand expression by HDACis. Conversely, co-administration of the Prostratin PKC agonist attenuated HDACi toxicity and, when combined with Romidepsin, stimulated HIV reactivation and further up-modulated NKG2D ligands on HIV+ T cells and NKG2D on NK cells, ultimately boosting NKG2D-mediated viral suppression by NK cells. These findings disclose limitations of LRA candidates and provide evidence that NK cell suppression of reactivated HIV may be modulated by specific 2-LRA combinations.


Assuntos
Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , Inibidores de Histona Desacetilases/uso terapêutico , Células Matadoras Naturais/imunologia , Linfócitos T/virologia , Latência Viral , Infecções por HIV/imunologia , Infecções por HIV/fisiopatologia , Infecções por HIV/terapia , Humanos , Células Matadoras Naturais/fisiologia
5.
Int J Mol Sci ; 21(22)2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-33203071

RESUMO

Schimke immuno-osseous dysplasia (SIOD) is a rare multisystemic disorder with a variable clinical expressivity caused by biallelic variants in SMARCAL1. A phenotype-genotype correlation has been attempted and variable expressivity of biallelic SMARCAL1 variants may be associated with environmental and genetic disturbances of gene expression. We describe two siblings born from consanguineous parents with a diagnosis of SIOD revealed by whole exome sequencing (WES). Results: A homozygous missense variant in the SMARCAL1 gene (c.1682G>A; p.Arg561His) was identified in both patients. Despite carrying the same variant, the two patients showed substantial renal and immunological phenotypic differences. We describe features not previously associated with SIOD-both patients had congenital anomalies of the kidneys and of the urinary tract and one of them succumbed to a classical type congenital mesoblastic nephroma. We performed an extensive characterization of the immunophenotype showing combined immunodeficiency characterized by a profound lymphopenia, lack of thymic output, defective IL-7Rα expression, and disturbed B plasma cells differentiation and immunoglobulin production in addition to an altered NK-cell phenotype and function. Conclusions: Overall, our results contribute to extending the phenotypic spectrum of features associated with SMARCAL1 mutations and to better characterizing the underlying immunologic disorder with critical implications for therapeutic and management strategies.


Assuntos
Arteriosclerose , DNA Helicases , Rim , Células Matadoras Naturais/imunologia , Mutação de Sentido Incorreto , Nefroma Mesoblástico , Síndrome Nefrótica , Osteocondrodisplasias , Fenótipo , Doenças da Imunodeficiência Primária , Embolia Pulmonar , Sistema Urinário , Substituição de Aminoácidos , Arteriosclerose/diagnóstico por imagem , Arteriosclerose/genética , Arteriosclerose/imunologia , DNA Helicases/genética , DNA Helicases/imunologia , Feminino , Humanos , Subunidade alfa de Receptor de Interleucina-7/genética , Subunidade alfa de Receptor de Interleucina-7/imunologia , Rim/anormalidades , Rim/diagnóstico por imagem , Rim/imunologia , Masculino , Nefroma Mesoblástico/diagnóstico por imagem , Nefroma Mesoblástico/genética , Nefroma Mesoblástico/imunologia , Síndrome Nefrótica/diagnóstico por imagem , Síndrome Nefrótica/genética , Síndrome Nefrótica/imunologia , Osteocondrodisplasias/diagnóstico por imagem , Osteocondrodisplasias/genética , Osteocondrodisplasias/imunologia , Doenças da Imunodeficiência Primária/diagnóstico por imagem , Doenças da Imunodeficiência Primária/genética , Doenças da Imunodeficiência Primária/imunologia , Embolia Pulmonar/diagnóstico por imagem , Embolia Pulmonar/genética , Embolia Pulmonar/imunologia , Sistema Urinário/anormalidades , Sistema Urinário/diagnóstico por imagem , Sistema Urinário/imunologia , Sequenciamento Completo do Genoma
6.
Nucleic Acids Res ; 45(1): 155-168, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-27658966

RESUMO

Adenosine deaminases acting on RNA (ADARs) are involved in RNA editing that converts adenosines to inosines in double-stranded RNAs. ADAR1 was demonstrated to be functional on different viruses exerting either antiviral or proviral effects. Concerning HIV-1, several studies showed that ADAR1 favors viral replication. The aim of this study was to investigate the composition of the ADAR1 ribonucleoprotein complex during HIV-1 expression. By using a dual-tag affinity purification procedure in cells expressing HIV-1 followed by mass spectrometry analysis, we identified 14 non-ribosomal ADAR1-interacting proteins, most of which are novel. A significant fraction of these proteins were previously demonstrated to be associated to the Long INterspersed Element 1 (LINE1 or L1) ribonucleoparticles and to regulate the life cycle of L1 retrotransposons that continuously re-enter host-genome.Hence, we investigated the function of ADAR1 in the regulation of L1 activity.By using different cell-culture based retrotransposition assays in HeLa cells, we demonstrated a novel function of ADAR1 as suppressor of L1 retrotransposition. Apparently, this inhibitory mechanism does not occur through ADAR1 editing activity. Furthermore, we showed that ADAR1 binds the basal L1 RNP complex. Overall, these data support the role of ADAR1 as regulator of L1 life cycle.


Assuntos
Adenosina Desaminase/genética , HIV-1/genética , Elementos Nucleotídeos Longos e Dispersos , Proteínas de Ligação a RNA/genética , Retroelementos , Ribonucleoproteínas/genética , Adenosina Desaminase/metabolismo , Bioensaio , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Células HEK293 , HIV-1/metabolismo , Células HeLa , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Anotação de Sequência Molecular , Ligação Proteica , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Transdução de Sinais
7.
Int J Mol Sci ; 20(18)2019 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-31514330

RESUMO

Viral persistency in latently infected CD4+ T cells despite antiretroviral therapy (ART) represents a major drawback in the fight against HIV-1. Efforts to purge latent HIV-1 have been attempted using latency reversing agents (LRAs) that activate expression of the quiescent virus. However, initial trials have shown that immune responses of ART-treated patients are ineffective at clearing LRA-reactivated HIV-1 reservoirs, suggesting that an adjuvant immunotherapy is needed. Here we overview multiple lines of evidence indicating that natural killer (NK) cells have the potential to induce anti-HIV-1 responses relevant for virus eradication. In particular, we focus on the role of the NKG2D activating receptor that crucially enables NK cell-mediated killing of HIV-1-infected cells. We describe recent data indicating that LRAs can synergize with HIV-1 at upregulating ligands for NKG2D (NKG2DLs), hence sensitizing T cells that exit from viral latency for recognition and lysis by NK cells; in addition, we report in vivo and ex vivo data showing the potential benefits and drawbacks that LRAs may have on NKG2D expression and, more in general, on the cytotoxicity of NK cells. Finally, we discuss how the NKG2D/NKG2DLs axis can be exploited for the development of effective HIV-1 eradication strategies combining LRA-induced virus reactivation with recently optimized NK cell-based immunotherapies.


Assuntos
Reservatórios de Doenças/virologia , Infecções por HIV/imunologia , HIV-1/imunologia , Células Matadoras Naturais/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Ensaios Clínicos como Assunto , Humanos
8.
Int J Mol Sci ; 20(15)2019 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-31366013

RESUMO

Natural Killer (NK) cells play a critical role in host defense against viral infections. The mechanisms of recognition and killing of virus-infected cells mediated by NK cells are still only partially defined. Several viruses induce, on the surface of target cells, the expression of molecules that are specifically recognized by NK cell-activating receptors. The main NK cell-activating receptors involved in the recognition and killing of virus-infected cells are NKG2D and DNAM-1. In particular, ligands for DNAM-1 are nectin/nectin-like molecules involved also in mechanisms allowing viral infection. Viruses adopt several immune evasion strategies, including those affecting NK cell-mediated immune surveillance, causing persistent viral infection and the development of virus-associated diseases. The virus's immune evasion efficacy depends on molecules differently expressed during the various phases of infection. In this review, we overview the molecular strategies adopted by viruses, specifically cytomegalovirus (CMV), human immunodeficiency virus (HIV-1), herpes virus (HSV), Epstein-Barr virus (EBV) and hepatitis C virus (HCV), aiming to evade NK cell-mediated surveillance, with a special focus on the modulation of DNAM-1 activating receptor and its ligands in various phases of the viral life cycle. The increasing understanding of mechanisms involved in the modulation of activating ligands, together with those mediating the viral immune evasion strategies, would provide critical tools leading to design novel NK cell-based immunotherapies aiming at viral infection control, thus improving cure strategies of virus-associated diseases.


Assuntos
Antígenos de Diferenciação de Linfócitos T/metabolismo , Vigilância Imunológica , Células Matadoras Naturais/imunologia , Viroses/imunologia , Animais , Humanos , Células Matadoras Naturais/virologia , Nectinas/metabolismo , Viroses/virologia , Replicação Viral
9.
J Virol ; 89(10): 5687-700, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25822027

RESUMO

UNLABELLED: Leukocyte recirculation between blood and lymphoid tissues is required for the generation and maintenance of immune responses against pathogens and is crucially controlled by the L-selectin (CD62L) leukocyte homing receptor. CD62L has adhesion and signaling functions and initiates the capture and rolling on the vascular endothelium of cells entering peripheral lymph nodes. This study reveals that CD62L is strongly downregulated on primary CD4(+) T lymphocytes upon infection with human immunodeficiency virus type 1 (HIV-1). Reduced cell surface CD62L expression was attributable to the Nef and Vpu viral proteins and not due to increased shedding via matrix metalloproteases. Both Nef and Vpu associated with and sequestered CD62L in perinuclear compartments, thereby impeding CD62L transport to the plasma membrane. In addition, Nef decreased total CD62L protein levels. Importantly, infection with wild-type, but not Nef- and Vpu-deficient, HIV-1 inhibited the capacity of primary CD4(+) T lymphocytes to adhere to immobilized fibronectin in response to CD62L ligation. Moreover, HIV-1 infection impaired the signaling pathways and costimulatory signals triggered in primary CD4(+) T cells by CD62L ligation. We propose that HIV-1 dysregulates CD62L expression to interfere with the trafficking and activation of infected T cells. Altogether, this novel HIV-1 function could contribute to virus dissemination and evasion of host immune responses. IMPORTANCE: L-selectin (CD62L) is an adhesion molecule that mediates the first steps of leukocyte homing to peripheral lymph nodes, thus crucially controlling the initiation and maintenance of immune responses to pathogens. Here, we report that CD62L is downmodulated on the surfaces of HIV-1-infected T cells through the activities of two viral proteins, Nef and Vpu, that prevent newly synthesized CD62L molecules from reaching the plasma membrane. We provide evidence that CD62L downregulation on HIV-1-infected primary T cells results in impaired adhesion and signaling functions upon CD62L triggering. Removal of cell surface CD62L may predictably keep HIV-1-infected cells away from lymph nodes, the privileged sites of both viral replication and immune response activation, with important consequences, such as systemic viral spread and evasion of host immune surveillance. Altogether, we propose that Nef- and Vpu-mediated subversion of CD62L function could represent a novel determinant of HIV-1 pathogenesis.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/imunologia , HIV-1/patogenicidade , Proteínas do Vírus da Imunodeficiência Humana/imunologia , Selectina L/metabolismo , Proteínas Virais Reguladoras e Acessórias/imunologia , Produtos do Gene nef do Vírus da Imunodeficiência Humana/imunologia , Adesão Celular/imunologia , Células Cultivadas , Regulação para Baixo , HIV-1/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Células Jurkat , Ativação Linfocitária , Transporte Proteico , Transdução de Sinais/imunologia
10.
FASEB J ; 27(6): 2440-50, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23395909

RESUMO

In humans, the interaction of the natural killer group 2 member D (NKG2D)-activating receptor on natural killer (NK) and CD8(+) T cells with its major histocompatibility complex class I-related chain (MIC) and UL16 binding protein (ULBP) ligands (NKG2DLs) promotes recognition and elimination of stressed cells, such as tumor or infected cells. Here, we investigated the capacity of HIV-1 to modulate NKG2DL expression and escape NGK2D-mediated immunosurveillance. In CD4(+) T lymphocytes, both cell surface expression and release of MICA, MICB, and ULBP2 were up-regulated >2-fold by HIV-1 infection. In HIV-infected CD4(+) T lymphocytes or Jurkat T-cell lines, increased shedding of soluble NKG2DLs (sNKG2DLs) was impaired by a matrix metalloproteinase inhibitor (MMPI). Moreover, naive HIV(+) patients displayed increased plasma sMICA and sULBP2 levels and reduced NKG2D expression on NK and CD8(+) T cells compared to patients receiving highly active antiretroviral therapy (HAART) or healthy donors. In individual patients, HAART uptake resulted in the drop of sNKG2DL and recovery of NKG2D expression. Finally, sNKG2DLs in patients' plasma down-regulated NKG2D on NK and CD8(+) T cells and impaired NKG2D-mediated cytotoxicity of NK cells. Thus, NKG2D detuning by sNKG2DLs may promote HIV-1 immune evasion and compromise host resistance to opportunistic infections, but HAART and MMPI have the potential to avoid such immune dysfunction.


Assuntos
Citotoxicidade Imunológica , Infecções por HIV/imunologia , HIV-1 , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/antagonistas & inibidores , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Adolescente , Adulto , Terapia Antirretroviral de Alta Atividade , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Estudos de Casos e Controles , Proteínas Ligadas por GPI/metabolismo , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células Jurkat , Células K562 , Células Matadoras Naturais/metabolismo , Ligantes , Metaloproteinase 1 da Matriz/metabolismo , Subfamília K de Receptores Semelhantes a Lectina de Células NK/sangue , Adulto Jovem
11.
RNA Biol ; 11(4): 334-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24717285

RESUMO

Several cellular microRNAs show substantial changes in expression during HIV-1 infection and their active role in the viral life cycle is progressively emerging. In the present study, we found that HIV-1 infection of Jurkat T cells significantly induces the expression of miR-222. We show that this induction depends on HIV-1 Tat protein, which is able to increase the transcriptional activity of NFkB on miR-222 promoter. Moreover, we demonstrate that miR-222 directly targets CD4, a key receptor for HIV-1, thus reducing its expression. We propose that Tat, by inducing miR-222 expression, complements the CD4 downregulation activity exerted by other viral proteins (i.e., Nef, Vpu, and Env), and we suggest that this represents a novel mechanism through which HIV-1 efficiently represses CD4 expression in infected cells.


Assuntos
Antígenos CD4/genética , Regulação da Expressão Gênica , Infecções por HIV/genética , HIV-1/fisiologia , MicroRNAs/genética , Subpopulações de Linfócitos T/metabolismo , Produtos do Gene tat do Vírus da Imunodeficiência Humana/metabolismo , Antígenos CD4/metabolismo , Linhagem Celular , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Humanos , NF-kappa B/metabolismo , RNA Mensageiro/genética , Subpopulações de Linfócitos T/imunologia
12.
J Gen Virol ; 94(Pt 12): 2664-2669, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24045107

RESUMO

Viral infection may induce the cell-surface expression of PVR (CD155) that, upon recognition by its cognate activating DNAM-1 receptor present on cytotoxic lymphocytes, may promote antiviral immune responses. Here we show that expression of the human immunodeficiency virus type 1 (HIV-1) Vpr protein in Jurkat T cells increases cell-surface and total PVR levels. Analysis of mutated Vpr variants indicated that Vpr uses the same protein surfaces, and hence probably the same mechanisms, to upregulate PVR and arrest the cell cycle in the G2 phase. Moreover, we found that PVR upregulation by Vpr relied on the ability of the protein to activate the ATR kinase that triggers the DNA damage response pathway and G2 arrest. Finally, we showed that Vpr contributes to PVR up-modulation in HIV-infected CD4(+) T lymphocytes and inhibits the PVR downregulating activity of the viral Nef protein.


Assuntos
Dano ao DNA/efeitos dos fármacos , Receptores Virais/metabolismo , Regulação para Cima , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/farmacologia , Linfócitos T CD4-Positivos/virologia , Fase G2/efeitos dos fármacos , Infecções por HIV/virologia , HIV-1 , Humanos , Células Jurkat , Receptores Virais/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/genética , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/farmacologia
13.
J Virol ; 86(8): 4496-504, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22301152

RESUMO

The human immunodeficiency virus type 1 (HIV-1) evades the immune responses of natural killer (NK) cells through mechanisms that have been partially deciphered. Here we show that in HIV-1-infected T lymphocytes, the early viral Nef protein downmodulates PVR (CD155, Necl-5), a ligand for the activating receptor DNAM-1 (CD226) expressed by all NK cells, CD8(+) T cells, and other cell types. This novel Nef activity is conserved by Nef proteins of laboratory HIV-1 strains (NL4-3, SF2) and of a patient-derived virus, but it is not maintained by HIV-2. Nef uses the same motifs to downregulate PVR and HLA-I molecules, likely by the same mechanisms. Indeed, as previously demonstrated for HLA-I, Nef reduces the total amounts of cell-associated PVR. Optimal downregulation of cell surface PVR by Nef also requires the presence of the late viral factor Vpu. In line with PVR reduction, the NK cell-mediated lysis of T cells infected by a wild-type but not Nef-deficient virus is virtually abrogated upon blocking of both DNAM-1 and another activating receptor, NKG2D, previously shown to mediate killing of HIV-infected cells. Together, these data demonstrate that the PVR downmodulation by Nef and Vpu is a strategy evolved by HIV-1 to prevent NK cell-mediated lysis of infected cells. The PVR downregulation reported here has the potential to affect the immune responses of other DNAM-1-positive cells besides NK cells and to alter multiple PVR-mediated cellular processes, such as adhesion and migration, and may thus greatly influence HIV-1 pathogenesis.


Assuntos
Regulação para Baixo , HIV-1/imunologia , HIV-1/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Células Matadoras Naturais/imunologia , Receptores Virais/metabolismo , Proteínas Virais Reguladoras e Acessórias/metabolismo , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular , Citotoxicidade Imunológica , Expressão Gênica , HIV-1/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Proteínas do Vírus da Imunodeficiência Humana/genética , Humanos , Células Matadoras Naturais/metabolismo , Ligação Proteica , Receptores Virais/genética , Proteínas Virais Reguladoras e Acessórias/genética , Produtos do Gene nef do Vírus da Imunodeficiência Humana/genética
14.
Cancers (Basel) ; 15(6)2023 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-36980798

RESUMO

The Epstein-Barr virus (EBV) is a ubiquitous herpesvirus most often transmitted during infancy and infecting the vast majority of human beings. Usually, EBV infection is nearly asymptomatic and results in life-long persistency of the virus in a latent state under the control of the host immune system. Yet EBV can cause an acute infectious mononucleosis (IM), particularly in adolescents, and is associated with several malignancies and severe diseases that pose a serious threat to individuals with specific inborn error of immunity (IEI). While there is a general consensus on the requirement for functional CD8 T cells to control EBV infection, the role of the natural killer (NK) cells of the innate arm of immunity is more enigmatic. Here we provide an overview of the interaction between EBV and NK cells in the immunocompetent host as well as in the context of primary and secondary immunodeficiencies. Moreover, we report in vitro data on the mechanisms that regulate the capacity of NK cells to recognize and kill EBV-infected cell targets and discuss the potential of recently optimized NK cell-based immunotherapies for the treatment of EBV-associated diseases.

15.
Sci Rep ; 12(1): 18567, 2022 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-36329160

RESUMO

Inhibitors of histone deacetylases (HDACis) are major latency reversing agent (LRA) candidates in 'shock and kill' strategies to eradicate the HIV reservoir in infected patients. The poor achievements of initial HDACi-based trials and subsequent studies have highlighted the need for more efficient approaches such as combinatory and immunostimulating therapies. Here we studied combinations of IL-15 with pan-HDACi (Vorinostat, Romidepsin, Panobinostat) or class I selective-HDACi (Entinostat) with or without a PKC agonist (Prostratin) for their impact on in vitro reactivation and NK cell-mediated suppression of latent HIV. Results showed that pan-HDACis but not Entinostat reduced NK cell viability and function; yet, combined IL-15 reverted the negative effects of pan-HDACis except for Panobinostat. All HDACis were ineffective at reactivating HIV in a CD4+ T cell model of latency, with pan-HDACis suppressing spontaneous and IL-15- or Prostratin-induced HIV release, while IL-15 + Prostratin combination showed maximal activity. Moreover, Panobinostat impaired STAT5 and NF-κB activation by IL-15 and Prostratin, respectively. Finally, by using effectors (NK) and targets (latently infected CD4+ T cells) equally exposed to drug combinations, we found that IL-15-mediated suppression of HIV reactivation by NK cells was inhibited by Panobinostat. Our data raise concerns and encouragements for therapeutic application of IL-15/LRA combinations.


Assuntos
Infecções por HIV , HIV-1 , Humanos , Latência Viral , HIV-1/fisiologia , Panobinostat/farmacologia , Panobinostat/uso terapêutico , Infecções por HIV/tratamento farmacológico , Interleucina-15/farmacologia , Linfócitos T CD4-Positivos , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Células Matadoras Naturais , Ativação Viral
16.
Cancers (Basel) ; 14(9)2022 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-35565433

RESUMO

The most widely accepted hypothesis for the development of glioblastoma suggests that glioblastoma stem-like cells (GSCs) are crucially involved in tumor initiation and recurrence as well as in the occurrence of chemo- and radio-resistance. Mesenchyme homeobox 2 (MEOX2) is a transcription factor overexpressed in glioblastoma, whose expression is negatively correlated with patient survival. Starting from our observation that MEOX2 expression is strongly enhanced in six GSC lines, we performed shRNA-mediated knock-down experiments in two different GSC lines and found that MEOX2 depletion resulted in the inhibition of cell growth and sphere-forming ability and an increase in apoptotic cell death. By a deep transcriptome analysis, we identified a core group of genes modulated in response to MEOX2 knock-down. Among these genes, the repressed ones are largely enriched in genes involved in the hypoxic response and glycolytic pathway, two strictly related pathways that contribute to the resistance of high-grade gliomas to therapies. An in silico study of the regulatory regions of genes differentially expressed by MEOX2 knock-down revealed that they mainly consisted of GC-rich regions enriched for Sp1 and Klf4 binding motifs, two main regulators of metabolism in glioblastoma. Our results show, for the first time, the involvement of MEOX2 in the regulation of genes of GSC metabolism, which is essential for the survival and growth of these cells.

17.
Front Immunol ; 13: 799564, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154113

RESUMO

The study of inborn errors of immunity (IEI) provides unique opportunities to elucidate the microbiome and pathogenic mechanisms related to severe viral infection. Several immunological and genetic anomalies may contribute to the susceptibility to develop Human Papillomavirus (HPV) pathogenesis. They include different acquired immunodeficiencies, EVER1-2 or CIB1 mutations underlying epidermodysplasia verruciformis (EV) syndrome and multiple IEI. Whereas EV syndrome patients are specifically unable to control infections with beta HPV, individuals with IEI show broader infectious and immune phenotypes. The WHIM (warts, hypogammaglobulinemia, infection, and myelokathexis) syndrome caused by gain-of-CXCR4-function mutation manifests by HPV-induced extensive cutaneous warts but also anogenital lesions that eventually progress to dysplasia. Here we report alterations of B and NK cells in a female patient suffering from cutaneous and mucosal HPV-induced lesions due to an as-yet unidentified genetic defect. Despite no detected mutations in CXCR4, B but not NK cells displayed a defective CXCR4-dependent chemotactic response toward CXCL12. In addition, NK cells showed an abnormal distribution with an expanded CD56bright cell subset and defective cytotoxicity of CD56dim cells. Our observations extend the clinical and immunological spectrum of IEI associated with selective susceptibility toward HPV pathogenesis, thus providing new insight on the immune control of HPV infection and potential host susceptibility factors.


Assuntos
Linfócitos B/imunologia , Linfócitos B/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Infecções por Papillomavirus/diagnóstico , Doenças da Imunodeficiência Primária/complicações , Doenças da Imunodeficiência Primária/etiologia , Receptores CXCR4/metabolismo , Biomarcadores , Suscetibilidade a Doenças , Humanos , Infecções por Papillomavirus/etiologia , Doenças da Imunodeficiência Primária/diagnóstico
18.
J Exp Med ; 202(6): 817-28, 2005 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-16157687

RESUMO

The presentation of exogenous protein antigens in a major histocompatibility complex class I-restricted fashion to CD8+ T cells is called cross-presentation. We demonstrate that cross-presentation of soluble viral antigens (derived from hepatitis C virus [HCV], hepatitis B virus [HBV], or human immunodeficiency virus) to specific CD8+ T cell clones is dramatically improved when antigen-presenting dendritic cells (DCs) are pulsed with the antigen in the presence of chloroquine or ammonium chloride, which reduce acidification of the endocytic system. The export of soluble antigen into the cytosol is considerably higher in chloroquine-treated than in untreated DCs, as detected by confocal microscopy of cultured cells and Western blot analysis comparing endocytic and cytosolic fractions. To pursue our findings in an in vivo setting, we boosted groups of HBV vaccine responder individuals with a further dose of hepatitis B envelope protein vaccine with or without a single dose of chloroquine. Although all individuals showed a boost in antibody titers to HBV, six of nine individuals who were administered chloroquine showed a substantial CD8+ T cell response to HBV antigen, whereas zero of eight without chloroquine lacked a CD8 response. Our results suggest that chloroquine treatment improves CD8 immunity during vaccination.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antimaláricos/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Cloroquina/farmacologia , Apresentação Cruzada/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Antígenos/imunologia , Antígenos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Cultivadas , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Hepacivirus/imunologia , Humanos , Microscopia Confocal , Solubilidade , Regulação para Cima/efeitos dos fármacos , Proteínas não Estruturais Virais/imunologia , Proteínas não Estruturais Virais/metabolismo
19.
J Gen Virol ; 92(Pt 1): 141-50, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20881088

RESUMO

Nef is a multifunctional protein of the human immunodeficiency virus type 1 (HIV-1) required for high viral replication and disease progression. Several findings indicate that the capacity of Nef to downregulate surface CD4 is essential for the protein's pathogenic activity, although the mechanisms that link the two functions are yet unclear. It is believed that, by reducing surface CD4 levels, Nef counteracts the receptor's negative effects on virion infectivity and release. Here, we show that, in 293T cells co-expressing CD4 and HIV-1, the capacity of Nef to enhance the virion incorporation of Env products and release of viral particles was mediated by retention-degradation of neo-synthesized CD4 rather than by accelerated receptor endocytosis. Different results were observed in primary CD4(+) T lymphocytes in which Nef-mediated CD4 downregulation occurs primarily by accelerated internalization. In HIV-infected T cell cultures, Nef was crucial for the removal of surface CD4 at the beginning of the infection, while later on maximal CD4 downregulation was achieved in a Nef-independent manner. Moreover, by means of in vivo selected Nef mutants, we observed that CD4 downregulation is not essential for Nef ability to enhance Env incorporation into virions and increase viral infectivity or replication in CD4(+) T lymphocytes. Notably, Nef expression itself was dispensable for efficient release of HIV-1 particles by T cells. In conclusion, we propose that the CD4 downregulation activity of Nef plays a role before the late productive phases of HIV-1 replication.


Assuntos
Antígenos CD4/biossíntese , Linfócitos T CD4-Positivos/virologia , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Replicação Viral , Produtos do Gene nef do Vírus da Imunodeficiência Humana/metabolismo , Células Cultivadas , Regulação para Baixo , Humanos
20.
J Gen Virol ; 92(Pt 5): 1228-1232, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21289159

RESUMO

The adenosine deaminases acting on RNA (ADAR) enzymes catalyse conversion of adenosine to inosine in dsRNA. A positive effect of ADAR1 on human immunodeficiency virus type 1 (HIV-1) replication has recently been reported. Here, we show that another ADAR enzyme, ADAR2, positively affects the replication process of HIV-1. We found that, analogously to ADAR1, ADAR2 enhances the release of progeny virions by an editing-dependent mechanism. However, differently from the ADAR1 enzyme, ADAR2 does not increase the infectious potential of the virus. Importantly, downregulation of ADAR2 in Jurkat cells significantly impairs viral replication. Therefore, ADAR2 shares some but not all proviral functions of ADAR1. These results suggest a novel role of ADAR2 as a viral regulator.


Assuntos
Adenosina Desaminase/metabolismo , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Provírus/patogenicidade , Replicação Viral , Humanos , Células Jurkat , Proteínas de Ligação a RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA