Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 27(6): 109960, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38832015

RESUMEN

Limited information exists regarding the impact of interferons (IFNs) on the information carried by extracellular vesicles (EVs). This study aimed at investigating whether IFN-α2b, IFN-ß, IFN-γ, and IFN-λ1/2 modulate the content of EVs released by primary monocyte-derived macrophages (MDM). Small-EVs (sEVs) were purified by size exclusion chromatography from supernatants of MDM treated with IFNs. To characterize the concentration and dimensions of vesicles, nanoparticle tracking analysis was used. SEVs surface markers were examined by flow cytometry. IFN treatments induced a significant down-regulation of the exosomal markers CD9, CD63, and CD81 on sEVs, and a significant modulation of some adhesion molecules, major histocompatibility complexes and pro-coagulant proteins, suggesting IFNs influence biogenesis and shape the immunological asset of sEVs. SEVs released by IFN-stimulated MDM also impact lymphocyte function, showing significant modulation of lymphocyte activation and IL-17 release. Altogether, our results show that sEVs composition and activity are affected by IFN treatment of MDM.

2.
Int J Mol Sci ; 24(11)2023 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-37298304

RESUMEN

Type I and III Interferons (IFNs) are the first lines of defense in microbial infections. They critically block early animal virus infection, replication, spread, and tropism to promote the adaptive immune response. Type I IFNs induce a systemic response that impacts nearly every cell in the host, while type III IFNs' susceptibility is restricted to anatomic barriers and selected immune cells. Both IFN types are critical cytokines for the antiviral response against epithelium-tropic viruses being effectors of innate immunity and regulators of the development of the adaptive immune response. Indeed, the innate antiviral immune response is essential to limit virus replication at the early stages of infection, thus reducing viral spread and pathogenesis. However, many animal viruses have evolved strategies to evade the antiviral immune response. The Coronaviridae are viruses with the largest genome among the RNA viruses. Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) caused the coronavirus disease 2019 (COVID-19) pandemic. The virus has evolved numerous strategies to contrast the IFN system immunity. We intend to describe the virus-mediated evasion of the IFN responses by going through the main phases: First, the molecular mechanisms involved; second, the role of the genetic background of IFN production during SARS-CoV-2 infection; and third, the potential novel approaches to contrast viral pathogenesis by restoring endogenous type I and III IFNs production and sensitivity at the sites of infection.


Asunto(s)
COVID-19 , Interferón Tipo I , Animales , Interferones/genética , SARS-CoV-2 , Antivirales/farmacología , Antivirales/uso terapéutico , Interferón Tipo I/genética , Citocinas , Inmunidad Innata , Evasión Inmune
3.
Front Endocrinol (Lausanne) ; 13: 961744, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36213288

RESUMEN

Thyroid hormones, T3 (triiodothyronine) and T4 (thyroxine), induce a variety of long-term effects on important physiological functions, ranging from development and growth to metabolism regulation, by interacting with specific nuclear or cytosolic receptors. Extranuclear or nongenomic effects of thyroid hormones are mediated by plasma membrane or cytoplasmic receptors, mainly by αvß3 integrin, and are independent of protein synthesis. A wide variety of nongenomic effects have now been recognized to be elicited through the binding of thyroid hormones to this receptor, which is mainly involved in angiogenesis, as well as in cell cancer proliferation. Several signal transduction pathways are modulated by thyroid hormone binding to αvß3 integrin: protein kinase C, protein kinase A, Src, or mitogen-activated kinases. Thyroid hormone-activated nongenomic effects are also involved in the regulation of Na+-dependent transport systems, such as glucose uptake, Na+/K+-ATPase, Na+/H+ exchanger, and amino acid transport System A. Of note, the modulation of these transport systems is cell-type and developmental stage-dependent. In particular, dysregulation of Na+/K+-ATPase activity is involved in several pathological situations, from viral infection to cancer. Therefore, this transport system represents a promising pharmacological tool in these pathologies.


Asunto(s)
Neoplasias , Triyodotironina , Adenosina Trifosfatasas/metabolismo , Sistema de Transporte de Aminoácidos A , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glucosa , Humanos , Integrinas/metabolismo , Mitógenos , Neoplasias/metabolismo , Proteína Quinasa C/metabolismo , Hormonas Tiroideas/metabolismo , Tiroxina/metabolismo , Triyodotironina/fisiología
4.
Front Cell Dev Biol ; 9: 651492, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33898447

RESUMEN

Interaction between thyroid hormones and the immune system is reported in the literature. Thyroid hormones, thyroxine, T4, but also T3, act non-genomically through mechanisms that involve a plasma membrane receptor αvß3 integrin, a co-receptor for insulin-like growth factor-1 (IGF-1). Previous data from our laboratory show a crosstalk between thyroid hormones and IGF-1 because thyroid hormones inhibit the IGF-1-stimulated glucose uptake and cell proliferation in L-6 myoblasts, and the effects are mediated by integrin αvß3. IGF-1 also behaves as a chemokine, being an important factor for tissue regeneration after damage. In the present study, using THP-1 human leukemic monocytes, expressing αvß3 integrin in their cell membrane, we focused on the crosstalk between thyroid hormones and either IGF-1 or monocyte chemoattractant protein-1 (MCP-1), studying cell migration and proliferation stimulated by the two chemokines, and the role of αvß3 integrin, using inhibitors of αvß3 integrin and downstream pathways. Our results show that IGF-1 is a potent chemoattractant in THP-1 monocytes, stimulating cell migration, and thyroid hormone inhibits the effect through αvß3 integrin. Thyroid hormone also inhibits IGF-1-stimulated cell proliferation through αvß3 integrin, an example of a crosstalk between genomic and non-genomic effects. We also studied the effects of thyroid hormone on cell migration and proliferation induced by MCP-1, together with the pathways involved, by a pharmacological approach and docking simulation. Our findings show a different downstream signaling for IGF-1 and MCP-1 in THP-1 monocytes mediated by the plasma membrane receptor of thyroid hormones, integrin αvß3.

5.
Viruses ; 14(1)2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-35062278

RESUMEN

Plasmacytoid dendritic cells (pDCs) are a unique dendritic cell subset specialized in type I interferon production, whose role in Human Immunodeficiency Virus (HIV) infection and pathogenesis is complex and not yet well defined. Considering the crucial role of the accessory protein Nef in HIV pathogenicity, possible alterations in intracellular signalling and extracellular vesicle (EV) release induced by exogenous Nef on uninfected pDCs have been investigated. As an experimental model system, a human plasmacytoid dendritic cell line, GEN2.2, stimulated with a myristoylated recombinant NefSF2 protein was employed. In GEN2.2 cells, Nef treatment induced the tyrosine phosphorylation of STAT-1 and STAT-2 and the production of a set of cytokines, chemokines and growth factors including IP-10, MIP-1ß, MCP-1, IL-8, TNF-α and G-CSF. The released factors differed both in type and amount from those released by macrophages treated with the same viral protein. Moreover, Nef treatment slightly reduces the production of small EVs, and the protein was found associated with the small (size < 200 nm) but not the medium/large vesicles (size > 200 nm) collected from GEN2.2 cells. These results add new information on the interactions between this virulence factor and uninfected pDCs, and may provide the basis for further studies on the interactions of Nef protein with primary pDCs.


Asunto(s)
Citocinas/metabolismo , Células Dendríticas/metabolismo , Vesículas Extracelulares/metabolismo , VIH-1/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Línea Celular , Quimiocinas/metabolismo , Células Dendríticas/virología , Infecciones por VIH/virología , Humanos , Macrófagos/metabolismo , Proteínas Recombinantes , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología
6.
J Agric Food Chem ; 68(52): 15428-15439, 2020 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-33305574

RESUMEN

Some constituents of the Mediterranean diet, such as extra-virgin olive oil (EVOO) contain substances such as hydroxytyrosol (HT) and its metabolite homovanillic alcohol (HA). HT has aroused much interest due to its antioxidant activity as a radical scavenger, whereas only a few studies have been made on the HA molecule. Both chemical synthesis and extraction techniques have been developed to obtain these molecules, with each method having its advantages and drawbacks. In this study, we report the use of tyrosol from olive mill wastewaters as a starting molecule to synthesize HT and HA, using a sustainable procedure characterized by high efficiency and low cost. The effects of HT and HA were evaluated on two cell lines, THP-1 human leukemic monocytes and L-6 myoblasts from rat skeletal muscle, after treating the cells with a radical generator. Both HT and HA efficiently inhibited ROS production. In particular, HT inhibited the proliferation of the THP-1 leukemic monocytes, while HA protected L-6 myoblasts from cytotoxicity.


Asunto(s)
Antioxidantes/aislamiento & purificación , Ácido Homovanílico/aislamiento & purificación , Alcohol Feniletílico/análogos & derivados , Extractos Vegetales/aislamiento & purificación , Aguas Residuales/química , Animales , Antioxidantes/química , Línea Celular , Proliferación Celular/efectos de los fármacos , Ácido Homovanílico/química , Humanos , Olea/química , Aceite de Oliva/química , Alcohol Feniletílico/sangre , Alcohol Feniletílico/aislamiento & purificación , Extractos Vegetales/química , Ratas , Especies Reactivas de Oxígeno/metabolismo , Residuos/análisis
7.
Viruses ; 12(5)2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32456011

RESUMEN

Extracellular vesicles (EVs) are lipid bilayer-enclosed entities containing proteins and nucleic acids that mediate intercellular communication, in both physiological and pathological conditions. EVs resemble enveloped viruses in both structural and functional aspects. In full analogy with viral biogenesis, some of these vesicles are generated inside cells and, once released into the extracellular milieu, are called "exosomes". Others bud from the plasma membrane and are generally referred to as "microvesicles". In this review, we will discuss the state of the art of the current studies on the relationship between EVs and viruses and their involvement in three important viral infections caused by HIV, HCV and Severe Acute Respiratory Syndrome (SARS) viruses. HIV and HCV are two well-known pathogens that hijack EVs content and release to create a suitable environment for viral infection. SARS viruses are a new entry in the world of EVs studies, but are equally important in this historical framework. A thorough knowledge of the involvement of the EVs in viral infections could be helpful for the development of new therapeutic strategies to counteract different pathogens.


Asunto(s)
Vesículas Extracelulares/metabolismo , Infecciones por VIH/metabolismo , Hepatitis C/metabolismo , Síndrome Respiratorio Agudo Grave/metabolismo , Comunicación Celular , Coronavirus , Infecciones por Coronavirus/metabolismo , Exosomas , VIH-1 , Hepacivirus , Humanos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Virosis/metabolismo
8.
Front Cell Dev Biol ; 8: 614030, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33553149

RESUMEN

The interdependence between thyroid hormones (THs), namely, thyroxine and triiodothyronine, and immune system is nowadays well-recognized, although not yet fully explored. Synthesis, conversion to a bioactive form, and release of THs in the circulation are events tightly supervised by the hypothalamic-pituitary-thyroid (HPT) axis. Newly synthesized THs induce leukocyte proliferation, migration, release of cytokines, and antibody production, triggering an immune response against either sterile or microbial insults. However, chronic patho-physiological alterations of the immune system, such as infection and inflammation, affect HPT axis and, as a direct consequence, THs mechanism of action. Herein, we revise the bidirectional crosstalk between THs and immune cells, required for the proper immune system feedback response among diverse circumstances. Available circulating THs do traffic in two distinct ways depending on the metabolic condition. Mechanistically, internalized THs form a stable complex with their specific receptors, which, upon direct or indirect binding to DNA, triggers a genomic response by activating transcriptional factors, such as those belonging to the Wnt/ß-catenin pathway. Alternatively, THs engage integrin αvß3 receptor on cell membrane and trigger a non-genomic response, which can also signal to the nucleus. In addition, we highlight THs-dependent inflammasome complex modulation and describe new crucial pathways involved in microRNA regulation by THs, in physiological and patho-physiological conditions, which modify the HPT axis and THs performances. Finally, we focus on the non-thyroidal illness syndrome in which the HPT axis is altered and, in turn, affects circulating levels of active THs as reported in viral infections, particularly in immunocompromised patients infected with human immunodeficiency virus.

9.
Cytokine Growth Factor Rev ; 51: 49-60, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31874738

RESUMEN

Extracellular vesicles (EVs) are small membrane-bound particles that are naturally released from cells. They are recognized as potent vehicles of intercellular communication both in prokaryotes and eukaryotes. Because of their capacity to carry biological macromolecules such as proteins, lipids and nucleic acids, EVs influence different physiological and pathological functions of both parental and recipient cells. Although multiple pathways have been proposed for cytokine secretion beyond the classical ER/Golgi route, EVs have recently recognized as an alternative secretory mechanism. Interestingly, cytokines/chemokines exploit these vesicles to be released into the extracellular milieu, and also appear to modulate their release, trafficking and/or content. In this review, we provide an overview of the cytokines/chemokines that are known to be associated with EVs or their regulation with a focus on TNFα, IL-1ß and IFNs.


Asunto(s)
Comunicación Celular/inmunología , Citocinas/inmunología , Vesículas Extracelulares/metabolismo , Animales , Quimiocinas/inmunología , Regulación de la Expresión Génica , Humanos , Interferones/inmunología , Ratones
10.
Int J Nanomedicine ; 14: 8755-8768, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31806970

RESUMEN

PURPOSE: Single-chain variable fragments (scFvs) are one of the smallest antigen-binding units having the invaluable advantage to be expressed by a unique short open reading frame (ORF). Despite their reduced size, spontaneous cell entry of scFvs remains inefficient, hence precluding the possibility to target intracellular antigens. Here, we describe an original strategy to deliver scFvs inside target cells through engineered extracellular vesicles (EVs). This approach relies on the properties of a Human Immunodeficiency Virus (HIV)-1 Nef mutant protein referred to as Nefmut. It is a previously characterized Nef allele lacking basically all functions of wt Nef, yet strongly accumulating in the EV lumen also when fused at its C-terminus with a foreign protein. To gain the proof-of-principle for the efficacy of the proposed strategy, the tumor-promoting Human Papilloma Virus (HPV)16-E7 protein was considered as a scFv-specific intracellular target. The oncogenic effect of HPV16-E7 relies on its binding to the tumor suppressor pRb protein leading to a dysregulated cell duplication. Interfering with this interaction means impairing the HPV16-E7-induced cell proliferation. METHODS: The Nefmut gene was fused in frame at its 3'-terminus with the ORF coding for a previously characterized anti-HPV16-E7 scFv. Interaction between the Nefmut-fused anti-HPV16-E7 scFv and the HPV16-E7 protein was tested by both confocal microscope and co-immunoprecipitation analyses on co-transfected cells. The in cis anti-proliferative effect of the Nefmut/anti-HPV16-E7 scFv was assayed by transfecting HPV16-infected cells. The anti-proliferative effect of EVs engineered with Nefmut/anti-HPV16-E7 scFv on HPV16-E7-expressing cells was evaluated in two ways: i) through challenge with purified EVs by a Real-Time Cell Analysis system and ii) in transwell co-cultures by an MTS-based assay. RESULTS: The Nefmut/anti-HPV16-E7 scFv chimeric product is efficiently uploaded in EVs, binds HPV16-E7, and inhibits the proliferation of HPV16-E7-expressing cells. Most important, challenge with cell-free EVs incorporating the Nefmut/anti-HPV16-E7 scFv led to the inhibition of proliferation of HPV16-E7-expressing cells. The proliferation of these cells was hindered also when they were co-cultured in transwells with cells producing EVs uploading Nefmut/anti-HPV16-E7 scFv. CONCLUSION: Our data represent the proof-of-concept for the possibility to target intracellular antigens through EV-mediated delivery of scFvs. This finding could be relevant to design novel methods of intracellular therapeutic interventions.


Asunto(s)
Vesículas Extracelulares/inmunología , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Anticuerpos de Cadena Única/administración & dosificación , Efecto Espectador , Línea Celular , Proliferación Celular , Técnicas de Cocultivo , Exosomas/inmunología , Exosomas/metabolismo , Vesículas Extracelulares/genética , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 16/patogenicidad , Humanos , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/prevención & control , Anticuerpos de Cadena Única/genética , Transfección , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
11.
Cell Div ; 14: 3, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31007707

RESUMEN

BACKGROUND: Resveratrol and its natural stilbene-containing derivatives have been extensively investigated as potential chemotherapeutic agents. The synthetic manipulation of the stilbene scaffold has led to the generation of new analogues with improved anticancer activity and better bioavailability. In the present study we investigated the anticancer activity of a novel trimethoxystilbene derivative (3,4,4'-trimethoxylstilbene), where two methoxyl groups are adjacent on the benzene ring (ortho configuration), and compared its activity to 3,5,4'-trimethoxylstilbene, whose methoxyl groups are in meta configuration. RESULTS: We provide evidence that the presence of the two methoxyl groups in ortho configuration renders 3,4,4'-trimethoxystilbene more efficient than the meta isomer in inhibiting cell proliferation and producing apoptotic death in colorectal cancer cells. Confocal microscopy of α- and γ-tubulin staining shows that the novel compound strongly depolymerizes the mitotic spindle and produces fragmentation of the pericentrosomal material. Computer assisted docking studies indicate that both molecules potentially interact with γ-tubulin, and that 3,4,4'-trimethoxystilbene is likely to establish stronger interactions with the protein. CONCLUSIONS: These findings demonstrate the ortho configuration confers higher specificity for γ-tubulin with respect to α-tubulin on 3,4,4' trimethoxystilbene, allowing it to be defined as a new γ-tubulin inhibitor. A strong interaction with γ-tubulin might be a defining feature of molecules with high anticancer activity, as shown for the 3,4,4' isomer.

12.
Cytokine Growth Factor Rev ; 40: 77-89, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29588163

RESUMEN

Plasmacytoid dendritic cells (pDCs) are a unique dendritic cell subset that are specialized in type I interferon (IFN) production. pDCs are key players in the antiviral immune response and serve as bridge between innate and adaptive immunity. Although pDCs do not represent the main reservoir of the Human Immunodeficiency Virus (HIV), they are a crucial subset in HIV infection as they influence viral transmission, target cell infection and antigen presentation. pDCs act as inflammatory and immunosuppressive cells, thus contributing to HIV disease progression. This review provides a state of art analysis of the interactions between HIV and pDCs and their potential roles in HIV transmission, chronic immune activation and immunosuppression. A thorough understanding of the roles of pDCs in HIV infection will help to improve therapeutic strategies to fight HIV infection, and will further increase our knowledge on this important immune cell subset.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Infecciones por VIH/inmunología , VIH-1/inmunología , Interferón Tipo I/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/patología , Infecciones por VIH/transmisión , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-28785545

RESUMEN

The opportunistic pathogen Staphylococcus aureus (S. aureus) is a major cause of nosocomial- and community-acquired infections. In addition, many antibiotic-resistant strains are emerging worldwide, thus, there is an urgent unmet need to pinpoint novel therapeutic and prophylactic strategies. In the present study, we characterized the impact of infection with the pandemic methicillin-resistant USA300 S. aureus strain on human primary dendritic cells (DC), key initiators and regulators of immune responses. In particular, among staphylococcal virulence factors, the function of EsxA and EsxB, two small acidic dimeric proteins secreted by the type VII-like secretion system Ess (ESAT-6-like secretion system), was investigated in human DC setting. A comparative analysis of bacterial entry, replication rate as well as DC maturation, apoptosis, signaling pathway activation and cytokine production was performed by using wild type (wt) USA300 and three isogenic mutants carrying the deletion of esxA (ΔesxA), esxB (ΔesxB), or both genes (ΔesxAB). The S. aureus mutant lacking only the EsxA protein (ΔesxA) stimulated a stronger pro-apoptotic phenotype in infected DC as compared to wt USA300, ΔesxAB, and ΔesxB strains. When the mutant carrying the esxB deletion (ΔesxB) was analyzed, a higher production of both regulatory and pro-inflammatory mediators was found in the infected DC with respect to those challenged with the wt counterpart and the other esx mutants. In accordance with these data, supernatant derived from ΔesxB-infected DC promoted a stronger release of both IFN-γ and IL-17 from CD4+ T cells as compared with those conditioned with supernatants derived from wild type USA300-, ΔesxAB-, and ΔesxA-infected cultures. Although, the interaction of S. aureus with human DC is not yet fully understood, our data suggest that both cytokine production and apoptotic process are modulated by Esx factors, thus indicating a possible role of these proteins in the modulation of DC-mediated immunity to S. aureus.


Asunto(s)
Proteínas Bacterianas/metabolismo , Citocinas/metabolismo , Células Dendríticas/inmunología , Interacciones Huésped-Patógeno , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/inmunología , Factores de Virulencia/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Células Cultivadas , Medios de Cultivo Condicionados , Células Dendríticas/microbiología , Eliminación de Gen , Humanos , Staphylococcus aureus/genética , Células TH1/inmunología , Células Th17/inmunología , Factores de Virulencia/genética , Factores de Virulencia/inmunología
14.
Mol Carcinog ; 56(3): 1117-1126, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27739192

RESUMEN

Natural compounds are extensively studied for their potential use in traditional and non-traditional medicine. Several natural and synthetic Resveratrol analogues have shown interesting biological activities in the field of cancer chemoprevention. In the present study, we have focused on the ability of Resveratrol and two methoxylated derivatives (Trimethoxystilbene and Pterostilbene) to inhibit human cancer cell growth particularly analyzing their ability to interfere with tubulin dynamics at mitosis. We show that Trimethoxystilbene, differently from Resveratrol and Pterostilbene, alters microtubule polymerization dynamics in HeLa cells specifically inducing multipolar spindles and mitotic arrest coupled to a reduction of cell growth and an increase in apoptotic death by mitotic catastrophe. This work demonstrates that the structural modification of Rsv causes substantial changes in the mechanism of action of the derivatives. The presence of three extra methyl groups renders Trimethoxy very efficient in impairing cell proliferation by inducing mitotic catastrophe in cancer cells. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Mitosis/efectos de los fármacos , Neoplasias/genética , Estilbenos/farmacología , Tubulina (Proteína)/metabolismo , Animales , Células CHO , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cricetulus , Células HeLa , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Resveratrol , Estilbenos/química , Tubulina (Proteína)/efectos de los fármacos
15.
Oncotarget ; 7(13): 15539-53, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26788990

RESUMEN

Single-chain variable fragments (scFvs) expressed as "intracellular antibodies" (intrabodies) can target intracellular antigens to hamper their function efficaciously and specifically. Here we use an intrabody targeting the E6 oncoprotein of Human papillomavirus 16 (HPV16) to address the issue of a non-invasive therapy for HPV cancer patients.A scFv against the HPV16 E6 was selected by Intracellular Antibody Capture Technology and expressed as I7nuc in the nucleus of HPV16-positive SiHa, HPV-negative C33A and 293T cells. Colocalization of I7nuc and recombinant E6 was observed in different cell compartments, obtaining evidence of E6 delocalization ascribable to I7nuc. In SiHa cells, I7nuc expressed by pLNCX retroviral vector was able to partially inhibit degradation of the main E6 target p53, and induced p53 accumulation in nucleus. When analyzing in vitro activity on cell proliferation and survival, I7nuc was able to decrease growth inducing late apoptosis and necrosis of SiHa cells.Finally, I7nuc antitumor activity was demonstrated in two pre-clinical models of HPV tumors. C57BL/6 mice were injected subcutaneously with HPV16-positive TC-1 or C3 tumor cells, infected with pLNCX retroviral vector expressing or non-expressing I7nuc. All the mice injected with I7nuc-expressing cells showed a clear delay in tumor onset; 60% and 40% of mice receiving TC-1 and C3 cells, respectively, remained tumor-free for 17 weeks of follow-up, whereas 100% of the controls were tumor-bearing 20 days post-inoculum. Our data support the therapeutic potential of E6-targeted I7nuc against HPV tumors.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Experimentales/virología , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Anticuerpos de Cadena Única/farmacología , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos C57BL , Infecciones por Papillomavirus/complicaciones
16.
PLoS One ; 10(6): e0130189, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26066624

RESUMEN

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.


Asunto(s)
Macrófagos/patología , Microglía/patología , Ácido Mirístico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/metabolismo , Animales , Western Blotting , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Humanos , Técnicas para Inmunoenzimas , Interferón gamma/genética , Interferón gamma/metabolismo , Macrófagos/metabolismo , Ratones , Microglía/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Fosforilación , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
17.
Cytokine Growth Factor Rev ; 26(2): 159-73, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25529283

RESUMEN

Several viruses manipulate host innate immune responses to avoid immune recognition and improve viral replication and spreading. The viral protein Nef of Human Immunodeficiency Virus is mainly involved in this "hijacking" activity and is a well established virulence factor. In the last few years there have been remarkable advances in outlining a defined framework of its functions. In particular Nef appears to be a shuttling molecular adaptor able to exert its effects both on infected and non infected bystander cell. In addition it is emerging fact that it has an important impact on the chemo-cytokine network. Nef protein represents an interesting new target to develop therapeutic drugs for treatment of seropositive patients. In this review we have tried to provide a unifying view of the multiple functions of this viral protein on the basis of recently available experimental data.


Asunto(s)
Citocinas/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/fisiología , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/fisiología , Antígenos CD4/inmunología , Citocinas/metabolismo , VIH-1/inmunología , Humanos , Macrófagos/inmunología , Transducción de Señal , Factor de Necrosis Tumoral alfa/sangre , Replicación Viral
18.
Int J Cancer ; 134(11): 2742-7, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24226851

RESUMEN

Human papillomavirus (HPV)-associated tumors still represent an urgent problem of public health in spite of the efficacy of the prophylactic HPV vaccines. Specific antibodies in single-chain format expressed as intracellular antibodies (intrabodies) are valid tools to counteract the activity of target proteins. We previously showed that the M2SD intrabody, specific for the E7 oncoprotein of HPV16 and expressed in the endoplasmic reticulum of the HPV16-positive SiHa cells, was able to inhibit cell proliferation. Here, we showed by confocal microscopy that M2SD and E7 colocalize in the endoplasmic reticulum of SiHa cells, suggesting that the E7 delocalization mediated by M2SD could account for the anti-proliferative activity of the intrabody. We then tested the M2SD antitumor activity in two mouse models for HPV tumors based respectively on TC-1 and C3 cells. The M2SD intrabody was delivered by retroviral vector to tumor cells before cell injection into C57BL/6 mice. In both models, a marked delay of tumor onset with respect to the controls was observed in all the mice injected with the M2SD-expressing tumor cells and, importantly, a significant percentage of mice remained tumor-free permanently. This is the first in vivo demonstration of the antitumor activity of an intrabody directed towards an HPV oncoprotein. We consider that these results could contribute to the development of new therapeutic molecules based on antibodies in single-chain format, to be employed against the HPV-associated lesions even in combination with other drugs.


Asunto(s)
Anticuerpos Antivirales/administración & dosificación , Papillomavirus Humano 16/inmunología , Proteínas E7 de Papillomavirus/antagonistas & inhibidores , Infecciones por Papillomavirus/terapia , Anticuerpos de Cadena Única/administración & dosificación , Neoplasias del Cuello Uterino/terapia , Animales , Anticuerpos Antivirales/genética , Anticuerpos Antivirales/inmunología , Proliferación Celular , Femenino , Terapia Genética , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/inmunología , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/virología , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Tasa de Supervivencia , Células Tumorales Cultivadas , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/virología
19.
Peptides ; 50: 100-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24120989

RESUMEN

Atrial natriuretic peptide (ANP), a cardiovascular hormone, elicits different biological actions in the immune system. The aim of the present study was to investigate in THP-1 monocytes the ANP effect on hydrogen peroxide (H2O2)-induced Reactive Oxygen Species (ROS), cell proliferation and migration. A significant increase of H2O2-dependent ROS production was induced by physiological concentration of ANP (10(-10)M). The ANP action was partially affected by cell pretreatment with PD98059, an inhibitor of mitogen activated-protein kinases (MAPK) as well as by wortmannin, an inhibitor of phosphatidylinositol 3-kinase (PI3K) and totally suppressed by diphenylene iodonium (DPI), an inhibitor of the enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. The hormone effect was mimicked by cANF and an ANP/NPR-C signaling pathway was studied using pertussis toxin (PTX). A significant increase of H2O2-induced cell migration was observed after ANP (10(-10)M) treatment, conversely a decrease of THP-1 proliferation, due to cell death, was found. Both ANP actions were partially prevented by DPI. Moreover, H2O2-induced release of IL-9, TNF-α, MIP-1α and MIP-1ß was not counteracted by DPI, whereas no effect was observed in any experimental condition for both IL-6 and IL-1ß. Our results support the view that ANP can play a key role during the inflammatory process.


Asunto(s)
Factor Natriurético Atrial/farmacología , Peróxido de Hidrógeno/farmacología , Monocitos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Androstadienos/farmacología , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/metabolismo , Flavonoides/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/citología , Monocitos/metabolismo , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Compuestos Onio/farmacología , Fragmentos de Péptidos/farmacología , Toxina del Pertussis/farmacología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Especies Reactivas de Oxígeno/agonistas , Transducción de Señal , Wortmanina
20.
PLoS One ; 7(5): e36909, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615843

RESUMEN

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.


Asunto(s)
Transformación Celular Viral , Interferón beta/metabolismo , Queratinocitos/metabolismo , Papillomaviridae/fisiología , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Ciclo Celular/genética , Procesos de Crecimiento Celular/genética , Células Cultivadas , Senescencia Celular/genética , Silenciador del Gen , Humanos , Interferón beta/genética , Queratinocitos/virología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Papillomaviridae/genética , Papillomaviridae/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Fosforilación , Proteína de la Leucemia Promielocítica , Procesamiento Proteico-Postraduccional , Transactivadores/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA