Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Physiol Cell Physiol ; 324(4): C821-C836, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36802732

RESUMEN

Pericytes are mural cells that play an important role in regulation of angiogenesis and endothelial function. Cadherins are a superfamily of adhesion molecules mediating Ca2+-dependent homophilic cell-cell interactions that control morphogenesis and tissue remodeling. To date, classical N-cadherin is the only cadherin described on pericytes. Here, we demonstrate that pericytes also express T-cadherin (H-cadherin, CDH13), an atypical glycosyl-phosphatidylinositol (GPI)-anchored member of the superfamily that has previously been implicated in regulation of neurite guidance, endothelial angiogenic behavior, and smooth muscle cell differentiation and progression of cardiovascular disease. The aim of the study was to investigate T-cadherin function in pericytes. Expression of T-cadherin in pericytes from different tissues was performed by immunofluorescence analysis. Using lentivirus-mediated gain-of-function and loss-of-function in cultured human pericytes, we demonstrate that T-cadherin regulates pericyte proliferation, migration, invasion, and interactions with endothelial cells during angiogenesis in vitro and in vivo. T-cadherin effects are associated with the reorganization of the cytoskeleton, modulation of cyclin D1, α-smooth muscle actin (αSMA), integrin ß3, metalloprotease MMP1, and collagen expression levels, and involve Akt/GSK3ß and ROCK intracellular signaling pathways. We also report the development of a novel multiwell 3-D microchannel slide for easy analysis of sprouting angiogenesis from a bioengineered microvessel in vitro. In conclusion, our data identify T-cadherin as a novel regulator of pericyte function and support that it is required for pericyte proliferation and invasion during active phase of angiogenesis, while T-cadherin loss shifts pericytes toward the myofibroblast state rendering them unable to control endothelial angiogenic behavior.


Asunto(s)
Células Endoteliales , Pericitos , Humanos , Pericitos/metabolismo , Células Endoteliales/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Morfogénesis , Neovascularización Fisiológica
2.
EMBO Rep ; 19(5)2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29643120

RESUMEN

Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis, whose best-understood mechanism is sprouting. However, therapeutic VEGF delivery to ischemic muscle induces angiogenesis by the alternative process of intussusception, or vascular splitting, whose molecular regulation is essentially unknown. Here, we identify ephrinB2/EphB4 signaling as a key regulator of intussusceptive angiogenesis and its outcome under therapeutically relevant conditions. EphB4 signaling fine-tunes the degree of endothelial proliferation induced by specific VEGF doses during the initial stage of circumferential enlargement of vessels, thereby limiting their size and subsequently enabling successful splitting into normal capillary networks. Mechanistically, EphB4 neither inhibits VEGF-R2 activation by VEGF nor its internalization, but it modulates VEGF-R2 downstream signaling through phospho-ERK1/2. In vivo inhibitor experiments show that ERK1/2 activity is required for EphB4 regulation of VEGF-induced intussusceptive angiogenesis. Lastly, after clinically relevant VEGF gene delivery with adenoviral vectors, pharmacological stimulation of EphB4 normalizes dysfunctional vascular growth in both normoxic and ischemic muscle. These results identify EphB4 as a druggable target to modulate the outcome of VEGF gene delivery and support further investigation of its therapeutic potential.


Asunto(s)
Efrina-B2/metabolismo , Mioblastos/metabolismo , Neovascularización Patológica/metabolismo , Receptor EphB4/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Humanos , Intususcepción , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Músculo Esquelético/patología , Neovascularización Patológica/patología , Fosforilación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
3.
J Virol ; 88(12): 6751-61, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24696478

RESUMEN

UNLABELLED: High-risk types of human papillomavirus (HPV) are the causative agents of virtually all cases of cervical cancer and a significant proportion of other anogenital cancers, as well as both oral and pharyngeal cancers. The high-risk types encode two viral oncogenes, E6 and E7, which work together to initiate cell transformation. Multiple steps involving the activities and interactions of both viral and cellular proteins are involved in the progression from HPV infection to cell transformation to cancer. The E6 oncoprotein is expressed as several isoforms: a full-length variant referred to as E6 and a few shorter isoforms collectively referred to as E6*. In this study, we found that expression of E6* increased the level of reactive oxygen species (ROS) in both HPV-positive and HPV-negative cells. This increased oxidative stress led to higher levels of DNA damage, as assessed by the comet assay, quantification of 8-oxoguanine, and poly(ADP-ribose) polymerase 1. The observed increase in ROS may be due to a decrease in cellular antioxidant activity, as we found that E6* expression also led to decreased expression of superoxide dismutase isoform 2 and glutathione peroxidase. These studies indicate that E6* may play an important role in virus-induced mutagenesis by increasing oxidative stress and DNA damage. IMPORTANCE: Our findings demonstrate for the first time that an HPV gene product, E6*, can increase ROS levels in host cells. This ability may play a significant role both in the viral life cycle and in cancer development, because an increase in oxidative DNA damage may both facilitate HPV genome amplification and increase the probability of HPV16 DNA integration. Integration, in turn, is thought to be an important step in HPV-mediated carcinogenesis.


Asunto(s)
Daño del ADN , Papillomavirus Humano 16/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Estrés Oxidativo , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular , Papillomavirus Humano 16/genética , Humanos , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/virología , Especies Reactivas de Oxígeno , Proteínas Represoras/genética
4.
Lab Chip ; 24(2): 292-304, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-38086670

RESUMEN

Leukocyte recruitment from blood to tissue is a process that occurs at the level of capillary vessels during both physiological and pathological conditions. This process is also relevant for evaluating novel adoptive cell therapies, in which the trafficking of therapeutic cells such as chimeric antigen receptor (CAR)-T cells throughout the capillaries of solid tumors is important. Local variations in blood flow, mural cell concentration, and tissue stiffness contribute to the regulation of capillary vascular permeability and leukocyte trafficking throughout the capillary microvasculature. We developed a platform to mimic a biologically functional human arteriole-venule microcirculation system consisting of pericytes (PCs) and arterial and venous primary endothelial cells (ECs) embedded within a hydrogel, which self-assembles into a perfusable, heterogeneous microvasculature. Our device shows a preferential association of PCs with arterial ECs that drives the flow-dependent formation of microvasculature networks. We show that PCs stimulate basement membrane matrix synthesis, which affects both vessel diameter and permeability in a manner correlating with the ratio of ECs to PCs. Moreover, we demonstrate that hydrogel concentration can affect capillary morphology but has no observed effect on vascular permeability. The biological function of our capillary network was demonstrated using an inflammation model, where significantly higher expression of cytokines, chemokines, and adhesion molecules was observed after tumor necrosis factor-alpha (TNF-α) treatment. Accordingly, T cell adherence and transendothelial migration were significantly increased in the immune-activated state. Taken together, our platform allows the generation of a perfusable microvasculature that recapitulates the structure and function of an in vivo capillary bed that can be used as a model for developing potential immunotherapies.


Asunto(s)
Células Endoteliales , Microvasos , Humanos , Microvasos/metabolismo , Capilares/fisiología , Leucocitos , Hidrogeles/metabolismo
5.
Biochim Biophys Acta ; 1821(7): 943-53, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22349266

RESUMEN

DNA damaging agents typically induce an apoptotic cascade in which p53 plays a central role. However, absence of a p53-mediated response does not necessarily abrogate programmed cell death, due to the existence of p53-independent apoptotic pathways, such as those mediated by the pro-apoptotic molecule ceramide. We compared ceramide levels before and after DNA damage in human osteosarcoma (U2OS) and colon cancer (HCT116) cells that were either expressing or deficient in p53. When treated with mitomycin C, p53-deficient cells, but not p53-expressing cells, showed a marked increase in ceramide levels. Microarray analysis of genes involved in ceramide metabolism identified acid ceramidase (ASAH1, up-regulated), ceramide glucosyltransferase (UGCG, down-regulated), and galactosylceramidase (GALC, up-regulated) as the three genes most affected. Experiments employing pharmacological and siRNA agents revealed that inhibition of UGCG is sufficient to increase ceramide levels and induce cell death. When inhibition of UGCG and treatment with mitomycin C were combined, p53-deficient, but not p53-expressing cells, showed a significant increase in cell death, suggesting that the regulation of sphingolipid metabolism could be used to sensitize cells to chemotherapeutic drugs.


Asunto(s)
Ceramidas/biosíntesis , Daño del ADN , Glucosiltransferasas/genética , Proteína p53 Supresora de Tumor/genética , Ceramidasa Ácida/genética , Ceramidasa Ácida/metabolismo , Apoptosis/genética , Neoplasias Óseas , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ceramidas/agonistas , Neoplasias del Colon , Galactosilceramidasa/genética , Galactosilceramidasa/metabolismo , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glucosiltransferasas/metabolismo , Humanos , Mitomicina/farmacología , Osteosarcoma , ARN Interferente Pequeño/genética , Transfección , Proteína p53 Supresora de Tumor/deficiencia
6.
J Biol Chem ; 286(15): 13657-68, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21335548

RESUMEN

Arrest of cell differentiation is one of the leading causes of leukemia and other cancers. Induction of cell differentiation using pharmaceutical agents has been clinically attempted for the treatment of these cancers. Epigenetic regulation may be one of the underlying molecular mechanisms controlling cell proliferation or differentiation. Here, we report on the use of proteomics-based differential protein expression analysis in conjunction with quantification of histone modifications to decipher the interconnections among epigenetic modifications, their modifying enzymes or mediators, and changes in the associated pathways/networks that occur during cell differentiation. During phorbol-12-myristate 13-acetate-induced differentiation of U937 cells, fatty acid synthesis and its metabolic processing, the clathrin-coated pit endocytosis pathway, and the ubiquitin/26 S proteasome degradation pathways were up-regulated. In addition, global histone H3/H4 acetylation and H2B ubiquitination were down-regulated concomitantly with impaired chromatin remodeling machinery, RNA polymerase II complexes, and DNA replication. Differential protein expression analysis established the networks linking histone hypoacetylation to the down-regulated expression/activity of p300 and linking histone H2B ubiquitination to the RNA polymerase II-associated FACT-RTF1-PAF1 complex. Collectively, our approach has provided an unprecedentedly systemic set of insights into the role of epigenetic regulation in leukemia cell differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Epigénesis Genética/fisiología , Regulación de la Expresión Génica/fisiología , Perfilación de la Expresión Génica/métodos , Humanos , Espectrometría de Masas/métodos , Células U937
7.
Bioorg Med Chem Lett ; 22(5): 2125-9, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22300659

RESUMEN

High-risk strains of human papillomaviruses (HPVs) cause nearly all cases of cervical cancer as well as a growing number of head and neck cancers. The oncogenicity of these viruses can be attributed to the activities of their two primary oncoproteins, E6 and E7. The E6 protein has among its functions the ability to prevent apoptosis of infected cells through its binding to FADD and caspase 8. A small molecule library was screened for candidates that could inhibit E6 binding to FADD and caspase 8. Flavonols were found to possess this activity with the rank order of myricetin>morin>quercetin>kaempferol=galangin≫(apigenin, 7-hydroxyflavonol, rhamnetin, isorhamnetin, geraldol, datiscetin, fisetin, 6-hydroxyflavonol). Counter screening, where the ability of these chosen flavonols to inhibit caspase 8 binding to itself was assessed, demonstrated that myricetin, morin and quercetin inhibited GST-E6 and His-caspase 8 binding in a specific manner. The structure-activity relationships suggested by these data are unique and do not match prior reports on flavonols in the literature for a variety of anticancer assays.


Asunto(s)
Inhibidores de Caspasas , Interacciones Huésped-Patógeno/efectos de los fármacos , Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Apoptosis/efectos de los fármacos , Caspasa 8/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/antagonistas & inhibidores , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Femenino , Flavonas/química , Flavonas/farmacología , Flavonoles/química , Flavonoles/farmacología , Papillomavirus Humano 16/efectos de los fármacos , Humanos , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/virología , Proteínas Represoras/metabolismo , Neoplasias del Cuello Uterino/virología
8.
J Virol ; 84(3): 1453-63, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19906919

RESUMEN

Human papillomavirus 16 is a causative agent of most cases of cervical cancer and has also been implicated in the development of some head and neck cancers. The early viral E6 gene codes for two alternatively spliced isoforms, E6(large) and E6*. We have previously demonstrated the differential effects of E6(large) and E6* binding on the expression and stability of procaspase 8, a key mediator of the apoptotic pathway. Additionally, we have reported that E6 binds to the FADD death effector domain (DED) at a novel E6 binding domain. Sequence similarities between the FADD and procaspase 8 DEDs suggested a specific region for E6(large)/procaspase 8 binding, which was subsequently confirmed by mutational analysis as well as by the ability of peptides capable of blocking E6/FADD binding to also block E6(large)/caspase 8 binding. However, the binding of the smaller isoform, E6*, to procaspase 8 occurs at a different region, as deletion and point mutations that disrupt E6(large)/caspase 8 DED binding do not disrupt E6*/caspase 8 DED binding. In addition, peptide inhibitors that can block E6(large)/procaspase 8 binding do not affect the binding of E6* to procaspase 8. These results demonstrate that the residues that mediate E6*/procaspase 8 DED binding localize to a different region on the protein and employ a separate binding motif. This provides a molecular explanation for our initial findings that the two E6 isoforms affect procaspase 8 stability in an opposing manner.


Asunto(s)
Caspasa 8/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Empalme del ARN , Proteínas Represoras/metabolismo , Apoptosis/fisiología , Sitios de Unión , Western Blotting , Línea Celular Tumoral , Electroforesis en Gel de Poliacrilamida , Humanos , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Proteínas Represoras/química , Proteínas Represoras/genética , Factor de Necrosis Tumoral alfa/fisiología
9.
Cytokine ; 56(2): 272-81, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21843952

RESUMEN

Interleukin-24 (mda-7/IL-24) is a cytokine in the IL-10 family that has received a great deal of attention for its properties as a tumor suppressor and as a potential treatment for cancer. In this study, we have identified and characterized five alternatively spliced isoforms of this gene. Several, but not all of these isoforms induce apoptosis in the osteosarcoma cell line U2OS, while none affect the survival of the non-cancerous NOK cell line. One of these isoforms, lacking three exons and encoding the N-terminal end of the mda-7/IL-24 protein sequence, caused levels of apoptosis that were higher than those caused by the full-length mda-7/IL-24 variant. Additionally, we found that the ratio of isoform expression can be modified by the splice factor SRp55. This regulation suggests that alternative splicing of mda-7/IL-24 is under tight control in the cell, and can be modified under various cellular conditions, such as DNA damage. In addition to providing new insights into the function of an important tumor suppressor gene, these findings may also point toward new avenues for cancer treatment.


Asunto(s)
Apoptosis/fisiología , Supervivencia Celular/fisiología , Interleucinas/fisiología , Empalme del ARN , Secuencia de Aminoácidos , Apoptosis/genética , Secuencia de Bases , Western Blotting , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/genética , Cartilla de ADN , Humanos , Interleucinas/química , Interleucinas/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Homología de Secuencia de Aminoácido
10.
J Virol ; 83(1): 210-27, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18842714

RESUMEN

High-risk strains of human papillomavirus (HPV) such as HPV type 16 (HPV16) and HPV18 are causative agents of most human cervical carcinomas. E6, one of the oncogenes encoded by HPV16, possesses a number of biological and transforming functions. We have previously shown that the binding of E6 to host apoptotic proteins such as tumor necrosis factor (TNF) R1, the adaptor protein FADD, and procaspase 8 results in a significant modification of the normal flow of apoptotic events. For example, E6 can bind to and accelerate the degradation of FADD. In addition, full-length E6 binds to the TNF R1 death domain and can also bind to and accelerate the degradation of procaspase 8. In contrast, the binding of small splice isoforms known as E6* results in the stabilization of procaspase 8. In this report, we propose a model for the ability of HPV16 E6 to both sensitize and protect cells from TNF as well as to protect cells from Fas. We demonstrate that both the level of E6 expression and the ratio between full-length E6 and E6* are important factors in the modification of the host extrinsic apoptotic pathways and show that at high levels of E6 expression, the further sensitization of U2OS, NOK, and Ca Ski cells to TNF-mediated apoptosis is most likely due to the formation of a pseudo-death-inducing signaling complex structure that includes complexes of E6 proteins.


Asunto(s)
Apoptosis , Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/metabolismo , Proteínas Represoras/metabolismo , Línea Celular , Humanos , Unión Proteica , Isoformas de Proteínas/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Receptor fas/inmunología
11.
Sci Rep ; 10(1): 19044, 2020 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-33149215

RESUMEN

High-risk human papillomaviruses (HPV) are the causative agents of cervical cancer. However, not all infected women develop cervical cancer. Cervical tumorigenesis is characterized by a multifactorial etiology, with oxidative stress (OS) likely playing a major role. In addition to exogenous sources, metabolic processes also contribute to OS. In principle, variability in levels of cervical OS has the potential to influence the likelihood of conversion to cervical cancer. To ask whether such variability indeed existed, we assessed the levels of ROS and the oxidative DNA damage biomarker 8-oxodG in normal non-cancerous cervical tissues and cells obtained from women with uterovaginal pelvic organ prolapse following vaginal hysterectomy. We demonstrated five and ten-fold variability between tissues isolated from the transformation zone (TZ) and ectocervix (EC) of different women, respectively. Despite the greater variability (likely due to differences in tissue composition), the overall pattern of ROS levels in EC tissues mirrored those obtained in their corresponding TZ tissues. Our results also show that the levels of ROS in TZ tissues were always higher than or equal to those found in the respective EC tissues, providing a possible explanation for TZ tissue being the primary target for HPV infection and cervical carcinogenesis. Interestingly, primary keratinocytes isolated and cultured from these cervical specimens also displayed high variability in ROS levels, with some strongly mirroring the levels of ROS observed in their corresponding tissues, while others were less closely associated. Finally, we demonstrated that the levels of DNA damage mirrored the levels of ROS in the cultured primary cells. Understanding the factors and mechanisms that dispose certain individuals to develop cervical cancer has the potential to enable the development of approaches that make the conversion of HPV infection to cancer development even more rare.


Asunto(s)
Cuello del Útero/metabolismo , Estrés Oxidativo , 8-Hidroxi-2'-Desoxicoguanosina/metabolismo , Biomarcadores , Transformación Celular Neoplásica/metabolismo , Células Cultivadas , Cuello del Útero/patología , Daño del ADN , Células Epiteliales/metabolismo , Femenino , Humanos , Queratinocitos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Neoplasias del Cuello Uterino/etiología , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
12.
Mol Cancer Res ; 6(8): 1293-307, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18708362

RESUMEN

There is increasing evidence that an augmented state of cellular oxidative stress modulates the expression of stress genes implicated in diseases associated with health disparities such as certain cancers and diabetes. Lens epithelium-derived growth factor p75 (LEDGF/p75), also known as DFS70 autoantigen, is emerging as a survival oncoprotein that promotes resistance to oxidative stress-induced cell death and chemotherapy. We previously showed that LEDGF/p75 is targeted by autoantibodies in prostate cancer patients and is overexpressed in prostate tumors, and that its stress survival activity is abrogated during apoptosis. LEDGF/p75 has a COOH-terminally truncated splice variant, p52, whose role in stress survival and apoptosis has not been thoroughly investigated. We observed unbalanced expression of these proteins in a panel of tumor cell lines, with LEDGF/p75 generally expressed at higher levels. During apoptosis, caspase-3 cleaved p52 to generate a p38 fragment that lacked the NH(2)-terminal PWWP domain and failed to transactivate the Hsp27 promoter in reporter assays. However, p38 retained chromatin association properties and repressed the transactivation potential of LEDGF/p75. Overexpression of p52 or its variants with truncated PWWP domains in several tumor cell lines induced apoptosis, an activity that was linked to the presence of an intron-derived COOH-terminal sequence. These results implicate the PWWP domain of p52 in transcription function but not in chromatin association and proapoptotic activities. Consistent with their unbalanced expression in tumor cells, LEDGF/p75 and p52 seem to play antagonistic roles in the cellular stress response and could serve as targets for novel antitumor therapies.


Asunto(s)
Empalme Alternativo/genética , Caspasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas Oncogénicas/genética , Apoptosis , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Cromatina/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/química , Espacio Intracelular/metabolismo , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Fragmentos de Péptidos/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Transcripción Genética , Activación Transcripcional/genética
13.
J Virol ; 82(19): 9600-14, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18632871

RESUMEN

High-risk strains of human papillomavirus, such as types 16 and 18, have been etiologically linked to cervical cancer. Most cervical cancer tissues are positive for both the E6 and E7 oncoproteins, since it is their cooperation that results in successful transformation and immortalization of infected cells. We have reported that E6 binds to tumor necrosis factor receptor 1 and to Fas-associated death domain (FADD) and, in doing so, prevents E6-expressing cells from responding to apoptotic stimuli. The binding site of E6 to FADD localizes to the first 23 amino acids of FADD and has now been further characterized by the use of deletion and site-directed mutants of FADD in pull-down and functional assays. The results from these experiments revealed that mutations of serine 16, serine 18, and leucine 20 obstruct FADD binding to E6, suggesting that these residues are part of the E6 binding domain on FADD. Because FADD does not contain the two previously identified E6 binding motifs, the LxxphiLsh motif, and the PDZ motif, a novel binding domain for E6 has been identified on FADD. Furthermore, peptides that correspond to this region can block E6/FADD binding in vitro and can resensitize E6-expressing cells to apoptotic stimuli in vivo. These results demonstrate the existence of a novel E6 binding domain.


Asunto(s)
Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Papillomavirus Humano 16/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Apoptosis , Línea Celular Tumoral , Femenino , Humanos , Conformación Molecular , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Receptores Tipo I de Factores de Necrosis Tumoral/química , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/terapia , Neoplasias del Cuello Uterino/virología
14.
Cancer Res ; 67(16): 7621-30, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699766

RESUMEN

Expression of the human papillomavirus 16 E6 oncogene interferes with several vital cellular processes, including the p53-dependent response to DNA damage. To assess the influence of E6 on the early response to DNA damage, we analyzed gene expression following mitomycin C-induced genotoxic stress in human E6-expressing U2OS cells (U2OSE64b) as well as in p53-expressing control cells (U2OSE6AS) by comparative global expression profiling. As expected, genes involved in p53-dependent pathways were activated in p53-expressing cells. In the U2OSE64b cells, however, a largely nonoverlapping group of genes was identified, including two splicing factors of the SR family. Immunoblot analysis revealed increased expression of several SR proteins during the early response to DNA damage, which was accompanied by activation of alternative splicing activity. Disruption of splicing activity by treatment with small interfering RNA directed against splicing factor SRp55 resulted in the increased viability of p53-deficient cells following DNA damage. To determine whether the transient activation of splicing activity was due to E6-mediated degradation of p53, or was due to some other activity of E6, we compared the early response of the p53 wild-type and p53-/- isogenic HCT116 cell lines, and found that the increase in splicing activity was observed only in the absence of p53. Finally, both the U2OSE64b and the p53-/- cells showed altered splicing patterns for the CD44 receptor. Together, these data show that cells lacking p53 can activate alternative splicing following DNA damage.


Asunto(s)
Daño del ADN/genética , Receptores de Hialuranos/genética , Empalme Alternativo/efectos de los fármacos , Empalme Alternativo/genética , Antibióticos Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Regulación hacia Abajo , Perfilación de la Expresión Génica , Silenciador del Gen , Células HCT116 , Humanos , Receptores de Hialuranos/biosíntesis , Mitomicina/farmacología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Oncogénicas Virales/biosíntesis , Proteínas Oncogénicas Virales/genética , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Osteosarcoma/metabolismo , Fosfoproteínas/biosíntesis , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Isoformas de Proteínas , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Factores de Empalme Serina-Arginina , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
15.
Oxid Med Cell Longev ; 2019: 1279250, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30992736

RESUMEN

Reactive oxygen species (ROS) are common by-products of normal aerobic cellular metabolism and play important physiological roles in intracellular cell signaling and homeostasis. The human body is equipped with antioxidant systems to regulate the levels of these free radicals and maintain proper physiological function. However, a condition known as oxidative stress (OS) occurs, when ROS overwhelm the body's ability to readily detoxify them. Excessive amounts of free radicals generated under OS conditions cause oxidative damage to proteins, lipids, and nucleic acids, severely compromising cell health and contributing to disease development, including cancer. Biomarkers of OS can therefore be exploited as important tools in the assessment of disease status in humans. In the present review, we discuss different approaches used for the evaluation of OS in clinical samples. The described methods are limited in their ability to reflect on OS only partially, revealing the need of more integrative approaches examining both pro- and antioxidant reactions with higher sensitivity to physiological/pathological alternations. We also provide an overview of recent findings of OS in patients with different types of cancer. Identification of OS biomarkers in clinical samples of cancer patients and defining their roles in carcinogenesis hold great promise in promoting the development of targeted therapeutic approaches and diagnostic strategies assessing disease status. However, considerable data variability across laboratories makes it difficult to draw general conclusions on the significance of these OS biomarkers. To our knowledge, no adequate comparison has yet been performed between different biomarkers and the methodologies used to measure them, making it difficult to conduct a meta-analysis of findings from different groups. A critical evaluation and adaptation of proposed methodologies available in the literature should therefore be undertaken, to enable the investigators to choose the most suitable procedure for each chosen biomarker.


Asunto(s)
Investigación Biomédica/métodos , Neoplasias/fisiopatología , Estrés Oxidativo/fisiología , Humanos
16.
Gene ; 420(1): 34-41, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18571879

RESUMEN

Alternative splicing is an important source of protein diversity, and is an established but not yet fully understood mechanism for gene regulation in higher eukaryotes. Its regulation is governed by a variety of mechanisms, including variation in the expression levels of splicing factors engaged in spliceosome formation. SRp55 is one of the most ubiquitous splicing factors and one that can be up-regulated by DNA damage in the absence of p53, and we had previously found that depletion of its activity increased resistance to DNA damage in p53-dependant manner. To assess its influence on the splicing patterns of genes involved in apoptosis, we performed splice-specific microarray analysis of cells treated with siRNA specific for this gene. This analysis, backed by RT-PCR verification, identified three genes, KSR1, ZAK and mda7/IL24, which are sensitive to SRp55 depletion. We also analyzed the splice patterns of apoptosis-related genes in p53-deficient U2OS cells following treatment with the genotoxic drug mitomycin C. This analysis revealed that DNA damage resulted in changes in splicing activity that modified the splicing pattern of Fas, a key pro-apoptotic, p53-inducible death receptor. Interestingly, this modification led to an enrichment of the anti-apoptotic soluble Fas isoform, and this secreted isoform was detected in the media surrounding cells subjected to DNA damage. These findings show that modulation of splicing activity in p53-deficient cells during the early response to sub-lethal DNA damage results in a change in the splicing of target genes, thus modifying the cellular response to genotoxic agents.


Asunto(s)
Empalme Alternativo , Daño del ADN , Proteínas Nucleares/genética , Fosfoproteínas/genética , Empalme del ARN , Apoptosis , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Análisis por Micromatrices/métodos , Mitomicina/farmacología , Isoformas de Proteínas , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN , Factores de Empalme Serina-Arginina , Empalmosomas , Receptor fas/genética
17.
Anal Biochem ; 378(1): 80-6, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18423390

RESUMEN

Ceramides play an important role in diverse cellular functions such as differentiation, cell cycle progression, cell-cell adhesion, senescence, and apoptosis. Here we report a method of extracting lipids from mammalian cells and quantifying ceramide, where the assay conditions were optimized for reproducibility, linearity, recovery, and sensitivity. Simultaneous chromatographic separations were carried out by reversed-phase high-performance liquid chromatography coupled to electrospray ionization using a Pursuit 3 Diphenyl column (50 x 2.0 mm) and supported by a mobile phase consisting of acetonitrile plus 0.1% formic acid and 25 mM ammonium acetate. Ceramides were detected in the multiple reaction mode by tandem mass spectrometry in the positive ion mode, and all extracted ion peaks were integrated for quantitative analysis. The limits of detection and quantification achieved were 0.2 and 1.0 pg on column, respectively. Using this method, we successfully quantified and compared differences in C(18) ceramide levels induced by two DNA-damaging agents, mitomycin C and daunorubicin, and two apoptosis-inducing ligands, tumor necrosis factor alpha (TNF-alpha) and TNF-related apoptosis-inducing ligand (TRAIL). This work, therefore, describes a method that will be helpful for investigating how ceramide is regulated by different chemotherapeutic agents and will help us to better understand the mechanisms of signal transduction involving ceramide.


Asunto(s)
Ceramidas/análisis , Cromatografía Líquida de Alta Presión/instrumentación , Cromatografía Líquida de Alta Presión/métodos , Espectrometría de Masas en Tándem/instrumentación , Espectrometría de Masas en Tándem/métodos , Línea Celular Tumoral , Ceramidas/química , Humanos , Estructura Molecular
18.
Cancer Genomics Proteomics ; 13(4): 259-73, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27365376

RESUMEN

BACKGROUND: High-risk human papillomaviruses (HPV) cause nearly all cases of cervical cancer, as well as many types of oral and anogenital cancer. Alternative splicing increases the capacity of the HPV genome to encode the proteins necessary for successful completion of its infectious life cycle. However, the roles of these splice variants, including E6*, the smaller splice isoform of the E6 oncogene, in carcinogenesis are not clear. MATERIALS AND METHODS: SiHa (HPV16(+)) and C33A (HPV(-)) cells were transfected with the E6* plasmid, and tandem mass tag-labeled protein levels were quantified by mass spectrometry. Proteomic analyses identified pathways affected by E6* in both HPV(+) and HPV(-) cells, and pathways were validated using in vitro methods. RESULTS: A total of 4,300 proteins were identified and quantified in lysates of SiHa and C33A cells with and without HPV16 E6* expression. SiHa and C33A cells expressing E6* underwent changes in protein expression affecting integrin signaling and mitochondrial dysfunction pathways, respectively. Subsequent experiments were performed to validate selected E6*-mediated alterations in protein levels. CONCLUSION: E6* modifies the expression of proteins involved in mitochondrial dysfunction and oxidative phosphorylation in C33A cells, and ß-integrin signaling in SiHa cells.


Asunto(s)
Expresión Génica , Integrinas/metabolismo , Mitocondrias/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas Represoras/genética , Transducción de Señal , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Línea Celular Tumoral , Daño del ADN , Femenino , Glutatión/metabolismo , Papillomavirus Humano 16/genética , Humanos , Potencial de la Membrana Mitocondrial , Proteínas Oncogénicas Virales/metabolismo , Estrés Oxidativo , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/virología , Proteómica/métodos , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/metabolismo
19.
Am J Cancer Res ; 6(4): 764-80, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27186429

RESUMEN

Cervical cancer is the second most common cancer, and the fourth most common cause of cancer death in women worldwide. Nearly all of these cases are caused by high-risk HPVs (HR HPVs), of which HPV16 is the most prevalent type. In most cervical cancer specimens, HR HPVs are found integrated into the human genome, indicating that integration is a key event in cervical tumor development. An understanding of the mechanisms that promote integration may therefore represent a unique opportunity to intercept carcinogenesis. To begin identifying these mechanisms, we tested the hypothesis that chronic oxidative stress (OS) induced by virus- and environmentallymediated factors can induce DNA damage, and thereby increase the frequency with which HPV integrates into the host genome. We found that virus-mediated factors are likely involved, as expression of E6*, a splice isoform of HPV16 E6, increased the levels of reactive oxygen species (ROS), caused oxidative DNA damage, and increased the frequency of plasmid DNA integration as assessed by colony formation assays. To assess the influence of environmentally induced chronic OS, we used L-Buthionine-sulfoximine (BSO) to lower the level of the intracellular antioxidant glutathione. Similar to our observations with E6*, glutathione depletion by BSO also increased ROS levels, caused oxidative DNA damage and increased the integration frequency of plasmid DNA. Finally, under conditions of chronic OS, we were able to induce and characterize a few independent events in which episomal HPV16 integrated into the host genome of cervical keratinocytes. Our results support a chain of events leading from induction of oxidative stress, to DNA damage, to viral integration, and ultimately to carcinogenesis.

20.
Viruses ; 7(9): 5040-65, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26393639

RESUMEN

The rise in human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC) has elicited significant interest in the role of high-risk HPV in tumorigenesis. Because patients with HPV-positive HNSCC have better prognoses than do their HPV-negative counterparts, current therapeutic strategies for HPV⁺ HNSCC are increasingly considered to be overly aggressive, highlighting a need for customized treatment guidelines for this cohort. Additional issues include the unmet need for a reliable screening strategy for HNSCC, as well as the ongoing assessment of the efficacy of prophylactic vaccines for the prevention of HPV infections in the head and neck regions. This review also outlines a number of emerging prospects for therapeutic vaccines, as well as for targeted, molecular-based therapies for HPV-associated head and neck cancers. Overall, the future for developing novel and effective therapeutic agents for HPV-associated head and neck tumors is promising; continued progress is critical in order to meet the challenges posed by the growing epidemic.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/cirugía , Detección Precoz del Cáncer/métodos , Neoplasias de Cabeza y Cuello/diagnóstico , Neoplasias de Cabeza y Cuello/cirugía , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/diagnóstico , Carcinoma de Células Escamosas/prevención & control , Neoplasias de Cabeza y Cuello/prevención & control , Humanos , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA