Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 21(1): 102, 2023 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-36759828

RESUMEN

BACKGROUND: In the past decades studies on anti-tumoral drugs inhibiting matrix metalloproteinase (MMPs) were disappointing. Recently, we demonstrated that mature endothelial cells (ECs) and endothelial colony forming cells (ECFCs) can switch between invasion modes to cope with challenging environments, performing the "amoeboid angiogenesis" in the absence of proteases activity. METHODS: We first set out to investigate by ELISA if the inhibitors of the main protease family involved in angiogenesis were differently expressed during breast cancer progression. We used Marimastat, a broad-spectrum MMP inhibitor, as a means of inducing amoeboid characteristics and studied VEGF role in amoeboid angiogenesis. Thus, we performed invasion and capillary morphogenesis assay, morphological, cell signaling and in vivo mouse studies. RESULTS: Our data showed that TIMP1, TIMP2, alpha2-antiplasmin, PAI-1 and cystatin increase in breast cancer serum of patients with primary cancer and lymph node positive compared to healthy women. In vitro results revealed that the most high-powered protease inhibitors able to induce amoeboid invasion of ECFCs were TIMP1, 2 and 3. Surprisingly, Marimastat promotes ECFC invasion and tubular formation in vitro and in vivo, inducing amoeboid characteristics. We observed that the combination of Marimastat plus VEGF doesn't boost neither cell invasion nor vessel formation capacity. Moreover, inhibition of VEGF activity with Bevacizumab in the presence of Marimastat confirmed that amoeboid angiogenesis is independent from the stimulus of the main vascular growth factor, VEGF. CONCLUSIONS: We underline the importance to consider the amoeboid mechanism of endothelial and cancer cell invasion, probably responsible for the failure of synthetic metalloproteinase inhibitors as cancer therapy and tumor resistance to VEGF-targeted therapies, to set-up new drugs to be used in cancer therapy.


Asunto(s)
Amoeba , Neoplasias , Animales , Femenino , Ratones , Amoeba/metabolismo , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Células Endoteliales/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Morfogénesis , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Sistema de Señalización de MAP Quinasas
2.
J Cell Mol Med ; 26(8): 2337-2350, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35278036

RESUMEN

Senescence occurs upon critical telomere shortening, or following DNA damage, oncogenic activation, hypoxia and oxidative stress, overall referred to stress-induced premature senescence (SIPS). In response to DNA damage, senescent cells release cytoplasmic chromatin fragments (CCFs), and express an altered secretome, the senescence-associated secretory phenotype (SASP), which contributes to generate a pro-inflammatory and pro-tumoral extracellular milieu. Polyphenols have gained significant attention owing to their anti-inflammatory and anti-tumour activities. Here, we studied the effect of oleuropein aglycone (OLE) and hydroxytyrosol (HT) on DNA damage, CCF appearance and SASP in a model of irradiation-induced senescence. Neonatal human dermal fibroblasts (NHDFs) were γ-irradiated and incubated with OLE, 5 µM and HT, 1 µM. Cell growth and senescence-associated (SA)-ß-Gal-staining were used as senescence markers. DNA damage was evaluated by Comet assay, lamin B1 expression, release of CCFs, cyclic GMP-AMP Synthase (cGAS) activation. IL-6, IL-8, MCP-1 and RANTES were measured by ELISA assay. Our results showed that OLE and HT exerted a protective effect on 8 Gy irradiation-induced senescence, preserving lamin B1 expression and reducing cGAS/STING/NFκB-mediated SASP. The ability of OLE and HT to mitigate DNA damage, senescence status and the related SASP in normal cells can be exploited to improve the efficacy and safety of cancer radiotherapy.


Asunto(s)
Neoplasias , Olea , Senescencia Celular , Daño del ADN , Humanos , Lamina Tipo B , FN-kappa B/genética , Neoplasias/metabolismo , Nucleotidiltransferasas/genética , Olea/metabolismo , Fenoles/farmacología , Radiación Ionizante
3.
Int J Cancer ; 150(2): 362-373, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34486752

RESUMEN

The role of fibroblast APC mutation in carcinogenesis is not clear. Apc+/- colon fibroblasts have been previously characterized: however, little is known about their behavior at very early-stage of colon carcinogenesis. We cultured colon mucosa fibroblasts (PCF, Apc+/- ) of Pirc rats (F344/NTac-Apcam1137 ) at an early stage of tumorigenesis, in absence of preneoplastic lesions, and of age-matched wt (WCF): DNA damage levels, inflammatory phenotype and the expression of known markers of CAFs were analyzed. The latter were also assessed by microarray analysis on colon normal mucosa of Pirc and wt animals. PCF exhibited higher proliferative rates (P < .001) and delayed replicative senescence onset (P < .05) compared to WCF, along with a lower level of oxidative DNA damage (P < .05). Furthermore, a constitutively higher expression of COX-2 and sensitivity to inflammatory stimuli was found in PCF compared to WCF (P < .05), accompanied by higher invasive capability (P < .05) and presence of cytoplasmic chromatin foci (cytoplasmic chromatin foci, P < .05). However, they neither expressed CAFs markers (α-SMA, IL-6) nor responded to CAFs activating stimuli (TGF-ß). Accordingly, CAFs markers and activating stimuli resulted down-regulated in Pirc normal mucosa compared to wt, whereas DNA damage response and tolerance pathways were overexpressed. These data show for the first time that a proliferative and inflammatory phenotype characterizes Apc+/- colon fibroblasts since very early stages of colon tumorigenesis, and indicate a role of Apc mutation in driving fibroblast phenotypic alterations that could support the establishment of a protumorigenic environment. Early pharmacological targeting of these dysfunctions might impact on tumor prevention in FAP patients.


Asunto(s)
Proliferación Celular , Colon/patología , Neoplasias del Colon/patología , Daño del ADN , Fibroblastos/patología , Genes APC , Inflamación/patología , Animales , Apoptosis , Colon/metabolismo , Neoplasias del Colon/etiología , Neoplasias del Colon/metabolismo , Fibroblastos/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Mutación , Fenotipo , Ratas , Ratas Endogámicas F344
4.
Eur J Immunol ; 51(1): 220-230, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32691428

RESUMEN

How T-helper (Th) lymphocyte subpopulations identified in synovial fluid from patients with juvenile idiopathic arthritis (JIA) (Th17, classic Th1, or nonclassic Th1) drive joint damage is of great interest for the possible use of biological drugs that inhibit the specific cytokines. Our objective was to clarify the role of such Th subpopulations in the pathogenesis of articular cartilage destruction by synovial fibroblasts (SFbs), and the effect of Th17 blockage in an animal model. SFbs were isolated from healthy subjects and patients with JIA, and peripheral blood Th lymphocytes subsets were obtained from healthy subjects. Fragments of human cartilage from healthy subjects in a collagen matrix containing JIA or normal SFbs grafted underskin in SCID mice were used to measure cartilage degradation under the effects of Th supernatants. JIA SFbs overexpress MMP9 and MMP2 and Th17 induce both MMPs in normal SFbs, while nonclassic Th1 upregulate urokinase plasminogen activator (uPA) activity. In vitro invasive phenotype of normal SFbs is stimulated with conditioned medium of Th17 and nonclassic-Th1. In the in vivo "inverse wrap" model, normal SFbs stimulated with supernatants of Th17-lymphocytes and nonclassic Th1 produced a cartilage invasion and degradation similar to JIA SFbs. Secukinumab inhibits the cartilage damage triggered by factors produced by Th17.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Artritis Juvenil/inmunología , Artritis Juvenil/terapia , Cartílago Articular/inmunología , Cartílago Articular/patología , Células Th17/inmunología , Células Th17/patología , Adolescente , Animales , Artritis Experimental/inmunología , Artritis Experimental/patología , Artritis Experimental/terapia , Artritis Juvenil/patología , Cartílago Articular/metabolismo , Estudios de Casos y Controles , Niño , Preescolar , Citocinas/inmunología , Modelos Animales de Enfermedad , Fibroblastos/inmunología , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Técnicas In Vitro , Interleucina-17/antagonistas & inhibidores , Ratones , Ratones SCID , Proteolisis , Membrana Sinovial/inmunología , Membrana Sinovial/metabolismo , Membrana Sinovial/patología
5.
Rheumatology (Oxford) ; 61(9): 3864-3874, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34888638

RESUMEN

OBJECTIVE: Emerging evidence demonstrates that excessive accumulation of senescent cells is associated with some chronic diseases and suggests a pathogenic role of cellular senescence in fibrotic processes, such as that occurring in ageing or in SSc. Recently we demonstrated that parvovirus B19 (B19V) activates normal human dermal fibroblasts and induces expression of different profibrotic/pro-inflammatory genes. This observation prompted us to investigate whether it is also able to induce fibroblast senescence as a potential pathogenetic mechanism in B19V-induced fibrosis. METHODS: Primary cultures of fibroblasts were infected with B19V and analysed for the acquisition of senescence markers, such as morphological modifications, senescence-associated ß-galactosidase (SA-ß-gal) activity, DNA damage response and expression of senescence-associated secretory phenotype (SASP)-related factors. RESULTS: We demonstrated that B19V-infected fibroblasts develop typical senescence features such as enlarged and flat-shaped morphology and SA-ß-gal activity similar to that observed in SSc skin fibroblasts. They also developed an SASP-like phenotype characterized by mRNA expression and release of some pro-inflammatory cytokines, along with activation of the transcription factor nuclear factor κB. Moreover, we observed B19V-induced DNA damage with the comet assay: a subpopulation of fibroblasts from B19V-infected cultures showed a significantly higher level of DNA strand breaks and oxidative damage compared with mock-infected cells. An increased level and nuclear localization of γH2AX, a hallmark of DNA damage response, were also found. CONCLUSIONS: B19V-induced senescence and production of SASP-like factors in normal dermal fibroblasts could represent a new pathogenic mechanism of non-productive B19V infection, which may have a role in the fibrotic process.


Asunto(s)
Parvovirus B19 Humano , Esclerodermia Sistémica , Senescencia Celular , Fibroblastos/metabolismo , Fibrosis , Humanos , Parvovirus B19 Humano/genética , Esclerodermia Sistémica/patología
6.
Cell Mol Life Sci ; 78(6): 3057-3072, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33237352

RESUMEN

Exosomes (Exos) have been reported to promote pre-metastatic niche formation, proliferation, angiogenesis and metastasis. We have investigated the role of uPAR in melanoma cell lines-derived Exos and their pro-angiogenic effects on human microvascular endothelial cells (HMVECs) and endothelial colony-forming cells (ECFCs). Melanoma Exos were isolated from conditioned media of A375 and M6 cells by differential centrifugation and filtration. Tunable Resistive Pulse Sensing (TRPS) and Nanoparticle tracking analysis were performed to analyze dimension and concentration of Exos. The CRISPR-Cas 9 technology was exploited to obtain a robust uPAR knockout. uPAR is expressed in melanoma Exos that are internalized by HMVECs and ECFCs, enhancing VE-Cadherin, EGFR and uPAR expression in endothelial cells that undergo a complete angiogenic program, including proliferation, migration and tube formation. uPAR loss reduced the pro-angiogenic effects of melanoma Exos in vitro and in vivo by inhibition of VE-Cadherin, EGFR and uPAR expression and of ERK1,2 signaling in endothelial cells. A similar effect was obtained with a peptide that inhibits uPAR-EGFR interaction and with the EGFR inhibitor Gefitinib, which also inhibited melanoma Exos-dependent EGFR phosphorylation. This study suggests that uPAR is required for the pro-angiogenic activity of melanoma Exos. We propose the identification of uPAR-expressing Exos as a potentially useful biomarker for assessing pro-angiogenic propensity and eventually monitoring the response to treatment in metastatic melanoma patients.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Exosomas/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal , Animales , Antígenos CD/genética , Cadherinas/genética , Línea Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gefitinib/farmacología , Edición Génica , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neovascularización Fisiológica , Fosforilación/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño , Receptores del Activador de Plasminógeno Tipo Uroquinasa/antagonistas & inhibidores , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética
7.
Rheumatology (Oxford) ; 60(10): 4508-4519, 2021 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33471123

RESUMEN

OBJECTIVES: SSc is an autoimmune disease characterized by peripheral vasculopathy and skin and internal organ fibrosis. Accumulating evidence underlines a close association between a metabolic reprogramming of activated fibroblasts and fibrosis. This prompted us to determine the metabolism of SSc dermal fibroblasts and the effect on the vasculopathy characterizing the disease. METHODS: A Seahorse XF96 Extracellular Flux Analyzer was used to evaluate SSc fibroblast metabolism. In vitro invasion and capillary morphogenesis assays were used to determine the angiogenic ability of endothelial cells (ECs). Immunofluorescence, flow cytometry and real-time PCR techniques provided evidence of the molecular mechanism behind the impaired vascularization that characterizes SSc patients. RESULTS: SSc fibroblasts, compared with controls, showed a boosted glycolytic metabolism with increased lactic acid release and subsequent extracellular acidification that in turn was found to impair EC invasion and organization in capillary-like networks without altering cell viability. A molecular link between extracellular acidosis and endothelial dysfunction was identified as acidic ECs upregulated MMP-12, which cleaves and inactivates urokinase-type plasminogen activator receptor, impairing angiogenesis in SSc. Moreover, the acidic environment was found to induce the loss of endothelial markers and the acquisition of mesenchymal-like features in ECs, thus promoting the endothelial-to-mesenchymal transition process that contributes to both capillary rarefaction and tissue fibrosis in SSc. CONCLUSION: This study showed the relationship of the metabolic reprogramming of SSc dermal fibroblasts, extracellular acidosis and endothelial dysfunction that may contribute to the impairment and loss of peripheral capillary networks in SSc disease.


Asunto(s)
Acidosis/fisiopatología , Microambiente Celular/fisiología , Endotelio Vascular/fisiopatología , Esclerodermia Sistémica/fisiopatología , Enfermedades Vasculares/fisiopatología , Acidosis/etiología , Adulto , Anciano , Western Blotting , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Fibroblastos/metabolismo , Glucólisis/fisiología , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica , Esclerodermia Sistémica/complicaciones , Piel/citología , Enfermedades Vasculares/etiología
8.
Langmuir ; 37(11): 3248-3260, 2021 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-33683133

RESUMEN

An alternative approach to classical surface plasmon resonance spectroscopy is dielectric-loaded waveguide (DLWG) spectroscopy, widely used in the past decades to investigate bio-interaction kinetics. Despite their wide application, a successful and clear approach to use the DLWGs for the one-step simultaneous determination of both the thickness and refractive index of organic thin films is absent in the literature. We propose here, for the first time, an experimental protocol based on the multimodal nature of DLWGs to be followed in order to evaluate the optical constants and thickness of transparent thin films with a unique measurement. The proposed method is general and can be applied to every class of transparent organic materials, with a resolution and accuracy which depend on the nature of the external medium (gaseous or liquid), the geometrical characteristics of the DLWG, and the values of both the thickness and dielectric constant of the thin film. From the experimental point of view, the method is demonstrated in a nitrogen environment with an accuracy of about 3%, for the special case of electroluminescent thin films of Eu3+ß-diketonate complexes, with an average thickness of about 20 nm. The high value of the refractive index measured for the thin film with the Eu(btfa)3(t-bpete) complex was confirmed by the use of a spectroscopic model based on the Judd-Ofelt theory, in which the magnetic dipole transition 5D0 → 7F1 (Eu3+) for similar films containing Eu3+ complexes is taken as a reference. The DLWGs are finally applied to control the refractive index changes of the organic thin films under UVA irradiation, with potential applications in dosimetry and monitoring light-induced transformation in organic thin films.

9.
Rheumatology (Oxford) ; 59(11): 3526-3532, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32556240

RESUMEN

OBJECTIVE: Fibrosis is the most characteristic pathological hallmark of SSc, a connective tissue disease characterized by vascular and immunological abnormalities, inflammation and enhanced extracellular matrix production, leading to progressive fibrosis of skin and internal organs. We previously demonstrated that parvovirus B19 (B19V) can infect normal human dermal fibroblasts (NHDFs) and that B19V persists in SSc fibroblasts. In this study, we investigated whether parvovirus B19V is able to activate in vitro NHDFs and to induce in these cells some phenotypic features similar to that observed in the SSc fibroblasts. METHODS: We preliminarily analysed the time course of B19V infection in cultured NHDFs, then we investigated the ability of B19V to induce cell migration, invasive phenotype and mRNA expression of some profibrotic and/or proinflammatory genes. RESULTS: We confirmed our previous findings that B19V infects NHDFs, but the infection is not productive. After incubation with B19V, NHDFs showed a significant increase of both migration and invasiveness, along with mRNA expression of different profibrotic genes (α-SMA, EDN-1, IL-6, TGF-ß1 receptors 1 and 2, Col1α2), some genes associated with inflammasome platform (AIM2, IFI16, IL-1ß, CASP-1) and genes for metalloprotease (MMP 2, 9 and 12). CONCLUSION: These data suggest that B19V can activate dermal fibroblasts and may have a role in the pathogenesis of fibrosis. B19V-induced fibroblast migration and invasiveness could be due to the B19V-associated MMP9 overexpression and activation. Moreover, the up-regulation of MMP12, typical of SSc, could link the B19V infection of fibroblasts to the anti-angiogenic process.


Asunto(s)
Movimiento Celular , Fibroblastos/metabolismo , Fibrosis/genética , Inflamación/genética , Infecciones por Parvoviridae/genética , ARN Mensajero/metabolismo , Actinas/genética , Caspasa 1/genética , Células Cultivadas , Colágeno Tipo I/genética , Proteínas de Unión al ADN/genética , Endotelina-1/genética , Fibroblastos/patología , Fibroblastos/virología , Fibrosis/patología , Humanos , Técnicas In Vitro , Interleucina-1beta/genética , Interleucina-6/genética , Metaloproteinasa 12 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Proteínas Nucleares/genética , Infecciones por Parvoviridae/patología , Parvovirus B19 Humano , Fosfoproteínas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Esclerodermia Sistémica/virología , Piel/citología , Piel/patología , Transcriptoma
10.
FASEB J ; 33(12): 13893-13904, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31618597

RESUMEN

Angiogenesis depends on a delicate balance between the different transcription factors, and their control should be considered necessary for preventing or treating diseases. Pre-B-cell leukemia transcription factor regulating protein 1 (Prep1) is a homeodomain transcription factor that plays a primary role in organogenesis and metabolism. Observations performed in a Prep1 hypomorphic mouse model, expressing 3-5% of the protein, show an increase of embryonic lethality due, in part, to defects in angiogenesis. In this study, we provide evidence that overexpression of Prep1 in mouse aortic endothelial cells (MAECs) stimulates migration, proliferation, and tube formation. These effects are paralleled by an increase of several proangiogenic factors and by a decrease of the antiangiogenic gene neurogenic locus notch homolog protein 1 (Notch1). Prep1-mediated angiogenesis involves the activation of the p160 Myb-binding protein (p160)/peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) pathway. Indeed, Prep1 overexpression increases its binding with p160 and induces a 4-fold increase of p160 and 70% reduction of PGC-1α compared with control cells. Incubation of MAECs with a synthetic Prep1(54-72) peptide, mimicking the Prep1 region involved in the interaction with p160, reverts the proangiogenic effects mediated by Prep1. In addition, Prep1 levels increase by 3.2-fold during the fibroblast growth factor ß (bFGF)-mediated endothelial colony-forming cells' activation, whereas Prep1(54-72) peptide reduces the capability of these cells to generate tubular-like structures in response to bFGF, suggesting a possible role of Prep1 both in angiogenesis from preexisting vessels and in postnatal vasculogenesis. Finally, Prep1 hypomorphic heterozygous mice, expressing low levels of Prep1, show attenuated placental angiogenesis and vessel formation within Matrigel plugs. All of these observations indicate that Prep1, complexing with p160, decreases PGC-1α and stimulates angiogenesis.-Cimmino, I., Margheri, F., Prisco, F., Perruolo, G., D'Esposito, V., Laurenzana, A., Fibbi, G., Paciello, O., Doti, N., Ruvo, M., Miele, C., Beguinot, F., Formisano, P., Oriente, F. Prep1 regulates angiogenesis through a PGC-1α-mediated mechanism.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Neovascularización Patológica/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Ratones
11.
Cell Mol Life Sci ; 74(15): 2761-2771, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28331999

RESUMEN

Although surgical excision, chemo-, and radio-therapy are clearly advanced, tumors may relapse due to cells of the so-called "minimal residual disease". Indeed, small clusters of tumor cells persist in host tissues after treatment of the primary tumor elaborating strategies to survive and escape from immunological attacks before their relapse: this variable period of remission is known as "cancer dormancy". Therefore, it is crucial to understand and consider the major concepts addressing dormancy, to identify new targets and disclose potential clinical strategies. Here, we have particularly focused the relationships between tumor microenvironment and cancer dormancy, looking at a re-appreciated aspect of this compartment that is the low extracellular pH. Accumulating evidences indicate that acidity of tumor microenvironment is associated with a poor prognosis of tumor-bearing patients, stimulates a chemo- and radio-therapy resistant phenotype, and suppresses the tumoricidal activity of cytotoxic lymphocytes and natural killer cells, and all these aspects are useful for dormancy. Therefore, this review discusses the possibility that acidity of tumor microenvironment may provide a new, not previously suggested, adequate milieu for "dormancy" of tumor cells.


Asunto(s)
Acidosis/complicaciones , Recurrencia Local de Neoplasia/etiología , Microambiente Tumoral , Acidosis/inmunología , Acidosis/patología , Animales , Apoptosis , Proliferación Celular , Humanos , Concentración de Iones de Hidrógeno , Vigilancia Inmunológica , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/patología , Neoplasia Residual/complicaciones , Neoplasia Residual/inmunología , Neoplasia Residual/patología , Neoplasias/inmunología , Neoplasias/patología , Neoplasias/terapia , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/patología , Neovascularización Patológica/etiología , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Pronóstico , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología
12.
Int J Cancer ; 141(6): 1190-1200, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28577299

RESUMEN

In this manuscript, we show the involvement of the uPA/uPAR system in the regulation of aerobic glycolysis of melanoma cells. uPAR over-expression in human melanoma cells controls an invasive and glycolytic phenotype in normoxic conditions. uPAR down-regulation by siRNA or its uncoupling from integrins, and hence from integrin-linked tyrosine kinase receptors (IL-TKRs), by an antagonist peptide induced a striking inhibition of the PI3K/AKT/mTOR/HIF1α pathway, resulting into impairment of glucose uptake, decrease of several glycolytic enzymes and of PKM2, a checkpoint that controls metabolism of cancer cells. Further, binding of uPA to uPAR regulates expression of molecules that govern cell invasion, including extracellular matrix metallo-proteinases inducer (EMPPRIN) and enolase, a glycolytyc enzyme that also serves as a plasminogen receptor, thus providing a common denominator between tumor metabolism and phenotypic invasive features. Such effects depend on the α5ß1-integrin-mediated uPAR connection with EGFR in melanoma cells with engagement of the PI3K-mTOR-HIFα pathway. HIF-1α trans-activates genes whose products mediate tumor invasion and glycolysis, thus providing the common denominator between melanoma metabolism and its invasive features. These findings unveil a unrecognized interaction between the invasion-related uPAR and IL-TKRs in the control of glycolysis and disclose a new pharmacological target (i.e., uPAR/IL-TKRs axis) for the therapy of melanoma.


Asunto(s)
Melanoma/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Glucólisis , Células HEK293 , Xenoinjertos , Humanos , Melanoma/patología , Ratones , Ratones Desnudos , Ratones SCID , Invasividad Neoplásica , Fenotipo
14.
Stem Cells ; 34(10): 2536-2547, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27300750

RESUMEN

Tumor stromal cells can supply appropriate signals that may develop aggressive phenotypes of carcinoma cells and establish a complex scenario which culminates in metastasis. Recent works proposed that bone marrow-derived mesenchymal stem cells (MSC) are recruited to primary tumors. However, the exact functions of these cells in the tumor microenvironment are not well characterized, as it is reported that MSC can either promote or inhibit tumor progression. In the present study, we aim at investigating the signaling molecules which regulate the interplay between MSC, prostate carcinoma (PCa) cells and two important cellular types constituting the tumor-associated stroma, macrophages and fibroblasts, during their progression toward malignancy. We identified TGF-ß1 as a crucial molecule able to attract MSC recruitment both to PCa cells as well as to tumor stroma components. Moreover, PCa- and tumor stroma-secreted TGF-ß1 is important to induce MSC transdifferentiation into carcinoma-associated fibroblast (CAF)-like cells. Consequently, the CAF-like phenotype acquired by MSC is central to promote tumor progression related effects. Thus, tumor-educated MSC enhance PCa invasiveness compared to nonactivated MSC. Additionally, differing from normal MSC, CAF-like MSC perform vascular mimicry and recruit monocytes, which can be further polarized to M2 macrophages within the PCa environment. Our findings indicate a prominent role for TGF-ß1 in MSC mobilization and activation strengthened by the fact that the blockade of TGF-ß1 signaling impairs MSC promotion of PCa progression. Stem Cells 2016;34:2536-2547.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Células Madre Mesenquimatosas/citología , Neoplasias de la Próstata/patología , Factor de Crecimiento Transformador beta1/metabolismo , Microambiente Tumoral , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Transdiferenciación Celular , Factores Quimiotácticos/metabolismo , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Células del Estroma/metabolismo
15.
Int J Mol Sci ; 18(11)2017 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-29084133

RESUMEN

Senescent cells display an increase in the secretion of growth factors, inflammatory cytokines and proteolytic enzymes, termed the "senescence-associated-secretory-phenotype" (SASP), playing a major role in many age-related diseases. The phenolic compounds present in extra-virgin olive oil are inhibitors of oxidative damage and have been reported to play a protective role in inflammation-related diseases. Particularly, hydroxytyrosol and oleuropein are the most abundant and more extensively studied. Pre-senescent human lung (MRC5) and neonatal human dermal (NHDF) fibroblasts were used as cellular model to evaluate the effect of chronic (4-6 weeks) treatment with 1 µM hydroxytyrosol (HT) or 10 µM oleuropein aglycone (OLE) on senescence/inflammation markers. Both phenols were effective in reducing ß-galactosidase-positive cell number and p16 protein expression. In addition, senescence/inflammation markers such as IL-6 and metalloprotease secretion, and Ciclooxigenase type 2 (COX-2) and α-smooth-actin levels were reduced by phenol treatments. In NHDF, COX-2 expression, Nuclear Factor κ-light-chain-enhancer of activated B cells (NFκB) protein level and nuclear localization were augmented with culture senescence and decreased by OLE and HT treatment. Furthermore, the inflammatory effect of Tumor Necrosis Factor α (TNFα) exposure was almost completely abolished in OLE- and HT-pre-treated NHDF. Thus, the modulation of the senescence-associated inflammatory phenotype might be an important mechanism underlying the beneficial effects of olive oil phenols.


Asunto(s)
Antiinflamatorios/farmacología , Senescencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Olea/química , Fenoles/farmacología , Extractos Vegetales/farmacología , Biomarcadores , Línea Celular , Células Cultivadas , Humanos , Glucósidos Iridoides , Iridoides/metabolismo , Fenotipo
16.
Cell Mol Life Sci ; 72(8): 1537-57, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25552244

RESUMEN

Research on the nanoscale membrane structures known as lipid rafts is relevant to the fields of cancer biology, inflammation and ischaemia. Lipid rafts recruit molecules critical to signalling and regulation of the invasion process in malignant cells, the leukocytes that provide immunity in inflammation and the endothelial cells that build blood and lymphatic vessels, as well as the patterning of neural networks. As angiogenesis is a common denominator, regulation of receptors and signalling molecules critical to angiogenesis is central to the design of new approaches aimed at reducing, promoting or normalizing the angiogenic process. The goal of this review is to highlight some of the key issues that indicate the involvement of endothelial cell lipid rafts at each step of so-called 'sprouting angiogenesis', from stimulation of the vascular endothelial growth factor to the choice of tip cells, activation of migratory and invasion pathways, recruitment of molecules that guide axons in vascular patterning and maturation of blood vessels. Finally, the review addresses opportunities for future studies to define how these lipid domains (and their constituents) may be manipulated to stimulate the so-called 'normalization' of vascular networks within tumors, and be identified as the main target, enabling the development of more efficient chemotherapeutics and cancer immunotherapies.


Asunto(s)
Vasos Sanguíneos/metabolismo , Microdominios de Membrana/metabolismo , Axones/metabolismo , Caveolinas/metabolismo , Células Endoteliales/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Neovascularización Patológica , Transducción de Señal , Factores de Crecimiento Endotelial Vascular/metabolismo
17.
J Enzyme Inhib Med Chem ; 31(sup4): 60-63, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27539792

RESUMEN

Juvenile idiopathic arthritis (JIA) is the most common form of chronic rheumatic disease affecting children worldwide, with some features similar to adult rheumatoid arthritis (RA). In the present study, we aim at investigating novel markers that will allow in the future for tailored, more personalized treatment strategies. Hence, taking notice of several reports proving the role of local acidosis as a causal link between inflammatory diseases and related pain, and the involvement of several carbonic anhydrases (CA, EC 4.2.1.1) isoforms in articular diseases, we evaluated in JIA patients the expression of these metalloenzymes. We identified that JIA patients show high levels of active CA IX and XII isoforms. Our results represent the first evidence of the identification of these enzymes as potential therapeutic targets and development of novel innovative therapies for arthritis, also considering that the two isoforms are validated antitumor targets.


Asunto(s)
Artritis Juvenil/enzimología , Anhidrasa Carbónica IX/genética , Anhidrasas Carbónicas/genética , Membrana Sinovial/enzimología , Adolescente , Artritis Juvenil/sangre , Artritis Juvenil/metabolismo , Anhidrasa Carbónica IX/metabolismo , Anhidrasas Carbónicas/metabolismo , Niño , Preescolar , Humanos , Estructura Molecular , Membrana Sinovial/metabolismo
18.
J Cell Mol Med ; 19(1): 113-23, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25313007

RESUMEN

Gangliosides and the urokinase plasminogen activator receptor (uPAR) tipically partition in specialized membrane microdomains called lipid-rafts. uPAR becomes functionally important in fostering angiogenesis in endothelial progenitor cells (EPCs) upon recruitment in caveolar-lipid rafts. Moreover, cell membrane enrichment with exogenous GM1 ganglioside is pro-angiogenic and opposite to the activity of GM3 ganglioside. On these basis, we first checked the interaction of uPAR with membrane models enriched with GM1 or GM3, relying on the adoption of solid-supported mobile bilayer lipid membranes with raft-like composition formed onto solid hydrophilic surfaces, and evaluated by surface plasmon resonance (SPR) the extent of uPAR recruitment. We estimated the apparent dissociation constants of uPAR-GM1/GM3 complexes. These preliminary observations, indicating that uPAR binds preferentially to GM1-enriched biomimetic membranes, were validated by identifying a pro-angiogenic activity of GM1-enriched EPCs, based on GM1-dependent uPAR recruitment in caveolar rafts. We have observed that addition of GM1 to EPCs culture medium promotes matrigel invasion and capillary morphogenesis, as opposed to the anti-angiogenesis activity of GM3. Moreover, GM1 also stimulates MAPKinases signalling pathways, typically associated with an angiogenesis program. Caveolar-raft isolation and Western blotting of uPAR showed that GM1 promotes caveolar-raft partitioning of uPAR, as opposed to control and GM3-challenged EPCs. By confocal microscopy, we have shown that in EPCs uPAR is present on the surface in at least three compartments, respectively, associated to GM1, GM3 and caveolar rafts. Following GM1 exogenous addition, the GM3 compartment is depleted of uPAR which is recruited within caveolar rafts thereby triggering angiogenesis.


Asunto(s)
Caveolas/metabolismo , Células Progenitoras Endoteliales/metabolismo , Gangliósido G(M1)/farmacología , Gangliósido G(M3)/farmacología , Microdominios de Membrana/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Caveolas/efectos de los fármacos , Caveolina 1/metabolismo , Ensayo de Unidades Formadoras de Colonias , Células Progenitoras Endoteliales/efectos de los fármacos , Humanos , Recién Nacido , Cinética , Microdominios de Membrana/efectos de los fármacos , Fenotipo , Transducción de Señal
19.
Mol Cell Proteomics ; 12(7): 1926-38, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23572564

RESUMEN

Endothelial cell caveolar-rafts are considered functional platforms that recruit several pro-angiogenic molecules to realize an efficient angiogenic program. Here we studied the differential caveolar-raft protein composition of endothelial colony-forming cells following stimulation with VEGF, which localizes in caveolae on interaction with its type-2 receptor. Endothelial colony-forming cells are a cell population identified in human umbilical blood that show all the properties of an endothelial progenitor cell and a high proliferative rate. Two-dimensional gel electrophoresis analysis was coupled with mass spectrometry to identify candidate proteins. The twenty-eight differentially expressed protein spots were grouped according to their function using Gene Ontology classification. In particular, functional categories relative to cell death inhibition and hydrogen peroxide metabolic processes resulted enriched. In these categories, Peroxiredoxin-2 and 6, that control hydrogen peroxide metabolic processes, are the main enriched molecules together with the anti-apoptotic 78 kDa glucose regulated protein. Some of the proteins we identified had never before identified as caveolar-raft components. Other identified proteins include calpain small subunit-1, known to mediates angiogenic response to VEGF, gelsolin, which regulates stress fiber assembly, and annexin A3, an angiogenic mediator that induces VEGF production. We validated the functional activity of the above proteins, showing that the siRNA silencing of these resulted in the inhibition of capillary morphogenesis. Overall, our data show that VEGF stimulation triggers the caveolar-raft recruitment of proteins that warrant a physiological amount of reactive oxygen species to maintain a proper angiogenic function of endothelial colony-forming cells and preserve the integrity of the actin cytoskeleton.


Asunto(s)
Caveolas/metabolismo , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Sangre Fetal/citología , Humanos , Neovascularización Fisiológica/fisiología , Proteómica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Arthritis Rheum ; 65(1): 258-69, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22972461

RESUMEN

OBJECTIVE: Clinical evidence suggests that the vascular abnormalities of systemic sclerosis (SSc) precede the onset of fibrosis, but molecular cues accounting for a possible vascular connection of SSc fibrosis have been elusive, although they have been searched for intensively. Since we had previously shown that connective tissue growth factor (CCN2), endowed with fibroblast-oriented activities, was overexpressed by endothelial cells (ECs) from SSc patients, we undertook this study to investigate its role and mechanisms in fibroblast activation. METHODS: Normal fibroblasts were challenged with conditioned medium of normal microvascular ECs (MVECs) and MVECs obtained from SSc patients with the diffuse form of the disease. Fibroblast invasion was studied using the Boyden chamber Matrigel assay. Fibroblast activation was evaluated by Western blotting and immunofluorescence of α-smooth muscle actin (α-SMA), vimentin, and type I collagen. Matrix metalloproteinase (MMP) production was evaluated by zymography and reverse transcription-polymerase chain reaction. Signal transduction and activation of the small GTPases RhoA and Rac1 were studied by Western blotting. Inhibition of SSc MVEC conditioned medium-dependent fibroblast activation was obtained by anti-CCN2 antibodies and the transforming growth factor ß (TGFß) antagonist peptide p17. RESULTS: SSc MVEC CCN2 stimulated fibroblast activation and invasion. Such activities depended on CCN2-induced overexpression of TGFß and its type I, II, and III receptors combined with overproduction of MMP-2 and MMP-9 gelatinases. All of these effects were reversed by the TGFß antagonist peptide p17. Motility increase required Rac1 activation and RhoA inhibition and was inhibited by an MMP inhibitor. These features connoted a mesenchymal style of cell invasion. Since fibroblast activation also fostered overexpression of α-SMA, vimentin, and type I collagen, the CCN2-dependent increase in fibroblast activities recapitulated the characteristics of a mesenchymal-to-mesenchymal transition. CONCLUSION: SSc MVECs recruit and activate dermal fibroblasts by induction of a CCN2/TGFß-dependent mesenchymal-to-mesenchymal transition.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Células Endoteliales/metabolismo , Fibroblastos/efectos de los fármacos , Esclerodermia Sistémica/metabolismo , Piel/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Western Blotting , Colágeno , Combinación de Medicamentos , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Técnica del Anticuerpo Fluorescente , Humanos , Laminina , Masculino , Mesodermo/patología , Proteoglicanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Esclerodermia Sistémica/patología , Transducción de Señal , Piel/patología , Factor de Crecimiento Transformador beta/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA