Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
JCI Insight ; 9(1)2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-37971882

RESUMEN

Despite strong indications that interactions between melanoma and lymphatic vessels actively promote melanoma progression, the molecular mechanisms are not yet completely understood. To characterize molecular factors of this crosstalk, we established human primary lymphatic endothelial cell (LEC) cocultures with human melanoma cell lines. Here, we show that coculture with melanoma cells induced transcriptomic changes in LECs and led to multiple changes in their function. WNT5B, a paracrine signaling molecule upregulated in melanoma cells upon LEC interaction, was found to contribute to the functional changes in LECs. Moreover, WNT5B transcription was regulated by Notch3 in melanoma cells following the coculture with LECs, and Notch3 and WNT5B were coexpressed in melanoma patient primary tumor and metastasis samples. Moreover, melanoma cells derived from LEC coculture escaped efficiently from the primary site to the proximal tumor-draining lymph nodes, which was impaired upon WNT5B depletion. This supported the role of WNT5B in promoting the metastatic potential of melanoma cells through its effects on LECs. Finally, DLL4, a Notch ligand expressed in LECs, was identified as an upstream inducer of the Notch3/WNT5B axis in melanoma. This study elucidated WNT5B as a key molecular factor mediating bidirectional crosstalk between melanoma cells and lymphatic endothelium and promoting melanoma metastasis.


Asunto(s)
Vasos Linfáticos , Melanoma , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al Calcio/metabolismo , Células Endoteliales/metabolismo , Metástasis Linfática/patología , Vasos Linfáticos/patología , Melanoma/patología , Transducción de Señal , Proteínas Wnt/metabolismo
2.
Mol Cancer Ther ; 22(11): 1319-1331, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37486980

RESUMEN

Chimeric antigen receptor (CAR) T-cell immunotherapies for solid tumors face critical challenges such as heterogeneous antigen expression. We characterized stage-specific embryonic antigen-4 (SSEA-4) cell-surface glycolipid as a target for CAR T-cell therapy. SSEA-4 is mainly expressed during embryogenesis but is also found in several cancer types making it an attractive tumor-associated antigen. Anti-SSEA-4 CAR-T cells were generated and assessed preclinically in vitro and in vivo for antitumor response and safety. SSEA-4 CAR-T cells effectively eliminated SSEA-4-positive cells in all the tested cancer cell lines, whereas SSEA-4-negative cells lines were not targeted. In vivo efficacy and safety studies using NSG mice and the high-grade serous ovarian cancer cell line OVCAR4 demonstrated a remarkable and specific antitumor response at all the CAR T-cell doses used. At high T-cell doses, CAR T cell-treated mice showed signs of health deterioration after a follow-up period. However, the severity of toxicity was reduced with a delayed onset when lower CAR T-cell doses were used. Our data demonstrate the efficacy of anti-SSEA-4 CAR T-cell therapy; however, safety strategies, such as dose-limiting and/or equipping CAR-T cells with combinatorial antigen recognition should be implemented for its potential clinical translation.


Asunto(s)
Carcinoma , Neoplasias Ováricas , Receptores Quiméricos de Antígenos , Humanos , Femenino , Animales , Ratones , Glicoesfingolípidos/metabolismo , Línea Celular Tumoral , Neoplasias Ováricas/metabolismo , Inmunoterapia Adoptiva , Linfocitos T , Carcinoma Epitelial de Ovario/metabolismo , Carcinoma/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Int J Mol Sci ; 24(7)2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-37047272

RESUMEN

After seeing a dramatic increase in the development and use of immunotherapy and precision medicine over the past few decades, oncological care now embraces the start of the adoptive cell therapy (ACT) era. This impulse towards a new treatment paradigm has been led by chimeric antigen receptor (CAR) T cells, the only type of ACT medicinal product to be commercialized so far. Brought about by an ever-growing understanding of cellular engineering, CAR T cells are T lymphocytes genetically modified with an appropriate DNA construct, which endows them with expression of a CAR, a fusion protein between a ligand-specific recognition domain, often an antibody-like structure, and the activating signaling domain of the T cell receptor. Through this genetic enhancement, CAR T cells are engineered from a cancer patient's own lymphocytes to better target and kill their cancer cells, and the current amassed data on clinical outcomes point to a stream of bright developments in the near future. Herein, from concept design and present-day manufacturing techniques to pressing hurdles and bright discoveries around the corner, we review and thoroughly describe the state of the art in CAR T cell therapy.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias , Humanos , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T , Neoplasias/terapia
4.
PLoS Pathog ; 19(1): e1010753, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36689549

RESUMEN

Kaposi's sarcoma herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS), a hyperplasia consisting of enlarged malformed vasculature and spindle-shaped cells, the main proliferative component of KS. While spindle cells express markers of lymphatic and blood endothelium, the origin of spindle cells is unknown. Endothelial precursor cells have been proposed as the source of spindle cells. We previously identified two types of circulating endothelial colony forming cells (ECFCs), ones that expressed markers of blood endothelium and ones that expressed markers of lymphatic endothelium. Here we examined both blood and lymphatic ECFCs infected with KSHV. Lymphatic ECFCs are significantly more susceptible to KSHV infection than the blood ECFCs and maintain the viral episomes during passage in culture while the blood ECFCs lose the viral episome. Only the KSHV-infected lymphatic ECFCs (K-ECFCLY) grew to small multicellular colonies in soft agar whereas the infected blood ECFCs and all uninfected ECFCs failed to proliferate. The K-ECFCLYs express high levels of SOX18, which supported the maintenance of high copy number of KSHV genomes. When implanted subcutaneously into NSG mice, the K-ECFCLYs persisted in vivo and recapitulated the phenotype of KS tumor cells with high number of viral genome copies and spindling morphology. These spindle cell hallmarks were significantly reduced when mice were treated with SOX18 inhibitor, SM4. These data suggest that KSHV-infected lymphatic ECFCs can be utilized as a KSHV infection model for in vivo translational studies to test novel inhibitors representing potential treatment modalities for KS.


Asunto(s)
Herpesvirus Humano 8 , Sarcoma de Kaposi , Animales , Ratones , Herpesvirus Humano 8/genética , Células Endoteliales , Endotelio Vascular/patología
7.
Nucleic Acids Res ; 49(17): 10018-10033, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34417603

RESUMEN

MicroRNAs (miRNAs) are small regulatory RNAs involved in virtually all biological processes. Although many of them are co-expressed from clusters, little is known regarding the impact of this organization on the regulation of their accumulation. In this study, we set to decipher a regulatory mechanism controlling the expression of the ten clustered pre-miRNAs from Kaposi's sarcoma associated herpesvirus (KSHV). We measured in vitro the efficiency of cleavage of each individual pre-miRNA by the Microprocessor and found that pre-miR-K1 and -K3 were the most efficiently cleaved pre-miRNAs. A mutational analysis showed that, in addition to producing mature miRNAs, they are also important for the optimal expression of the whole set of miRNAs. We showed that this feature depends on the presence of a canonical pre-miRNA at this location since we could functionally replace pre-miR-K1 by a heterologous pre-miRNA. Further in vitro processing analysis suggests that the two stem-loops act in cis and that the cluster is cleaved in a sequential manner. Finally, we exploited this characteristic of the cluster to inhibit the expression of the whole set of miRNAs by targeting the pre-miR-K1 with LNA-based antisense oligonucleotides in cells either expressing a synthetic construct or latently infected with KSHV.


Asunto(s)
Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/genética , MicroARNs/genética , Procesamiento Postranscripcional del ARN/genética , ARN Viral/genética , Línea Celular , Células HEK293 , Humanos , Oligonucleótidos Antisentido/genética , Pliegue del ARN/genética
8.
Sci Rep ; 11(1): 8856, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893375

RESUMEN

Erythropoietin producing hepatocellular (Eph) receptors and their membrane-bound ligands ephrins are variably expressed in epithelial cancers, with context-dependent implications to both tumor-promoting and -suppressive processes in ways that remain incompletely understood. Using ovarian cancer tissue microarrays and longitudinally collected patient cells, we show here that ephrinA5/EFNA5 is specifically overexpressed in the most aggressive high-grade serous carcinoma (HGSC) subtype, and increased in the HGSC cells upon disease progression. Among all the eight ephrin genes, high EFNA5 expression was most strongly associated with poor overall survival in HGSC patients from multiple independent datasets. In contrast, high EFNA3 predicted improved overall and progression-free survival in The Cancer Genome Atlas HGSC dataset, as expected for a canonical inducer of tumor-suppressive Eph receptor tyrosine kinase signaling. While depletion of either EFNA5 or the more extensively studied, canonically acting EFNA1 in HGSC cells increased the oncogenic EphA2-S897 phosphorylation, EFNA5 depletion left unaltered, or even increased the ligand-dependent EphA2-Y588 phosphorylation. Moreover, treatment with recombinant ephrinA5 led to limited EphA2 tyrosine phosphorylation, internalization and degradation compared to ephrinA1. Altogether, our results suggest a unique function for ephrinA5 in Eph-ephrin signaling and highlight the clinical potential of ephrinA5 as a cell surface biomarker in the most aggressive HGSCs.


Asunto(s)
Efrina-A5/metabolismo , Recurrencia Local de Neoplasia , Neoplasias Ováricas/patología , Receptor EphA2/metabolismo , Regulación hacia Arriba , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Ováricas/metabolismo , Transducción de Señal , Análisis de Supervivencia
9.
Methods Mol Biol ; 2265: 141-154, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33704712

RESUMEN

Three-dimensional (3D) cell culture has allowed a deeper understanding of complex pathological and physiological processes, overcoming some of the limitations of 2D cell culture on plastic and avoiding the costs and ethical issues related to experiments involving animals. Here we describe a protocol to embed single melanoma cells alone or together with primary human lymphatic endothelial cells in a 3D cross-linked matrix, to investigate the invasion and molecular crosstalk between these two cell types, respectively. After fixation and staining with antibodies and fluorescent conjugates, phenotypic changes in both cell types can be specifically analyzed by confocal microscopy.


Asunto(s)
Técnicas de Cocultivo/métodos , Células Endoteliales/metabolismo , Melanoma/metabolismo , Esferoides Celulares/metabolismo , Línea Celular Tumoral , Células Endoteliales/citología , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Melanoma/patología , Microscopía Confocal , Invasividad Neoplásica
10.
Nat Commun ; 12(1): 512, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33479235

RESUMEN

To achieve replicative immortality, cancer cells must activate telomere maintenance mechanisms to prevent telomere shortening. ~85% of cancers circumvent telomeric attrition by re-expressing telomerase, while the remaining ~15% of cancers induce alternative lengthening of telomeres (ALT), which relies on break-induced replication (BIR) and telomere recombination. Although ALT tumours were first reported over 20 years ago, the mechanism of ALT induction remains unclear and no study to date has described a cell-based model that permits the induction of ALT. Here, we demonstrate that infection with Kaposi's sarcoma herpesvirus (KSHV) induces sustained acquisition of ALT-like features in previously non-ALT cell lines. KSHV-infected cells acquire hallmarks of ALT activity that are also observed in KSHV-associated tumour biopsies. Down-regulating BIR impairs KSHV latency, suggesting that KSHV co-opts ALT for viral functionality. This study uncovers KSHV infection as a means to study telomere maintenance by ALT and reveals features of ALT in KSHV-associated tumours.


Asunto(s)
Neoplasias/genética , Homeostasis del Telómero/genética , Acortamiento del Telómero/genética , Telómero/genética , Carcinogénesis , Línea Celular , Línea Celular Tumoral , Daño del ADN , Replicación del ADN/genética , Células HeLa , Herpesvirus Humano 8/fisiología , Interacciones Huésped-Patógeno , Humanos , Hibridación Fluorescente in Situ , Neoplasias/patología , Neoplasias/virología , Proteoma/genética , Proteoma/metabolismo , Telomerasa/genética , Telomerasa/metabolismo
11.
Viruses ; 12(9)2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32867368

RESUMEN

CRISPR activation (CRISPRa) has revealed great potential as a tool to modulate the expression of targeted cellular genes. Here, we successfully applied the CRISPRa system to trigger the Kaposi's sarcoma-associated herpesvirus (KSHV) reactivation in latently infected cells by selectively activating ORF50 gene directly from the virus genome. We found that a nuclease-deficient Cas9 (dCas9) fused to a destabilization domain (DD) and 12 copies of the VP16 activation domain (VP192) triggered a more efficient KSHV lytic cycle and virus production when guided to two different sites on the ORF50 promoter, instead of only a single site. To our surprise, the virus reactivation induced by binding of the stable DD-dCas9-VP192 on the ORF50 promoter was even more efficient than reactivation induced by ectopic expression of ORF50. This suggests that recruitment of additional transcriptional activators to the ORF50 promoter, in addition to ORF50 itself, are needed for the efficient virus production. Further, we show that CRISPRa can be applied to selectively express the early lytic gene, ORF57, without disturbing the viral latency. Therefore, CRISPRa-based systems can be utilized to facilitate virus-host interaction studies by controlling the expression of not only cellular but also of specific KSHV genes.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Herpesvirus Humano 8/fisiología , Proteínas Inmediatas-Precoces/genética , Sarcoma de Kaposi/virología , Transactivadores/genética , Proteína 9 Asociada a CRISPR/química , Proteína 9 Asociada a CRISPR/genética , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Regulación Viral de la Expresión Génica , Herpesvirus Humano 8/genética , Humanos , Proteínas Inmediatas-Precoces/metabolismo , Regiones Promotoras Genéticas , Dominios Proteicos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transactivadores/metabolismo , Activación Transcripcional , Activación Viral , Latencia del Virus
12.
mSphere ; 5(3)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32581076

RESUMEN

Infections with the nine human herpesviruses (HHVs) are globally prevalent and characterized by lifelong persistence. Reactivations can potentially manifest as life-threatening conditions for which the demonstration of viral DNA is essential. In the present study, we developed HERQ-9, a pan-HHV quantitative PCR designed in triplex reactions to differentiate and quantify each of the HHV-DNAs: (i) herpes simplex viruses 1 and 2 and varicella-zoster virus; (ii) Epstein-Barr virus, human cytomegalovirus, and Kaposi's sarcoma-associated herpesvirus; and (iii) HHV-6A, -6B, and -7. The method was validated with prequantified reference standards as well as with mucocutaneous swabs and cerebrospinal fluid, plasma, and tonsillar tissue samples. Our findings highlight the value of multiplexing in the diagnosis of many unsuspected, yet clinically relevant, herpesviruses. In addition, we report here frequent HHV-DNA co-occurrences in clinical samples, including some previously unknown. HERQ-9 exhibited high specificity and sensitivity (LOD95s of ∼10 to ∼17 copies/reaction), with a dynamic range of 101 to 106 copies/µl. Moreover, it performed accurately in the coamplification of both high- and low-abundance targets in the same reaction. In conclusion, we demonstrated that HERQ-9 is suitable for the diagnosis of a plethora of herpesvirus-related diseases. Besides its significance to clinical management, the method is valuable for the assessment of hitherto-unexplored synergistic effects of herpesvirus coinfections. Furthermore, its high sensitivity enables studies on the human virome, often dealing with minute quantities of persisting HHVs.IMPORTANCE By adulthood, almost all humans become infected by at least one herpesvirus (HHV). The maladies inflicted by these microbes extend beyond the initial infection, as they remain inside our cells for life and can reactivate, causing severe diseases. The diagnosis of active infection by these ubiquitous pathogens includes the detection of DNA with sensitive and specific assays. We developed the first quantitative PCR assay (HERQ-9) designed to identify and quantify each of the nine human herpesviruses. The simultaneous detection of HHVs in the same sample is important since they may act together to induce life-threatening conditions. Moreover, the high sensitivity of our method is of extreme value for assessment of the effects of these viruses persisting in our body and their long-term consequences on our health.


Asunto(s)
Infecciones por Herpesviridae/diagnóstico , Herpesviridae/clasificación , Reacción en Cadena de la Polimerasa Multiplex/métodos , Adolescente , Adulto , Anciano , Línea Celular , Niño , Preescolar , Simulación por Computador , Cartilla de ADN/genética , Sondas de ADN/genética , ADN Viral/genética , Infecciones por Herpesviridae/virología , Humanos , Persona de Mediana Edad , Tonsila Palatina/virología , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Adulto Joven
13.
Cancer Res ; 80(15): 3116-3129, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32518203

RESUMEN

Kaposi sarcoma is a tumor caused by Kaposi sarcoma herpesvirus (KSHV) infection and is thought to originate from lymphatic endothelial cells (LEC). While KSHV establishes latency in virtually all susceptible cell types, LECs support spontaneous expression of oncogenic lytic genes, high viral genome copies, and release of infectious virus. It remains unknown the contribution of spontaneous virus production to the expansion of KSHV-infected tumor cells and the cellular factors that render the lymphatic environment unique to KSHV life cycle. We show here that expansion of the infected cell population, observed in LECs, but not in blood endothelial cells, is dependent on the spontaneous virus production from infected LECs. The drivers of lymphatic endothelium development, SOX18 and PROX1, regulated different steps of the KSHV life cycle. SOX18 enhanced the number of intracellular viral genome copies and bound to the viral origins of replication. Genetic depletion or chemical inhibition of SOX18 caused a decrease of KSHV genome copy numbers. PROX1 interacted with ORF50, the viral initiator of lytic replication, and bound to the KSHV genome in the promoter region of ORF50, increasing its transactivation activity and KSHV spontaneous lytic gene expression and infectious virus release. In Kaposi sarcoma tumors, SOX18 and PROX1 expression correlated with latent and lytic KSHV protein expression. These results demonstrate the importance of two key transcriptional drivers of LEC fate in the regulation of the tumorigenic KSHV life cycle. Moreover, they introduce molecular targeting of SOX18 as a potential novel therapeutic avenue in Kaposi sarcoma. SIGNIFICANCE: SOX18 and PROX1, central regulators of lymphatic development, are key factors for KSHV genome maintenance and lytic cycle in lymphatic endothelial cells, supporting Kaposi sarcoma tumorigenesis and representing attractive therapeutic targets.


Asunto(s)
Transformación Celular Viral/genética , Herpesvirus Humano 8/fisiología , Proteínas de Homeodominio/fisiología , Factores de Transcripción SOXF/fisiología , Sarcoma de Kaposi/genética , Proteínas Supresoras de Tumor/fisiología , Replicación Viral/genética , Carcinogénesis/genética , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células Endoteliales/virología , Regulación Viral de la Expresión Génica/genética , Genoma Viral/genética , Células HEK293 , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidad , Proteínas de Homeodominio/genética , Humanos , Sistema Linfático/metabolismo , Sistema Linfático/patología , Sistema Linfático/virología , Factores de Transcripción SOXF/genética , Sarcoma de Kaposi/patología , Sarcoma de Kaposi/virología , Proteínas Supresoras de Tumor/genética
14.
J Virol ; 94(13)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32295923

RESUMEN

The anaphase-promoting complex, or cyclosome (APC/C), is a large E3 ubiquitin ligase composed of 14 subunits. The activity of APC/C oscillates during the cell cycle to ensure a timely transition through each phase by promoting the degradation of important cell cycle regulators. Of the human herpesviruses, cytomegalovirus (HCMV) and Epstein-Barr virus (EBV) both impair the activity of APC/C during their lytic replication cycle through virus-encoded protein kinases. Here, we addressed whether the oncogenic Kaposi's sarcoma-associated herpesvirus (KSHV) deregulates the activity of APC/C during the lytic replication cycle. To this end, we used the well-characterized iSLK.219 cell model of KSHV infection and established a new infection model of primary lymphatic endothelial cells (LECs) infected with a lytically replicating KSHV BAC16 mutant. In contrast to those of EBV and HCMV, the KSHV lytic cycle occurs while the APC/C is active. Moreover, interfering with the activity of APC/C did not lead to major changes in the production of infectious virus. We further investigated whether rereplication stress induced by the unscheduled activation of the APC/C-CDH1 complex affects the number and integrity of KSHV viral episomes. Deep sequencing of the viral episomes and host chromosomes in iSLK.219 cells revealed that, while distinct regions in the cellular chromosomes were severely affected by rereplication stress, the integrity of the viral episomes remained unaltered.IMPORTANCE DNA viruses have evolved complex strategies to gain control over the cell cycle. Several of them target APC/C, a key cellular machinery that controls the timely progression of the cell cycle, by either blocking or enhancing its activity. Here, we investigated the activity of APC/C during the lytic replication cycle of KSHV and found that, in contrast to that of KSHV's close relatives EBV and HCMV, KSHV lytic replication occurs while the APC/C is active. Perturbing APC/C activity by depleting a core protein or the adaptor proteins of the catalytic domain, and hence interfering with normal cell-cycle progression, did not affect virus replication. This suggests that KSHV has evolved to replicate independently of the activity of APC/C and in various cell cycle conditions.


Asunto(s)
Herpesvirus Humano 8/metabolismo , Latencia del Virus/genética , Replicación Viral/fisiología , Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Línea Celular , Células Endoteliales/metabolismo , Regulación Viral de la Expresión Génica/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Cultivo Primario de Células , Sarcoma de Kaposi/virología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Proteínas Virales/metabolismo , Activación Viral/genética , Latencia del Virus/fisiología
15.
Cancer Med ; 8(16): 6995-7005, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31560170

RESUMEN

Matrix metalloproteinase 14 (MMP14), a membrane-associated matrix metalloproteinase, has been shown to influence the invasion and metastasis of several solid tumors. Prospero homeobox protein 1 (PROX1), involved in the development and cell fate determination, is also expressed in malignant diseases functioning either as a tumor-suppressing or oncogenic factor. In certain cancers PROX1 appears to transcriptionally suppress MMP14 expression. This study, therefore, aimed to explore the association between MMP14 and PROX1 and understand their potential as prognostic biomarkers in gastric cancer. The cohort consisted of 313 individuals operated for gastric adenocarcinoma between 2000 and 2009 in the Department of Surgery, Helsinki University Hospital. MMP14 and PROX1 expressions were studied using immunohistochemistry in the patient sample and using immunoblotting and immunofluorescence in gastric cancer cell lines. We generated survival curves using the Kaplan-Meier method, determining significance via the log-rank test. A high MMP14 expression associated with being ≥67 years (P = .041), while a positive nuclear PROX1 expression associated with tumors of a diffuse histological type (P = .041) and a high cytoplasmic PROX1 expression (P < .001). Five-year disease-specific survival among patients with a high MMP14 expression was 35.9% (95% confidence interval [CI] 24.9-46.9), compared to 45.3% (95% CI 38.0-52.6) for patients with a low MMP14 (P = .030). Survival was worse specifically among those with a high MMP14 and absent nuclear PROX1 expression (hazard ratio [HR] 1.65; 95% CI 1.09-2.51; P = .019). Thus, this study confirms that a high MMP14 expression predicts a worse survival in gastric cancer, revealing for the first time that survival is particularly worse when PROX1 is low.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/mortalidad , Biomarcadores de Tumor/genética , Proteínas de Homeodominio/genética , Metaloproteinasa 14 de la Matriz/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Proteínas Supresoras de Tumor/genética , Adenocarcinoma/patología , Anciano , Línea Celular Tumoral , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Estadificación de Neoplasias , Pronóstico , Neoplasias Gástricas/patología
16.
Cells ; 8(9)2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31466240

RESUMEN

Sarcomas are deadly malignant tumors of mesenchymal origin occurring at all ages. The expression and function of the membrane-type matrix metalloproteinase MMP14 is closely related to the mesenchymal cell phenotype, and it is highly expressed in most sarcomas. MMP14 regulates the activity of multiple extracellular and plasma membrane proteins, influencing cell-cell and cell-extracellular matrix (ECM) communication. This regulation mediates processes such as ECM degradation and remodeling, cell invasion, and cancer metastasis. Thus, a comprehensive understanding of the biology of MMP14 in sarcomas will shed light on the mechanisms controlling the key processes in these diseases. Here, we provide an overview of the function and regulation of MMP14 and we discuss their relationship with clinical and pre-clinical MMP14 data in both adult and childhood sarcomas.


Asunto(s)
Tejido Conectivo/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Sarcoma/metabolismo , Adulto , Comunicación Celular , Niño , Matriz Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Sarcoma/patología , Microambiente Tumoral
17.
Mol Metab ; 28: 135-143, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31327756

RESUMEN

OBJECTIVE: Heat Shock Proteins (HSPs) maintain cellular homeostasis under stress. HSP70 represents a major stress-inducible family member and has been identified as a druggable target in inherited cholesterol-sphingolipid storage diseases. We investigated if HSP70 modulates cholesterol accumulation in more common conditions related to atherogenesis. METHODS: We studied the effects of recombinant HSP70 in cholesterol-laden primary macrophages from human blood donors and pharmacological HSP70 upregulation in high-cholesterol diet fed zebrafish. RESULTS: Recombinant HSP70 facilitated cholesterol removal from primary human macrophage foam cells. RNA sequencing revealed that HSP70 induced a robust transcriptional re-programming, including upregulation of key targets of liver X receptors (LXR), master regulators of whole-body cholesterol removal. Mechanistically, HSP70 interacted with the macrophage LXRalpha promoter, increased LXRalpha and its target mRNAs, and led to elevated levels of key proteins facilitating cholesterol efflux, including ATP-binding cassette transporters A1 and G1. Pharmacological augmentation of endogenous HSP70 in high-cholesterol diet fed zebrafish activated LXR and its target mRNAs and reduced cholesterol storage at the whole organism level. CONCLUSION: These data demonstrate that HSP70 exerts a cholesterol lowering effect in primary human cells and animals and uncover a nuclear action of HSP70 in mediating cross-talk between HSP and LXR transcriptional regulation.


Asunto(s)
Colesterol/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Receptores X del Hígado/metabolismo , Animales , Colesterol/administración & dosificación , Dieta , Humanos , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Proteínas Recombinantes/metabolismo , Pez Cebra
18.
Sci Rep ; 8(1): 9531, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29934628

RESUMEN

The transcription factor PROX1 is essential for development and cell fate specification. Its function in cancer is context-dependent since PROX1 has been shown to play both oncogenic and tumour suppressive roles. Here, we show that PROX1 suppresses the transcription of MMP14, a metalloprotease involved in angiogenesis and cancer invasion, by binding and suppressing the activity of MMP14 promoter. Prox1 deletion in murine dermal lymphatic vessels in vivo and in human LECs increased MMP14 expression. In a hepatocellular carcinoma cell line expressing high endogenous levels of PROX1, its silencing increased both MMP14 expression and MMP14-dependent invasion in 3D. Moreover, PROX1 ectopic expression reduced the MMP14-dependent 3D invasiveness of breast cancer cells and angiogenic sprouting of blood endothelial cells in conjunction with MMP14 suppression. Our study uncovers a new transcriptional regulatory mechanism of cancer cell invasion and endothelial cell specification.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Metaloproteinasa 14 de la Matriz/genética , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular , Células Endoteliales/metabolismo , Células Endoteliales/patología , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/genética , Humanos , Vasos Linfáticos/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
19.
Elife ; 72018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29712618

RESUMEN

Lymphatic invasion and lymph node metastasis correlate with poor clinical outcome in melanoma. However, the mechanisms of lymphatic dissemination in distant metastasis remain incompletely understood. We show here that exposure of expansively growing human WM852 melanoma cells, but not singly invasive Bowes cells, to lymphatic endothelial cells (LEC) in 3D co-culture facilitates melanoma distant organ metastasis in mice. To dissect the underlying molecular mechanisms, we established LEC co-cultures with different melanoma cells originating from primary tumors or metastases. Notably, the expansively growing metastatic melanoma cells adopted an invasively sprouting phenotype in 3D matrix that was dependent on MMP14, Notch3 and ß1-integrin. Unexpectedly, MMP14 was necessary for LEC-induced Notch3 induction and coincident ß1-integrin activation. Moreover, MMP14 and Notch3 were required for LEC-mediated metastasis of zebrafish xenografts. This study uncovers a unique mechanism whereby LEC contact promotes melanoma metastasis by inducing a reversible switch from 3D growth to invasively sprouting cell phenotype.


Asunto(s)
Neoplasias de la Mama/patología , Endotelio Linfático/patología , Integrina beta1/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Metaloproteinasa 14 de la Matriz/metabolismo , Receptor Notch3/metabolismo , Animales , Apoptosis , Neoplasias de la Mama/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Endotelio Linfático/metabolismo , Femenino , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Pulmonares/metabolismo , Metástasis Linfática , Ratones , Ratones SCID , Invasividad Neoplásica , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
20.
Bio Protoc ; 8(18): e3027, 2018 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-34395813

RESUMEN

Xenograft models, and in particular the mouse xenograft model, where human cancer cells are transplanted into immunocompromised mice, have been used extensively in cancer studies. Although these models have contributed enormously to our understanding of cancer biology, the zebrafish xenograft model offers several advantages over the mouse model. Zebrafish embryos can be easily cultured in large quantities, are small and easy to handle, making it possible to use a high number of embryos for each experimental condition. Young embryos lack an efficient immune system. Therefore the injected cancer cells are not rejected, and the formation of primary tumors and micrometastases is rapid. Transparency of the embryos enables imaging of primary tumors and metastases in an intact and living embryo. Here we describe a method where GFP expressing tumor cells are injected into pericardial space of zebrafish embryos. At four days post-injection, the embryos are imaged and the formation of primary tumor and distant micrometastases are analyzed.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...