Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cancer Metastasis Rev ; 36(1): 7-21, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28321632

RESUMEN

Progress in understanding and treating metastatic melanoma is the result of decades of basic and translational research as well as the development of better in vitro tools for modeling the disease. Here, we review the latest therapeutic options for metastatic melanoma and the known genetic and non-genetic mechanisms of resistance to these therapies, as well as the in vitro toolbox that has provided the greatest insights into melanoma progression. These include next-generation sequencing technologies and more complex 2D and 3D cell culture models to functionally test the data generated by genomics approaches. The combination of hypothesis generating and hypothesis testing paradigms reviewed here will be the foundation for the next phase of metastatic melanoma therapies in the coming years.


Asunto(s)
Melanoma/patología , Melanoma/terapia , Medicina de Precisión/métodos , Investigación Biomédica Traslacional/métodos , Animales , Humanos
2.
Exp Dermatol ; 27(5): 578-586, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29697862

RESUMEN

Melanoma is one of the most studied neoplasia, although laboratory techniques used for investigating this tumor are not fully reliable. Animal models may not predict the human response due to differences in skin physiology and immunity. In addition, international guidelines recommend to develop processes that contribute to the reduction, refinement and replacement of animals for experiments (3Rs). Adherent cell culture has been widely used for the study of melanoma to obtain important information regarding melanoma biology. Nonetheless, these cells grow in adhesion on the culture substrate which differs considerably from the situation in vivo. Melanoma grows in a 3D spatial conformation where cells are subjected to a heterogeneous exposure to oxygen and nutrient. In addition, cell-cell and cell-matrix interaction play a crucial role in the pathobiology of the tumor as well as in the response to therapeutic agents. To better study, melanoma new techniques, including spherical models, tumorospheres and melanoma skin equivalents, have been developed. These 3D models allow to study tumors in a microenvironment that is more close to the in vivo situation and are less expensive and time-consuming than animal studies. This review will also describe the new technologies applied to skin reconstructs such as organ-on-a-chip that allows skin perfusion through microfluidic platforms. 3D in vitro models, based on the new technologies, are becoming more sophisticated, representing at a great extent the in vivo situation, the "perfect" model that will allow less involvement of animals up to their complete replacement, is still far from being achieved.


Asunto(s)
Técnicas de Cultivo de Célula , Melanoma , Modelos Biológicos , Humanos
3.
Int J Mol Sci ; 18(7)2017 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-28718799

RESUMEN

Skin cancer, which includes melanoma and squamous cell carcinoma, represents the most common type of cutaneous malignancy worldwide, and its incidence is expected to rise in the near future. This condition derives from acquired genetic dysregulation of signaling pathways involved in the proliferation and apoptosis of skin cells. The development of animal models has allowed a better understanding of these pathomechanisms, with the possibility of carrying out toxicological screening and drug development. In particular, the zebrafish (Danio rerio) has been established as one of the most important model organisms for cancer research. This model is particularly suitable for live cell imaging and high-throughput drug screening in a large-scale fashion. Thanks to the recent advances in genome editing, such as the clustered regularly-interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) methodologies, the mechanisms associated with cancer development and progression, as well as drug resistance can be investigated and comprehended. With these unique tools, the zebrafish represents a powerful platform for skin cancer research in the development of target therapies. Here, we will review the advantages of using the zebrafish model for drug discovery and toxicological and phenotypical screening. We will focus in detail on the most recent progress in the field of zebrafish model generation for the study of melanoma and squamous cell carcinoma (SCC), including cancer cell injection and transgenic animal development. Moreover, we will report the latest compounds and small molecules under investigation in melanoma zebrafish models.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Pez Cebra/fisiología , Animales , Modelos Animales de Enfermedad , Descubrimiento de Drogas , Humanos
4.
Int J Mol Sci ; 17(1)2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26771605

RESUMEN

Squamous Cell Carcinoma-derived Stem-like Cells (SCC-SC) originate from alterations in keratinocyte stem cells (KSC) gene expression and sustain tumor development, invasion and recurrence. Since survivin, a KSC marker, is highly expressed in SCC-SC, we evaluate its role in SCC-SC cell growth and SCC models. Survivin silencing by siRNA decreases clonal growth of SCC keratinocytes and viability of total, rapidly adhering (RAD) and non-RAD (NRAD) cells from primary SCC. Similarly, survivin silencing reduces the expression of stem cell markers (OCT4, NOTCH1, CD133, ß1-integrin), while it increases the level of differentiation markers (K10, involucrin). Moreover, survivin silencing improves the malignant phenotype of SCC 3D-reconstruct, as demonstrated by reduced epidermal thickness, lower Ki-67 positive cell number, and decreased expression of MMP9 and psoriasin. Furthermore, survivin depletion by siRNA in Ras(G12V)-IκBα-derived tumors leads to smaller tumor formation characterized by lower mitotic index and reduced expression of the tumor-associated marker HIF1α, VEGF and CD51. Therefore, our results indicate survivin as a key gene in regulating SCC cancer stem cell formation and cSCC development.


Asunto(s)
Carcinogénesis/genética , Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Inhibidoras de la Apoptosis/genética , Queratinocitos/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias Cutáneas/genética , Antígeno AC133 , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Técnicas de Cultivo de Célula , Proliferación Celular , Supervivencia Celular , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Inhibidoras de la Apoptosis/metabolismo , Integrina alfaV/genética , Integrina alfaV/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Queratinocitos/patología , Ratones , Ratones SCID , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Péptidos/genética , Péptidos/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transducción de Señal , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Survivin , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
Int J Mol Sci ; 16(11): 26291-302, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26540052

RESUMEN

The Notch signaling pathway orchestrates cell fate by either inducing cell differentiation or maintaining cells in an undifferentiated state. This study aims to evaluate Notch expression and function in normal human keratinocytes. Notch1 is expressed in all epidermal layers, though to a different degree of intensity, with a dramatic decrease during ageing. Notch1 intracellular domain (N1ICD) levels are decreased during transit from keratinocyte stem cells (KSC) to transit amplifying (TA) cells, mimicking survivin expression in samples from donors of all ages. Calcium markedly reduces N1ICD levels in keratinocytes. N1ICD overexpression induces the up-regulation of survivin and the down-regulation of keratin 10 and involucrin, while increasing the S phase of the cell cycle. On the other hand, Notch1 inhibition (DAPT) dose-dependently decreases survivin, stimulates differentiation, and reduces keratinocyte proliferation in samples from donors of all ages. Silencing Notch downgrades survivin and increases keratin 10. In addition, Notch1 inhibition decreases survivin levels and proliferation both in KSC and TA cells. Finally, while survivin overexpression decreases keratinocyte differentiation and increases N1ICD expression both in KSC and TA cells, silencing survivin results in N1ICD down-regulation and an increase in differentiation markers. These results suggest that the Notch1/survivin crosstalk contributes to the maintenance of stemness in human keratinocytes.


Asunto(s)
Autorrenovación de las Células , Senescencia Celular , Proteínas Inhibidoras de la Apoptosis/metabolismo , Queratinocitos/metabolismo , Receptores Notch/metabolismo , Células Madre/metabolismo , Adolescente , Adulto , Factores de Edad , Anciano , Calcio/metabolismo , Proliferación Celular , Senescencia Celular/genética , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Persona de Mediana Edad , Unión Proteica , Receptor Notch1/metabolismo , Transducción de Señal , Survivin , Adulto Joven
6.
Exp Dermatol ; 22(4): 255-61, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23528210

RESUMEN

Epidermal fatty acid-binding protein (E-FABP) is a lipid carrier, originally discovered in human epidermis. We show that E-FABP is almost exclusively expressed in postmitotic (PM) keratinocytes, corresponding to its localization in the highest suprabasal layers, while it is barely expressed in keratinocyte stem cells (KSC) and transit amplifying (TA) keratinocytes. Transfection of normal human keratinocytes with recombinant (r) E-FABP induces overexpression of K10 and involucrin. On the other hand, E-FABP inhibition by siRNA downregulates K10 and involucrin expression in normal keratinocytes through NF-κB and JNK signalling pathways. E-FABP is highly expressed in psoriatic epidermis, and it is mainly localized in stratum spinosum. Psoriatic PM keratinocytes overexpress E-FABP as compared to the same population in normal epidermis. E-FABP inhibition in psoriatic keratinocytes markedly reduces differentiation, while it upregulates psoriatic markers such as survivin and K16. However, under high-calcium conditions, E-FABP silencing downregulates K10 and involucrin, while survivin and K16 expression is completely abolished. These data strongly indicate that E-FABP plays an important role in keratinocyte differentiation. Moreover, E-FABP modulates differentiation in psoriatic keratinocytes.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/metabolismo , Queratinocitos/citología , Queratinocitos/fisiología , Psoriasis/metabolismo , Psoriasis/patología , Diferenciación Celular/fisiología , Células Cultivadas , Proteínas de Unión a Ácidos Grasos/antagonistas & inhibidores , Proteínas de Unión a Ácidos Grasos/genética , Humanos , Queratina-10/metabolismo , Queratinocitos/patología , Precursores de Proteínas/metabolismo , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
Int J Mol Sci ; 14(10): 19540-55, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24077125

RESUMEN

In human epidermis, keratinocyte stem cells (KSC) are characterized by high levels of ß1-integrin, resulting in the rapid adhesion to type IV collagen. Since epithelial tumors originate from KSC, we evaluated the features of rapidly adhering (RAD) keratinocytes derived from primary human squamous cell carcinoma of the skin (cSCC). RAD cells expressed higher levels of survivin, a KSC marker, as compared to non-rapidly adhering (NRAD) cells. Moreover, RAD cells proliferated to a greater extent and were more efficient in forming colonies than NRAD cells. RAD cells also migrated significantly better than NRAD cells. When seeded in a silicone chamber and grafted onto the back skin of NOD SCID mice, RAD cells formed tumors 2-4 fold bigger than those derived from NRAD cells. In tumors derived from RAD cells, the mitotic index was significantly higher than in those derived from NRAD cells, while Ki-67 and survivin expression were more pronounced in RAD tumors. This study suggests that SCC RAD stem cells play a critical role in the formation and development of epithelial tumors.


Asunto(s)
Carcinoma de Células Escamosas/patología , Neoplasias Cutáneas/patología , Células Madre/patología , Células 3T3 , Animales , Carcinogénesis/patología , Carcinoma de Células Escamosas/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular , Células Cultivadas , Epidermis/metabolismo , Epidermis/patología , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , Integrina beta1/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Ratones SCID , Proteínas Represoras/metabolismo , Neoplasias Cutáneas/metabolismo , Células Madre/metabolismo , Survivin
8.
Cancer Res ; 83(7): 1128-1146, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-36946761

RESUMEN

Clinical management of melanomas with NRAS mutations is challenging. Targeting MAPK signaling is only beneficial to a small subset of patients due to resistance that arises through genetic, transcriptional, and metabolic adaptation. Identification of targetable vulnerabilities in NRAS-mutated melanoma could help improve patient treatment. Here, we used multiomics analyses to reveal that NRAS-mutated melanoma cells adopt a mesenchymal phenotype with a quiescent metabolic program to resist cellular stress induced by MEK inhibition. The metabolic alterations elevated baseline reactive oxygen species (ROS) levels, leading these cells to become highly sensitive to ROS induction. In vivo xenograft experiments and single-cell RNA sequencing demonstrated that intratumor heterogeneity necessitates the combination of a ROS inducer and a MEK inhibitor to inhibit both tumor growth and metastasis. Ex vivo pharmacoscopy of 62 human metastatic melanomas confirmed that MEK inhibitor-resistant tumors significantly benefited from the combination therapy. Finally, oxidative stress response and translational suppression corresponded with ROS-inducer sensitivity in 486 cancer cell lines, independent of cancer type. These findings link transcriptional plasticity to a metabolic phenotype that can be inhibited by ROS inducers in melanoma and other cancers. SIGNIFICANCE: Metabolic reprogramming in drug-resistant NRAS-mutated melanoma cells confers sensitivity to ROS induction, which suppresses tumor growth and metastasis in combination with MAPK pathway inhibitors.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Especies Reactivas de Oxígeno , Proteínas Proto-Oncogénicas B-raf/genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Línea Celular Tumoral , Mutación , Proteínas de la Membrana/genética , GTP Fosfohidrolasas/genética
9.
J Invest Dermatol ; 142(4): 1171-1182.e10, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34757069

RESUMEN

The STK11/LKB1 is a tumor suppressor involved in metabolism and cell motility. In BRAFV600E melanoma, STK11 is inactivated by extracellular signal‒regulated kinase and RSK, preventing it from binding and activating adenosine monophosphate-activated protein kinase and promoting melanoma cell proliferation. Although STK11 mutations occur in 5‒10% of cutaneous melanoma, few functional studies have been performed. By knocking out STK11 with CRISPR/Cas9 in two human BRAF-mutant melanoma cell lines, we found that STK11 loss reduced the sensitivity to a BRAF inhibitor. More strikingly, STK11 loss led to an increased invasive phenotype in both three-dimensional spheroids and in vivo zebrafish xenograft models. STK11 overexpression consistently reversed the invasive phenotype. Interestingly, STK11 knockout increased invasion also in an NRAS-mutant melanoma cell line. Furthermore, although STK11 was expressed in primary human melanoma tumors, its expression significantly decreased in melanoma metastases, especially in brain metastases. In the STK11-knockout cells, we observed increased activating phosphorylation of signal transducer and activator of transcription 3/5 and FAK. Using inhibitors of signal transducer and activator of transcription 3/5 and FAK, we reversed the invasive phenotype in both BRAF- and NRAS-mutated cells. Our findings confirm an increased invasive phenotype on STK11 inactivation in BRAF- and NRAS-mutant cutaneous melanoma that can be targeted by signal transducer and activator of transcription 3/5 and FAK inhibition.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal , Humanos , Melanoma/patología , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Factor de Transcripción STAT3/metabolismo , Neoplasias Cutáneas/genética , Pez Cebra/metabolismo , Melanoma Cutáneo Maligno
10.
Foods ; 10(8)2021 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-34441474

RESUMEN

The anti-proliferative/pro-oxidant efficacy of green pea, soybean, radish, Red Rambo radish, and rocket microgreens, cultivated under either fluorescent lighting (predominant spectral peaks in green and orange) or combination light-emitting diode (LED, predominant spectral peak in blue) was investigated using Ewing sarcoma lines, RD-ES and A673, respectively. All aqueous microgreen extracts significantly reduced cell proliferation (cancer prevention effect) to varying extents in two-dimensional sarcoma cell cultures. The effect of the polyphenol fraction in the aqueous food matrix was unrelated to total polyphenol content, which differed between species and light treatment. Only Pisum sativum (LED-grown) extracts exercised anti-proliferative and pro-apoptotic effects in both three-dimensional RD-ES and A673 spheroids (early tumor progression prevention), without cytotoxic effects on healthy L929 fibroblasts. A similar anti-tumor effect of Red Rambo radish (LED and fluorescent-grown) was evident only in the RD-ES spheroids. Aside from the promising anti-tumor potential of the polyphenol fraction of green pea microgreens, the latter also displayed favorable growth quality parameters, along with radish, under both light treatments over the 10 day cultivation period.

11.
J Vis Exp ; (173)2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34309602

RESUMEN

Breast cancer is the leading cause of mortality in women. The growth of breast cancer cells and their subsequent metastasis is a key factor for its progression. Although the mechanisms involved in promoting breast cancer growth have been intensively studied using monocultures of breast cancer cells such as MCF-7 cells, the contribution of other cell types, such as vascular and lymphatic endothelial cells that are intimately involved in tumor growth, has not been investigated in depth. Cell-cell interaction plays a key role in tumor growth and progression. Neoangiogenesis, or the development of vessels, is essential for tumor growth, whereas the lymphatic system serves as a portal for cancer cell migration and subsequent metastasis. Recent studies provide evidence that vascular and lymphatic endothelial cells can significantly influence cancer cell growth. These observations imply a need for developing in vitro models that would more realistically reflect breast cancer growth processes in vivo. Moreover, restrictions in animal research require the development of ex vivo models to elucidate better the mechanisms involved. This article describes the development of breast cancer spheroids composed of both breast cancer cells (estrogen receptor-positive MCF-7 cells) and vascular and/or lymphatic endothelial cells. The protocol describes a detailed step-by-step approach in creating dual-cell spheroids using two different approaches, hanging drop (gold standard and cheap) and 96-well U-bottom plates (expensive). In-depth instructions are provided for how to delicately pick up the formed spheroids to monitor growth by microscopic sizing and assessing viability using dead and live cell staining. Moreover, procedures to fix the spheroids for sectioning and staining with growth-specific antibodies to differentiate growth patterns in spheroids are delineated. Additionally, details for preparing spheroids with transfected cells and methods to extract RNA for molecular analysis are provided. In conclusion, this article provides in-depth instructions for preparing multi-cell spheroids for breast cancer research.


Asunto(s)
Neoplasias de la Mama , Animales , Mama , Células Endoteliales , Femenino , Humanos , Neovascularización Patológica , Esferoides Celulares
12.
Cancer Res ; 81(23): 6044-6057, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34645608

RESUMEN

CD271 (NGFR) is a neurotrophin receptor that belongs to the tumor necrosis receptor (TNFR) family. Upon ligand binding, CD271 can mediate either survival or cell death. Although the role of CD271 as a marker of tumor-initiating cells is still a matter of debate, its role in melanoma progression has been well documented. Moreover, CD271 has been shown to be upregulated after exposure to both chemotherapy and targeted therapy. In this study, we demonstrate that activation of CD271 by a short ß-amyloid-derived peptide (Aß(25-35)) in combination with either chemotherapy or MAPK inhibitors induces apoptosis in 2D and 3D cultures of eight melanoma cell lines. This combinatorial treatment significantly reduced metastasis in a zebrafish xenograft model and led to significantly decreased tumor volume in mice. Administration of Aß(25-35) in ex vivo tumors from immunotherapy- and targeted therapy-resistant patients significantly reduced proliferation of melanoma cells, showing that activation of CD271 can overcome drug resistance. Aß(25-35) was specific to CD271-expressing cells and induced CD271 cleavage and phosphorylation of JNK (pJNK). The direct protein-protein interaction of pJNK with CD271 led to PARP1 cleavage, p53 and caspase activation, and pJNK-dependent cell death. Aß(25-35) also mediated mitochondrial reactive oxygen species (mROS) accumulation, which induced CD271 overexpression. Finally, CD271 upregulation inhibited mROS production, revealing the presence of a negative feedback loop in mROS regulation. These results indicate that targeting CD271 can activate cell death pathways to inhibit melanoma progression and potentially overcome resistance to targeted therapy. SIGNIFICANCE: The discovery of a means to specifically activate the CD271 death domain reveals unknown pathways mediated by the receptor and highlights new treatment possibilities for melanoma.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma/tratamiento farmacológico , Terapia Molecular Dirigida , Proteínas del Tejido Nervioso/agonistas , Receptores de Factor de Crecimiento Nervioso/agonistas , Animales , Apoptosis , Proliferación Celular , Quimioterapia Combinada , Femenino , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
13.
Nat Cancer ; 2(12): 1387-1405, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34957415

RESUMEN

Secreted extracellular vesicles (EVs) influence the tumor microenvironment and promote distal metastasis. Here, we analyzed the involvement of melanoma-secreted EVs in lymph node pre-metastatic niche formation in murine models. We found that small EVs (sEVs) derived from metastatic melanoma cell lines were enriched in nerve growth factor receptor (NGFR, p75NTR), spread through the lymphatic system and were taken up by lymphatic endothelial cells, reinforcing lymph node metastasis. Remarkably, sEVs enhanced lymphangiogenesis and tumor cell adhesion by inducing ERK kinase, nuclear factor (NF)-κB activation and intracellular adhesion molecule (ICAM)-1 expression in lymphatic endothelial cells. Importantly, ablation or inhibition of NGFR in sEVs reversed the lymphangiogenic phenotype, decreased lymph node metastasis and extended survival in pre-clinical models. Furthermore, NGFR expression was augmented in human lymph node metastases relative to that in matched primary tumors, and the frequency of NGFR+ metastatic melanoma cells in lymph nodes correlated with patient survival. In summary, we found that NGFR is secreted in melanoma-derived sEVs, reinforcing lymph node pre-metastatic niche formation and metastasis.


Asunto(s)
Vesículas Extracelulares , Melanoma , Animales , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Humanos , Linfangiogénesis/fisiología , Metástasis Linfática , Melanoma/metabolismo , Ratones , Proteínas del Tejido Nervioso , Receptores de Factor de Crecimiento Nervioso/genética , Microambiente Tumoral
15.
J Invest Dermatol ; 136(10): 2049-2058, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27328305

RESUMEN

CD271 is a neurotrophin receptor variably expressed in melanoma. Although contradictory data are reported on its role as a marker of tumor-initiating cells, little is known about its function in tumor progression. CD271 expression was higher in spheroids derived from freshly isolated cells of primary melanomas and in primary WM115 and WM793-B cell lines, and it decreased during progression to advanced stages in cells isolated from metastatic melanomas and in metastatic WM266-4 and 1205Lu cell lines. Moreover, CD271 was scarcely detected in the highly invasive spheroids (SKMEL28 and 1205Lu). CD271, originally expressed in the epidermis of skin reconstructs, disappeared when melanoma started to invade the dermis. SKMEL8 CD271(-) cells showed greater proliferation and invasiveness in vitro and were associated with a higher number of metastases in zebrafish compared with CD271(+) cells. CD271 silencing in WM115 induced a more aggressive phenotype in vitro and in vivo. On the contrary, CD271 overexpression in SKMEL28 cells reduced invasion in vitro, and CD271 overexpressing 1205Lu cells was associated with a lower percentage of metastases in zebrafish. A reduced cell-cell adhesion was also observed in the absence of CD271. Taken together, these results indicate that CD271 loss is critical for melanoma progression and metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Melanoma/patología , Proteínas del Tejido Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/genética , Neoplasias Cutáneas/patología , Animales , Adhesión Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Regulación hacia Abajo , Silenciador del Gen , Humanos , Melanoma/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neoplasias Cutáneas/genética , Pez Cebra
16.
Cell Signal ; 28(7): 753-63, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26806620

RESUMEN

Phosphodiesterases 4 (PDE4) act as proinflammatory enzymes via degradation of cAMP, whereas PDE4 inhibitors play an anti-inflammatory role in vitro and in vivo. In particular, apremilast has been recently approved for the treatment of psoriasis and psoriatic arthritis. However, little is known on the expression pattern of PDE4 in psoriasis. We report that PDE4B and PDE4D mRNA are overexpressed in peripheral blood mononuclear cells (PBMC) from psoriasis, as compared with normal controls, while apremilast reduces PBMC production of a number of pro-inflammatory cytokines and increases the levels of anti-inflammatory mediators. PDE4 expression is up-regulated in psoriatic dermis as compared with normal skin, with particular regard to fibroblasts. This is confirmed in vitro, where both dermal fibroblasts (DF) and, to a greater extent, myofibroblasts (DM) express all PDE4 isoforms at the mRNA and protein level. Because PDE4 interacts with the nerve growth factor (NGF) receptor CD271 in lung fibroblasts, we evaluated the relationship and function of PDE4 and CD271 in normal human skin fibroblasts. All PDE4 isoforms co-immunoprecipitate with CD271 in DM, while apremilast inhibits apoptosis induced by ß-amyloid, a CD271 ligand, in DM. Furthermore, apremilast significantly reduces NGF- and transforming growth factor-ß1 (TGF-ß1)-induced fibroblast migration, and inhibits DF differentiation into DM mediated by NGF or TGF-ß1. Finally, in DM, apremilast significantly reduces cAMP degradation induced by treatment with ß-amyloid. Taken together, these results indicate that PDE4 play an important role in psoriasis. In addition, the study reveals that the PDE4/CD271 complex could be important in modulating fibroblast functions.


Asunto(s)
Adapaleno/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Dermis/patología , Inflamación/enzimología , Miofibroblastos/metabolismo , Psoriasis/sangre , Psoriasis/enzimología , Talidomida/análogos & derivados , Adulto , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Citocinas/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica , Humanos , Inmunohistoquímica , Inmunoprecipitación , Inflamación/sangre , Inflamación/patología , Mediadores de Inflamación/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Leucocitos Mononucleares/metabolismo , Masculino , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Psoriasis/patología , Talidomida/farmacología
17.
J Invest Dermatol ; 135(3): 786-795, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25330297

RESUMEN

CD271 is the low-affinity neurotrophin (p75NTR) receptor that belongs to the tumor necrosis factor receptor superfamily. Because in human epidermis, CD271 is predominantly expressed in transit-amplifying (TA) cells, we evaluated the role of this receptor in keratinocyte differentiation and in the transition from keratinocyte stem cells (KSCs) to progeny. Calcium induced an upregulation of CD271 in subconfluent keratinocytes, which was prevented by CD271 small interfering RNA. Furthermore, CD271 overexpression provoked the switch of KSCs to TA cells, whereas silencing CD271 induced TA cells to revert to a KSC phenotype, as shown by the expression of ß1-integrin and by the increased clonogenic ability. CD271(+) keratinocytes sorted from freshly isolated TA cells expressed more survivin and keratin 15 (K15) compared with CD271(-) cells and displayed a higher proliferative capacity. Early differentiation markers and K15 were more expressed in the skin equivalent generated from CD271(+) TA than from those derived from CD271(-) TA cells. By contrast, late differentiation markers were more expressed in skin equivalents from CD271(-) than in reconstructs from CD271(+) TA cells. Finally, skin equivalents originated from CD271(-) TA cells displayed a psoriatic phenotype. These results indicate that CD271 is critical for keratinocyte differentiation and regulates the transition from KSCs to TA cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Epidérmicas , Queratinocitos/citología , Proteínas del Tejido Nervioso/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Células Madre/citología , Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Epidermis/efectos de los fármacos , Epidermis/metabolismo , Humanos , Técnicas In Vitro , Proteínas Inhibidoras de la Apoptosis/metabolismo , Queratina-15/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Fenotipo , Psoriasis/patología , ARN Interferente Pequeño/farmacología , Receptores de Factor de Crecimiento Nervioso/deficiencia , Receptores de Factor de Crecimiento Nervioso/genética , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Survivin
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA