Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 883
Filtrar
1.
Fitoterapia ; 177: 106100, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38972550

RESUMEN

Melanoma is the most aggressive form of skin cancer and originates from genetic mutations in melanocytes. The disease is multifactorial, but its main cause is overexposure to UV radiation. Currently, available chemotherapy expresses little to no results, which may justify the extensive use of natural products to treat this cancer. In this study, we reviewed the inhibition of melanoma angiogenesis by natural products and its potential mechanisms using literature from PubMed, EMBASE, Web of Science, Ovid, ScienceDirect and China National Knowledge Infrastructure databases. According to summarizes 27 natural products including alkaloids, polyphenols, terpenoids, flavonoids, and steroids that effectively inhibit angiogenesis in melanoma. In addition to these there are 15 crude extracts that can be used as promising agents to inhibit angiogenesis, but their core components still deserve further investigation. There are current studies on melanoma angiogenesis involving oxidative stress, immune-inflammatory response, cell proliferation and migration and capillary formation. The above natural products can be involved in melanoma angiogenesis through core targets such as VE-cadherin, COX-2, iNOS, VEGF, bFGF, FGF2,MMP2,MMP9,IL-1ß,IL-6 play a role in inhibiting melanoma angiogenesis. Effective excavation of natural products can not only clarify the mechanism of drug action and key targets, but also help to promote the preclinical research of natural products for melanoma treatment and further promote the development of new clinical drugs, which will bring the gospel to the vast number of patients who are deeply afflicted by melanoma.


Asunto(s)
Inhibidores de la Angiogénesis , Productos Biológicos , Melanoma , Neovascularización Patológica , Neoplasias Cutáneas , Melanoma/tratamiento farmacológico , Melanoma/irrigación sanguínea , Humanos , Productos Biológicos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos Fitogénicos/farmacología , Angiogénesis
2.
J Biophotonics ; 17(10): e202400208, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39080837

RESUMEN

Early invasive skin melanoma (EISM) associated with partial tumor invasion to the thin and optically complex papillary dermis (PD) represents a critical stage before the onset of metastasis. EISM lesions may be accompanied by angiogenesis, which can alter the PD's blood and fibril contents. A comprehensive understanding about these interconnected processes is essential for enhancing the efficacy of EISM optical evaluation methodologies. Employing a first-principles computational approach supported by measured data, we systematically assess the impact that angiogenesis can have on the EISM's spectral responses. Our findings indicate that these responses are discernibly affected by angiogenesis under distinct physiological conditions, with more substantial tissue alterations leading to accentuated spectral changes in the 550-600 nm region. Accordingly, we propose the use of a customized low-cost spectral index to monitor these processes. Furthermore, our investigation provides a high-fidelity in silico platform for interdisciplinary research on the photobiology of evolving skin melanomas.


Asunto(s)
Progresión de la Enfermedad , Melanoma , Invasividad Neoplásica , Neovascularización Patológica , Neoplasias Cutáneas , Melanoma/patología , Melanoma/irrigación sanguínea , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/irrigación sanguínea , Humanos , Neovascularización Patológica/patología , Melanoma Cutáneo Maligno , Angiogénesis
3.
Br J Cancer ; 131(3): 457-467, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38902534

RESUMEN

BACKGROUND/OBJECTIVES: Pseudo-vascular network formation in vitro is considered a key characteristic of vasculogenic mimicry. While many cancer cell lines form pseudo-vascular networks, little is known about the spatiotemporal dynamics of these formations. METHODS: Here, we present a framework for monitoring and characterising the dynamic formation and dissolution of pseudo-vascular networks in vitro. The framework combines time-resolved optical microscopy with open-source image analysis for network feature extraction and statistical modelling. The framework is demonstrated by comparing diverse cancer cell lines associated with vasculogenic mimicry, then in detecting response to drug compounds proposed to affect formation of vasculogenic mimics. Dynamic datasets collected were analysed morphometrically and a descriptive statistical analysis model was developed in order to measure stability and dissimilarity characteristics of the pseudo-vascular networks formed. RESULTS: Melanoma cells formed the most stable pseudo-vascular networks and were selected to evaluate the response of their pseudo-vascular networks to treatment with axitinib, brucine and tivantinib. Tivantinib has been found to inhibit the formation of the pseudo-vascular networks more effectively, even in dose an order of magnitude less than the two other agents. CONCLUSIONS: Our framework is shown to enable quantitative analysis of both the capacity for network formation, linked vasculogenic mimicry, as well as dynamic responses to treatment.


Asunto(s)
Neovascularización Patológica , Humanos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Línea Celular Tumoral , Melanoma/patología , Melanoma/irrigación sanguínea , Melanoma/tratamiento farmacológico , Axitinib/farmacología
4.
Proc Natl Acad Sci U S A ; 119(33): e2122716119, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35960843

RESUMEN

The microenvironment of malignant melanomas defines the properties of tumor blood vessels and regulates infiltration and vascular dissemination of immune and cancer cells, respectively. Previous research in other cancer entities suggested the complement system as an essential part of the tumor microenvironment. Here, we confirm activation of the complement system in samples of melanoma patients and murine melanomas. We identified the tumor endothelium as the starting point of the complement cascade. Generation of complement-derived C5a promoted the recruitment of neutrophils. Upon contact with the vascular endothelium, neutrophils were further activated by complement membrane attack complexes (MACs). MAC-activated neutrophils release neutrophil extracellular traps (NETs). Close to the blood vessel wall, NETs opened the endothelial barrier as indicated by an enhanced vascular leakage. This facilitated the entrance of melanoma cells into the circulation and their systemic spread. Depletion of neutrophils or lack of MAC formation in complement component 6 (C6)-deficient animals protected the vascular endothelium and prevented vascular intravasation of melanoma cells. Our data suggest that inhibition of MAC-mediated neutrophil activation is a potent strategy to abolish hematogenous dissemination in melanoma.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento , Endotelio Vascular , Trampas Extracelulares , Melanoma , Neutrófilos , Microambiente Tumoral , Animales , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Proteínas del Sistema Complemento , Endotelio Vascular/fisiopatología , Humanos , Melanoma/irrigación sanguínea , Melanoma/inmunología , Melanoma/patología , Ratones , Neutrófilos/inmunología , Permeabilidad
5.
Adv Sci (Weinh) ; 9(10): e2105226, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35088579

RESUMEN

The ability of melanoma to acquire metastasis through the induction of angiogenesis is one of the major causes of skin cancer death. Here, it is found that high transcript levels of DEP domain containing 1B (DEPDC1B) in cutaneous melanomas are significantly associated with a poor prognosis. Tissue microarray analysis indicates that DEPDC1B expression is positively correlated with SOX10 in the different stages of melanoma. Consistently, DEPDC1B is both required and sufficient for melanoma growth, metastasis, angiogenesis, and functions as a direct downstream target of SOX10 to partly mediate its oncogenic activity. In contrast to other tumor types, the DEPDC1B-mediated enhancement of melanoma metastatic potential is not dependent on the activities of RHO GTPase signaling and canonical Wnt signaling, but is acquired through secretion of signal peptide, CUB domain and EGF like domain containing 3 (SCUBE3), which is crucial for promoting angiogenesis in vitro and in vivo. Mechanistically, DEPDC1B regulates SCUBE3 protein stability through the competitive association with ubiquitin ligase cell division cycle 16 (CDC16) to prevent SCUBE3 from undergoing degradation via the ubiquitin-proteasome pathway. Importantly, expression of SOX10, DEPDC1B, and SCUBE3 are positively correlated with microvessel density in the advanced stage of melanomas. In conclusion, it is revealed that a SOX10-DEPDC1B-SCUBE3 regulatory axis promotes melanoma angiogenesis and metastasis, which suggests that targeting secreted SCUBE3 can be a therapeutic strategy against metastatic melanoma.


Asunto(s)
Subunidad Apc6 del Ciclosoma-Complejo Promotor de la Anafase , Proteínas de Unión al Calcio , Proteínas Activadoras de GTPasa , Melanoma , Ubiquitina , Subunidad Apc6 del Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas de Unión al Calcio/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Melanoma/irrigación sanguínea , Ubiquitina/metabolismo
6.
Microvasc Res ; 140: 104305, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34958805

RESUMEN

Angiogenesis is critical for solid tumor growth beyond its minimal size. Previously, we reported that Down Syndrome Candidate Region 1 isoform 1L (DSCR1-1L) was one of the most up-regulated genes in endothelial cells induced by VEGF and histamine, and regulated endothelial cell proliferation, migration and angiogenesis. However, it was not known whether DSCR1-1L played a role in tumor growth. In this study, we found that DSCR1-1L shRNAs significantly inhibited the growth of transplanted melanoma in mice and its associated tumoral angiogenesis. In the gain of function assay, overexpression of DSCR1-1L cDNA in mouse endothelium is sufficient to significantly increase the tumor initiation induced by carcinogen, the growth of xenografted tumor, and the tumor metastasis in our endothelially-expressed DSCR1-1L transgenic mice, in which angiogenesis was induced. It was the first time to find that DSCR1-1L was also expressed in various tumor cells. DSCR1-1L shRNAs inhibited, but overexpression of DSCR1-1L cDNA increased, the tumor cell proliferation and migration. Most recently, we reported that DSCR1-1L modulated angiogenesis by down-regulation of VE-cadherin expression. Here, we found that DSCR1-1L down-regulated the expression of E-cadherin. Hence, DSCR1-1L is an excellent therapeutic target for cancers by regulation of both the endothelial and tumor cells through down-regulating (V)E-cadherin. DSCR1-1L shRNAs have the potential to be developed for clinical application.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Endoteliales/metabolismo , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Proteínas Musculares/metabolismo , Neovascularización Patológica , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Proteínas de Unión al ADN/genética , Células Endoteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanoma/genética , Melanoma/patología , Ratones Desnudos , Proteínas Musculares/genética , Invasividad Neoplásica , Isoformas de Proteínas , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Carga Tumoral
7.
J Invest Dermatol ; 142(7): 1923-1933.e5, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34968503

RESUMEN

Matrix metalloproteinase (MMP) 14 belongs to a large family of zinc-dependent endopeptidases and plays a critical role in skin physiological and pathological processes. Complete loss of the protease resulted in severe developmental defects leading to early death. However, because of the premature death of the mice, the functional significance for endothelial cell (EC) expression of MMP14 in skin physiology and pathology in vivo after birth is yet unknown. Using a mouse model with constitutive EC-specific deletion of Mmp14 (Mmp14EC‒/‒), we showed that mice developed and bred normal, but melanoma growth and metastasis were reduced. Although vascularity was unaltered, vessel permeability was decreased. Deletion of MMP14 in ECs led to increased vessel coverage by pericytes and vascular endothelial-cadherin expression in mice in vivo and in vitro but not in human ECs. Endothelial nitric oxide synthase expression and nitric oxide production were significantly reduced in Mmp14EC‒/‒ ECs and MMP14-silenced human umbilical vein ECs. A direct correlation between endothelial nitric oxide synthase and MMP14 expression was detected in intratumoral vessels of human malignant melanomas. Altogether, we show that endothelial MMP14 controls tumor vessel function during melanoma growth. These data suggest that EC-derived MMP14 direct targeting alone or with vascular stabilizing agents may be therapeutically crucial in inhibiting melanoma growth and metastasis.


Asunto(s)
Metaloproteinasa 14 de la Matriz , Melanoma , Animales , Permeabilidad Capilar , Células Endoteliales/metabolismo , Humanos , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Melanoma/irrigación sanguínea , Melanoma/patología , Ratones , Metástasis de la Neoplasia , Óxido Nítrico Sintasa de Tipo III/metabolismo
8.
Cells ; 10(10)2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34685720

RESUMEN

Different types of cells, such as endothelial cells, tumor-associated fibroblasts, pericytes, and immune cells, release extracellular vesicles (EVs) in the tumor microenvironment. The components of EVs include proteins, DNA, RNA, and microRNA. One of the most important functions of EVs is the transfer of aforementioned bioactive molecules, which in cancer cells may affect tumor growth, progression, angiogenesis, and metastatic spread. Furthermore, EVs affect the presentation of antigens to immune cells via the transfer of nucleic acids, peptides, and proteins to recipient cells. Recent studies have also explored the potential application of EVs in cancer treatment. This review summarizes the mechanisms by which EVs regulate melanoma development, progression, and their potentials to be applied in therapy. We initially describe vesicle components; discuss their effects on proliferation, anti-melanoma immunity, and drug resistance; and finally focus on the effects of EV-derived microRNAs on melanoma pathobiology. This work aims to facilitate our understanding of the influence of EVs on melanoma biology and initiate ideas for the development of novel therapeutic strategies.


Asunto(s)
Vesículas Extracelulares/metabolismo , Melanoma/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , Melanoma/irrigación sanguínea , Melanoma/patología , Melanoma/terapia , Modelos Biológicos , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Microambiente Tumoral , Melanoma Cutáneo Maligno
9.
Int J Mol Sci ; 22(5)2021 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-33803452

RESUMEN

Apoptosis is the main mechanism by which multicellular organisms eliminate damaged or unwanted cells. To regulate this process, a balance between pro-survival and pro-apoptotic proteins is necessary in order to avoid impaired apoptosis, which is the cause of several pathologies, including cancer. Among the anti-apoptotic proteins, Bcl-xL exhibits a high conformational flexibility, whose regulation is strictly controlled by alternative splicing and post-transcriptional regulation mediated by transcription factors or microRNAs. It shows relevant functions in different forms of cancer, including melanoma. In melanoma, Bcl-xL contributes to both canonical roles, such as pro-survival, protection from apoptosis and induction of drug resistance, and non-canonical functions, including promotion of cell migration and invasion, and angiogenesis. Growing evidence indicates that Bcl-xL inhibition can be helpful for cancer patients, but at present, effective and safe therapies targeting Bcl-xL are lacking due to toxicity to platelets. In this review, we summarized findings describing the mechanisms of Bcl-xL regulation, and the role that Bcl-xL plays in melanoma pathobiology and response to therapy. From these findings, it emerged that even if Bcl-xL plays a crucial role in melanoma pathobiology, we need further studies aimed at evaluating the involvement of Bcl-xL and other members of the Bcl-2 family in the progression of melanoma and at identifying new non-toxic Bcl-xL inhibitors.


Asunto(s)
Apoptosis , Resistencia a Antineoplásicos , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Proteína bcl-X/metabolismo , Humanos , Melanoma/tratamiento farmacológico , Melanoma/patología , MicroARNs/metabolismo , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , ARN Neoplásico/metabolismo
10.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800878

RESUMEN

Uveal melanoma (UM) is a malignant tumor that arises in the melanocytes of the uveal tract. It is the most frequent eye cancer, and despite new therapeutic approaches, prognosis is still poor, with up to 50% of patients developing metastasis with no efficient treatment options available. In contrast to cutaneous melanoma, UM is considered an "immune-cold" tumor due to the low mutational burden and the unique immunosuppressive microenvironment. To gain insight into the role of the UM microenvironment in regard to prognosis and metastatic progression, we have performed a pool analysis characterizing the UM microenvironment by using a bioinformatic approach. A variety of scores based on gene expression measuring stromal infiltration were calculated and used to assess association with prognosis. As a result, the highest immune and stromal scores were associated with poor prognosis. Specifically, stromal cells (fibroblasts and endothelial cells), T cells CD8+, natural killer (NK) cells, and macrophages M1 and M2 infiltration were associated with poor prognosis. Contrary to other tumors, lymphocytic infiltration is related to poor prognosis. Only B cells were associated with more favorable prognosis. UM samples scoring high in both angiogenesis (Angio) and antigen presentation (AP) pathways showed a poor prognosis suggesting an additive role of both functions. Almost all these tumors exhibited a chromosome 3 monosomy. Finally, an enrichment analysis showed that tumors classified as high Angio-high AP also activated metabolic pathways such as glycolysis or PI3K-AKT-MTOR. In summary, our pool analysis identified a cluster of samples with angiogenic and inflammatory phenotypes exhibiting poor prognosis and metabolic activation. Our analysis showed robust results replicated in a pool analysis merging different datasets from different analytic platforms.


Asunto(s)
Linfocitos Infiltrantes de Tumor/patología , Melanoma/patología , Neovascularización Patológica/fisiopatología , Neoplasias de la Úvea/patología , Anciano , Animales , Presentación de Antígeno , Análisis por Conglomerados , Conjuntos de Datos como Asunto , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia , Estimación de Kaplan-Meier , Subgrupos Linfocitarios/patología , Macrófagos/patología , Masculino , Melanoma/irrigación sanguínea , Melanoma/genética , Melanoma/inmunología , Redes y Vías Metabólicas , Persona de Mediana Edad , Neovascularización Patológica/genética , Pronóstico , Transducción de Señal , Células del Estroma/patología , Microambiente Tumoral , Neoplasias de la Úvea/irrigación sanguínea , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/inmunología
11.
Cornea ; 40(12): 1519-1524, 2021 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-33591034

RESUMEN

PURPOSE: To investigate whether transvascular indocyanine green (ICG) dye leakage is associated with conjunctival malignancy. METHODS: This is a prospective interventional study. Patients presenting with circumscribed conjunctival melanocytic disorders (CMDs) were included and examined using color photography, anterior segment optical coherence tomography to measure lesion size, and fluorescein and ICG angiography to measure vascular pattern and leakage. Time to vascular leakage was measured by 2 independent observers. Lesions were characterized as benign or malignant based on histopathological features. RESULTS: Thirty patients with CMD were included: 22 lesions were benign (conjunctival nevus, n = 20; conjunctival melanocytic intraepithelial neoplasia without atypia, n = 2) and 8 were malignant (in situ conjunctival melanoma n = 2; invasive conjunctival melanoma, n = 6). Malignant lesions had larger mean maximal diameters (11.0 ± 4.5 vs. 4.2 ± 2.5 mm, P = 0.003) and more frequently showed intrinsic tumor vasculature (8 of 8 vs. 10 of 22, P = 0.007). The mean time to ICG leakage was 350.9 ± 165.9 seconds in benign and 59.6 ± 22.1 seconds (P = 0.002) in malignant lesions and was inversely correlated with lesion size and thickness. CONCLUSIONS: Time to angiographic ICG dye leakage is significantly shorter in malignant versus benign CMD.


Asunto(s)
Conjuntiva/patología , Neoplasias de la Conjuntiva/diagnóstico , Angiografía con Fluoresceína/métodos , Verde de Indocianina/farmacología , Melanoma/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Colorantes/farmacología , Conjuntiva/irrigación sanguínea , Neoplasias de la Conjuntiva/irrigación sanguínea , Femenino , Estudios de Seguimiento , Fondo de Ojo , Humanos , Masculino , Melanoma/irrigación sanguínea , Persona de Mediana Edad , Estudios Prospectivos , Tomografía de Coherencia Óptica/métodos
12.
Clin Neurol Neurosurg ; 202: 106537, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33571782

RESUMEN

OBJECTIVE: Perifocal edema of brain tumors is associated with survival and neurological symptoms. Our aim was to analyze associations between perifocal edema and immunohistochemical features including proliferation potential, microvessel density, neoangiogenesis and invasiveness in brain metastasis (BM). METHODS: 35 patients with BM were included into the retrospective study. The tumors were localized supratentorial in 25 lesions (71.4%) and infratentorial in 10 lesions (28.6%). The following immunohistochemical features were calculated on histopathological specimens: microvessel density, proliferation index Ki 67, matrix-metallopeptidase 9 (MMP9) extracellular matrix metalloproteinase inducer (EMMPRIN) and vascular endothelial growth factor (VEGF) expression. Tumor and edema volumes were estimated semiautomatically on magnetic resonance images. RESULTS: There were no correlations between tumor volume and edema volume. Moreover, no correlation was identified between the investigated immunohistochemical features and tumor/edema volume. In the non-small cell lung cancer subgroup, a positive correlation between tumor volume and VEGF expression was observed (r = 0.52, P = 0.02) and edema volume correlated inversely with MMP9 expression (r = -0.53, P = 0.02). CONCLUSION: In BM, no linear associations exist between tumor volumes, edema volumes and immunohistochemical features reflecting proliferation potential, neoangiogenesis, microvessel density and MMP9 expression. However, in the subgroup of non-small cell lung cancer, there might be associations between MMP9 expression and edema volume as well as between tumor volume and angiogenesis.


Asunto(s)
Edema Encefálico/diagnóstico por imagen , Neoplasias Encefálicas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/patología , Neovascularización Patológica/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Basigina/metabolismo , Neoplasias de los Conductos Biliares/patología , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/patología , Carcinoma/irrigación sanguínea , Carcinoma/diagnóstico por imagen , Carcinoma/metabolismo , Carcinoma/secundario , Carcinoma de Pulmón de Células no Pequeñas/irrigación sanguínea , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/secundario , Carcinoma de Células Transicionales/irrigación sanguínea , Carcinoma de Células Transicionales/diagnóstico por imagen , Carcinoma de Células Transicionales/metabolismo , Carcinoma de Células Transicionales/secundario , Proliferación Celular , Colangiocarcinoma/irrigación sanguínea , Colangiocarcinoma/diagnóstico por imagen , Colangiocarcinoma/metabolismo , Colangiocarcinoma/secundario , Neoplasias Colorrectales/patología , Neoplasias Endometriales/patología , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Imagen por Resonancia Magnética , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Melanoma/irrigación sanguínea , Melanoma/diagnóstico por imagen , Melanoma/metabolismo , Melanoma/secundario , Densidad Microvascular , Persona de Mediana Edad , Carcinoma Nasofaríngeo/irrigación sanguínea , Carcinoma Nasofaríngeo/diagnóstico por imagen , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/secundario , Neoplasias Nasofaríngeas/patología , Invasividad Neoplásica , Neoplasias Cutáneas/patología , Neoplasias Gástricas/patología , Carga Tumoral , Neoplasias Urológicas/patología
13.
Retina ; 41(6): 1182-1192, 2021 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-33315830

RESUMEN

PURPOSE: To evaluate the use of swept-source optical coherence tomography angiography to detect distinct vascular features in small choroidal melanomas and choroidal nevi. METHODS: Patients with a choroidal nevus or a treatment-naïve choroidal melanoma were imaged with color fundus photography, ultrasound, and swept-source optical coherence tomography angiography (12 × 12 mm). High-risk features including overlying fluid, orange pigment, shaggy photoreceptors, acoustic hollowness, depth >2 mm, and basal diameter >5 mm were assessed. Optical coherence tomography angiography vascular markers included: choroidal vessel visualization, choroidal vessel depth, and choriocapillaris flow signal, assessed qualitatively by comparison with surrounding, unaffected choriocapillaris. RESULTS: Twenty-nine lesions were included in this study, seven flat choroidal nevi, 17 elevated choroidal nevi, and 5 choroidal melanomas. Distinct vascular patterns were noted between flat nevi, elevated nevi, and small choroidal melanomas. Choroidal melanomas displayed two types of vasculature: "nevus-like" vasculature with straight parallel vessels and complex vasculature with vascular loops and crosslinking. Visualized choroidal vessels were significantly deeper in melanomas (110 µm) than elevated (84 µm) or flat nevi (70 µm). In a size-matched subanalysis of 5 elevated choroidal nevi and 5 choroidal melanomas, choroidal melanomas had increased mean choroidal vessel depth (P = 0.015), deepest choroidal vessel visualized (P = 0.034), and presence of a deep choroidal vessel >155 µm (P = 0.048). CONCLUSION: Swept-source optical coherence tomography angiography may detect distinct vascular features in choroidal nevi and small choroidal melanomas.


Asunto(s)
Neoplasias de la Coroides/diagnóstico , Coroides/diagnóstico por imagen , Angiografía con Fluoresceína/métodos , Melanoma/diagnóstico , Nevo Pigmentado/diagnóstico , Tomografía de Coherencia Óptica/métodos , Anciano , Coroides/irrigación sanguínea , Neoplasias de la Coroides/irrigación sanguínea , Estudios Transversales , Femenino , Estudios de Seguimiento , Fondo de Ojo , Humanos , Masculino , Melanoma/irrigación sanguínea , Persona de Mediana Edad , Reproducibilidad de los Resultados , Estudios Retrospectivos , Ultrasonografía
14.
J Invest Dermatol ; 141(4): 840-851.e4, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32890629

RESUMEN

Acquired BRAF/MAPK/extracellular signal‒regulated kinase inhibitor resistance in melanoma results in a new transcriptional state associated with an increased risk of metastasis. In this study, we identified noncanonical ephrin receptor (Eph) EphA2 signaling as a driver of the resistance-associated metastatic state. We used mass spectrometry‒based proteomic and phenotypic assays to demonstrate that the expression of active noncanonical EphA2-S897E in melanoma cells led to a mesenchymal-to-amoeboid transition driven by Cdc42 activation. The induction of mesenchymal-to-amoeboid transition promoted melanoma cell invasion, survival under shear stress, adhesion to endothelial cells under continuous-flow conditions, increased permeability of endothelial cell monolayers, and stimulated melanoma transendothelial cell migration. In vivo, melanoma cells expressing EphA2-S897E or active Cdc42 showed superior lung retention after tail-vain injection. Analysis of BRAF inhibitor‒sensitive and ‒resistant melanoma cells demonstrated resistance to be associated with a mesenchymal-to-amoeboid transition switch, upregulation of Cdc42 activity, increased invasion, and transendothelial migration. The drug-resistant metastatic state was dependent on histone deacetylase 8 activity. Silencing of histone deacetylase 8 led to the inhibition of EphA2 and protein kinase B phosphorylation, reduced invasion, and impaired melanoma cell-endothelial cell interactions. In summary, we have demonstrated that the metastatic state associated with acquired BRAF inhibitor resistance is dependent on noncanonical EphA2 signaling, leading to increased melanoma-endothelial cell interactions and enhanced tumor dissemination.


Asunto(s)
Resistencia a Antineoplásicos/genética , Melanoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Receptor EphA2/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Comunicación Celular/genética , Línea Celular Tumoral , Células Endoteliales/patología , Endotelio Vascular/citología , Endotelio Vascular/patología , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Melanoma/irrigación sanguínea , Melanoma/genética , Melanoma/patología , Ratones , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Receptor EphA2/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal/genética , Piel/irrigación sanguínea , Piel/patología , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Migración Transendotelial y Transepitelial/genética , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Sci Rep ; 10(1): 20493, 2020 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-33235311

RESUMEN

Angiogenesis is a physiological process for the formation of new blood vessels from the pre-existing vessels and it has a vital role in the survival and growth of neoplasms. During tumor angiogenesis, the activation of the gene transcriptions in vascular endothelial cells (ECs) plays an essential role in the promotion of EC proliferation, migration, and vascular network development. However, the molecular mechanisms underlying transcriptional regulation of EC and tumor angiogenesis remains to be fully elucidated. Here we report that the transcription factor Yin Yang 1 (YY1) in ECs is critically involved in tumor angiogenesis. First, we utilized a tamoxifen-inducible EC-specific YY1 deficient mouse model and showed that YY1 deletion in ECs inhibited the tumor growth and tumor angiogenesis. Using the in vivo matrigel plug assay, we then found that EC-specific YY1 ablation inhibited growth factor-induced angiogenesis. Furthermore, vascular endothelial growth factor (VEGF)-induced EC migration was diminished in YY1-depleted human umbilical vein endothelial cells (HUVECs). Finally, a rescue experiment revealed that YY1-regulated BMP6 expression in ECs was involved in EC migration. Collectively, our results demonstrate that endothelial YY1 has a crucial role in tumor angiogenesis and suggest that targeting endothelial YY1 could be a potential therapeutic strategy for cancer treatment.


Asunto(s)
Células Endoteliales/metabolismo , Melanoma/irrigación sanguínea , Melanoma/patología , Neovascularización Patológica/metabolismo , Factor de Transcripción YY1/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Colágeno/metabolismo , Combinación de Medicamentos , Células Endoteliales/patología , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Laminina/metabolismo , Melanoma/genética , Ratones Noqueados , Neovascularización Patológica/patología , Proteoglicanos/metabolismo , ARN Interferente Pequeño/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de Transcripción YY1/genética
16.
Exp Dermatol ; 29(11): 1046-1054, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32998178

RESUMEN

Metastatic melanoma is a devastating disease with a marginal-albeit increasing-hope for cure. Melanoma has a high mutation rate which correlates to the expression of numerous neo-antigens and thus is associated with the potential to induce and strengthen effective antitumoral immunity. However, the incomplete and potentially insufficient response to established immunotherapies (response rates usually do not markedly exceed 60%) already points to the need of further studies to improve treatment strategies. Multiple tumor escape mechanisms that allow melanoma to evade from antitumoral immune responses have been characterized and must be overcome to achieve a better clinical efficacy of immunotherapies. Recently, promising progress has been made in targeting tumor vasculature to control and increase the infiltration of tumors with effector lymphocytes. It has been hypothesized that amplified lymphocytic infiltrates in melanoma metastases result in a switch of the tumor microenvironment from a non-inflammatory to an inflammatory state. In this view point essay, we discuss the requirements for successful homing of lymphocytes to melanoma tissue and we present a mouse melanoma xenograft model that allows the investigation of human tumor vessels in vivo. Furthermore, current clinical studies dealing with the activation of melanoma vasculature for enhanced effectiveness of immunotherapy protocols are presented and open questions for routine clinical application are addressed.


Asunto(s)
Endotelio/inmunología , Inmunoterapia , Linfocitos/inmunología , Melanoma/terapia , Neoplasias Cutáneas/terapia , Animales , Vasos Sanguíneos/inmunología , Movimiento Celular , Modelos Animales de Enfermedad , Humanos , Melanoma/irrigación sanguínea , Melanoma/inmunología , Melanoma/secundario , Ratones , Receptores Mensajeros de Linfocitos , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Escape del Tumor/inmunología
17.
Sci Rep ; 10(1): 11372, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32647211

RESUMEN

Administration of hemoglobin-based oxygen carriers (HBOCs) into the systemic circulation is a potential strategy to relieve solid tumor hypoxia in order to increase the effectiveness of chemotherapeutics. Previous computational analysis indicated that the oxygen (O2) status of the tumor and HBOC O2 affinity may play a role in increased O2 delivery to the tumor. However, no study has experimentally investigated how low- and high-affinity HBOCs would perform in normoxic and hypoxic tumors. In this study, we examined how the HBOC, polymerized human hemoglobin (PolyhHb), in the relaxed (R) or tense (T) quaternary state modulates O2 delivery to hypoxic (FME) and normoxic (LOX) human melanoma xenografts in a murine window chamber model. We examined microcirculatory fluid flow via video shearing optical microscopy, and O2 distributions via phosphorescence quenching microscopy. Additionally, we examined how weekly infusion of a 20% top-load dose of PolyhHb influences growth rate, vascularization, and regional blood flow in the FME and LOX tumor xenografts. Infusion of low-affinity T-state PolyhHb led to increased tissue oxygenation, decreased blood flow, decreased tumor growth, and decreased vascularization in hypoxic tumors. However, infusion of both T-state and R-state PolyhHbs led to worse outcomes in normoxic tumors. Of particular concern was the high-affinity R-state PolyhHb, which led to no improvement in hypoxic tumors and significantly worsened outcomes in normoxic tumors. Taken together, the results of this study indicate that the tumor O2 status is a primary determinant of the potency and outcomes of infused PolyhHb.


Asunto(s)
Hemoglobinas/farmacología , Melanoma/tratamiento farmacológico , Oxígeno/metabolismo , Polímeros/farmacología , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Hipoxia de la Célula/efectos de los fármacos , Eritrocitos/química , Femenino , Hemoglobinas/química , Hemoglobinas/aislamiento & purificación , Hemoglobinas/uso terapéutico , Humanos , Infusiones Intravenosas , Melanoma/irrigación sanguínea , Melanoma/patología , Ratones , Microcirculación/efectos de los fármacos , Peso Molecular , Oxígeno/análisis , Polimerizacion , Polímeros/química , Polímeros/uso terapéutico , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancer Res ; 80(16): 3345-3358, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32586981

RESUMEN

Platelet-derived growth factor B (PDGFB) plays a crucial role in recruitment of PDGF receptor ß-positive pericytes to blood vessels. The endothelium is an essential source of PDGFB in this process. Platelets constitute a major reservoir of PDGFB and are continuously activated in the tumor microenvironment, exposing tumors to the plethora of growth factors contained in platelet granules. Here, we show that tumor vascular function, as well as pericyte coverage is significantly impaired in mice with conditional knockout of PDGFB in platelets. A lack of PDGFB in platelets led to enhanced hypoxia and epithelial-to-mesenchymal transition in the primary tumors, elevated levels of circulating tumor cells, and increased spontaneous metastasis to the liver or lungs in two mouse models. These findings establish a previously unknown role for platelet-derived PDGFB, whereby it promotes and maintains vascular integrity in the tumor microenvironment by contributing to the recruitment of pericytes. SIGNIFICANCE: Conditional knockout of PDGFB in platelets demonstrates its previously unknown role in the maintenance of tumor vascular integrity and host protection against metastasis.


Asunto(s)
Movimiento Celular , Endotelio Vascular/metabolismo , Pericitos/fisiología , Proteínas Proto-Oncogénicas c-sis/fisiología , Animales , Vasos Sanguíneos , Neoplasias del Colon/irrigación sanguínea , Transición Epitelial-Mesenquimal , Matriz Extracelular , Técnicas de Inactivación de Genes , Hibridación Genética , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/secundario , Melanoma/irrigación sanguínea , Melanoma/secundario , Ratones , Células Neoplásicas Circulantes , Neoplasias Pancreáticas , Pericitos/metabolismo , Activación Plaquetaria/fisiología , Proteínas Proto-Oncogénicas c-sis/deficiencia , Proteínas Proto-Oncogénicas c-sis/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Trombocitopenia , Hipoxia Tumoral , Microambiente Tumoral
19.
Nat Commun ; 11(1): 2810, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32499572

RESUMEN

The overexpression of the protein tyrosine kinase, Focal adhesion kinase (FAK), in endothelial cells has implicated its requirement in angiogenesis and tumour growth, but how pericyte FAK regulates tumour angiogenesis is unknown. We show that pericyte FAK regulates tumour growth and angiogenesis in multiple mouse models of melanoma, lung carcinoma and pancreatic B-cell insulinoma and provide evidence that loss of pericyte FAK enhances Gas6-stimulated phosphorylation of the receptor tyrosine kinase, Axl with an upregulation of Cyr61, driving enhanced tumour growth. We further show that pericyte derived Cyr61 instructs tumour cells to elevate expression of the proangiogenic/protumourigenic transmembrane receptor Tissue Factor. Finally, in human melanoma we show that when 50% or more tumour blood vessels are pericyte-FAK negative, melanoma patients are stratified into those with increased tumour size, enhanced blood vessel density and metastasis. Overall our data uncover a previously unknown mechanism of tumour growth by pericytes that is controlled by pericyte FAK.


Asunto(s)
Proteína 61 Rica en Cisteína/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neovascularización Patológica , Pericitos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Aorta Torácica/patología , Carcinoma Pulmonar de Lewis/metabolismo , Adhesión Celular , Proliferación Celular , Femenino , Quinasa 1 de Adhesión Focal/genética , Humanos , Linfocinas/metabolismo , Masculino , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/patología , Factor de Crecimiento Placentario/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Transducción de Señal , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Tirosina Quinasa del Receptor Axl
20.
Biomed Pharmacother ; 127: 110135, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32334374

RESUMEN

Melanoma is the most aggressive form of skin cancer. Malignant melanoma in particular has a poor prognosis and although treatment has improved, drug resistance continues to be a challenge. Angiogenesis, the formation of blood vessels from existing microvessels, precedes the progression of melanoma from a radial growth phase to a malignant phenotype. In addition, melanoma cells can form networks of vessel-like fluid conducting channels through vasculogenic mimicry (VM). Both angiogenesis and VM have been postulated to contribute to the development of resistance to treatment and to enable metastasis. Also, the metastatic spread of melanoma is highly dependent on lymphangiogenesis, the formation of lymphatic vessels from pre-existing vessels. Interestingly, the design and clinical testing of drugs that target VM and lymphangiogenesis lag behind that of angiogenesis inhibitors. Despite this, antiangiogenic drugs have not significantly improved the overall survival of melanoma patients, thus necessitating the targeting of alternative mechanisms. In this article, I review the roles of the three paradigms of tissue perfusion, namely, angiogenesis, VM and lymphangiogenesis, in promoting melanoma progression and metastasis. This article also explores the latest development and potential opportunities in the therapeutic targeting of these processes.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Melanoma/irrigación sanguínea , Neovascularización Patológica/etiología , Humanos , Linfangiogénesis , Melanoma/tratamiento farmacológico , Melanoma/secundario
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA