Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 880
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(33): e2122716119, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35960843

RESUMO

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.


Assuntos
Complexo de Ataque à Membrana do Sistema Complemento , Endotélio Vascular , Armadilhas Extracelulares , Melanoma , Neutrófilos , Microambiente Tumoral , Animais , Complexo de Ataque à Membrana do Sistema Complemento/imunologia , Proteínas do Sistema Complemento , Endotélio Vascular/fisiopatologia , Humanos , Melanoma/irrigação sanguínea , Melanoma/imunologia , Melanoma/patologia , Camundongos , Neutrófilos/imunologia , Permeabilidade
2.
Adv Sci (Weinh) ; 9(10): e2105226, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35088579

RESUMO

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.


Assuntos
Subunidade Apc6 do Ciclossomo-Complexo Promotor de Anáfase , Proteínas de Ligação ao Cálcio , Proteínas Ativadoras de GTPase , Melanoma , Ubiquitina , Subunidade Apc6 do Ciclossomo-Complexo Promotor de Anáfase/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Melanoma/irrigação sanguínea , Ubiquitina/metabolismo
3.
Microvasc Res ; 140: 104305, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34958805

RESUMO

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.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Endoteliais/metabolismo , Melanoma/irrigação sanguínea , Melanoma/metabolismo , Proteínas Musculares/metabolismo , Neovascularização Patológica , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/genética , Caderinas/metabolismo , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteínas de Ligação a DNA/genética , Células Endoteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Melanoma/genética , Melanoma/patologia , Camundongos Nus , Proteínas Musculares/genética , Invasividade Neoplásica , Isoformas de Proteínas , Transdução de Sinais , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Carga Tumoral
4.
J Invest Dermatol ; 142(7): 1923-1933.e5, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34968503

RESUMO

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.


Assuntos
Metaloproteinase 14 da Matriz , Melanoma , Animais , Permeabilidade Capilar , Células Endoteliais/metabolismo , Humanos , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Melanoma/irrigação sanguínea , Melanoma/patologia , Camundongos , Metástase Neoplásica , Óxido Nítrico Sintase Tipo III/metabolismo
5.
Cells ; 10(10)2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34685720

RESUMO

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.


Assuntos
Vesículas Extracelulares/metabolismo , Melanoma/metabolismo , Neoplasias Cutâneas/metabolismo , Animais , Progressão da Doença , Resistencia a Medicamentos Antineoplásicos , Humanos , Melanoma/irrigação sanguínea , Melanoma/patologia , Melanoma/terapia , Modelos Biológicos , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/terapia , Microambiente Tumoral , Melanoma Maligno Cutâneo
6.
Int J Mol Sci ; 22(5)2021 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-33803452

RESUMO

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.


Assuntos
Apoptose , Resistencia a Medicamentos Antineoplásicos , Melanoma/irrigação sanguínea , Melanoma/metabolismo , Neovascularização Patológica/tratamento farmacológico , Proteína bcl-X/metabolismo , Humanos , Melanoma/tratamento farmacológico , Melanoma/patologia , MicroRNAs/metabolismo , Invasividade Neoplásica , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , RNA Neoplásico/metabolismo
7.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-33800878

RESUMO

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.


Assuntos
Linfócitos do Interstício Tumoral/patologia , Melanoma/patologia , Neovascularização Patológica/fisiopatologia , Neoplasias Uveais/patologia , Idoso , Animais , Apresentação de Antígeno , Análise por Conglomerados , Conjuntos de Dados como Assunto , Intervalo Livre de Doença , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoterapia , Estimativa de Kaplan-Meier , Subpopulações de Linfócitos/patologia , Macrófagos/patologia , Masculino , Melanoma/irrigação sanguínea , Melanoma/genética , Melanoma/imunologia , Redes e Vias Metabólicas , Pessoa de Meia-Idade , Neovascularização Patológica/genética , Prognóstico , Transdução de Sinais , Células Estromais/patologia , Microambiente Tumoral , Neoplasias Uveais/irrigação sanguínea , Neoplasias Uveais/genética , Neoplasias Uveais/imunologia
8.
Clin Neurol Neurosurg ; 202: 106537, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33571782

RESUMO

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.


Assuntos
Edema Encefálico/diagnóstico por imagem , Neoplasias Encefálicas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/patologia , Neovascularização Patológica/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Basigina/metabolismo , Neoplasias dos Ductos Biliares/patologia , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Carcinoma/irrigação sanguínea , Carcinoma/diagnóstico por imagem , Carcinoma/metabolismo , Carcinoma/secundário , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/secundário , Carcinoma de Células de Transição/irrigação sanguínea , Carcinoma de Células de Transição/diagnóstico por imagem , Carcinoma de Células de Transição/metabolismo , Carcinoma de Células de Transição/secundário , Proliferação de Células , Colangiocarcinoma/irrigação sanguínea , Colangiocarcinoma/diagnóstico por imagem , Colangiocarcinoma/metabolismo , Colangiocarcinoma/secundário , Neoplasias Colorretais/patologia , Neoplasias do Endométrio/patologia , Feminino , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Imageamento por Ressonância Magnética , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Melanoma/irrigação sanguínea , Melanoma/diagnóstico por imagem , Melanoma/metabolismo , Melanoma/secundário , Densidade Microvascular , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/irrigação sanguínea , Carcinoma Nasofaríngeo/diagnóstico por imagem , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/secundário , Neoplasias Nasofaríngeas/patologia , Invasividade Neoplásica , Neoplasias Cutâneas/patologia , Neoplasias Gástricas/patologia , Carga Tumoral , Neoplasias Urológicas/patologia
9.
Cornea ; 40(12): 1519-1524, 2021 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-33591034

RESUMO

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.


Assuntos
Túnica Conjuntiva/patologia , Neoplasias da Túnica Conjuntiva/diagnóstico , Angiofluoresceinografia/métodos , Verde de Indocianina/farmacologia , Melanoma/diagnóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Corantes/farmacologia , Túnica Conjuntiva/irrigação sanguínea , Neoplasias da Túnica Conjuntiva/irrigação sanguínea , Feminino , Seguimentos , Fundo de Olho , Humanos , Masculino , Melanoma/irrigação sanguínea , Pessoa de Meia-Idade , Estudos Prospectivos , Tomografia de Coerência Óptica/métodos
10.
Retina ; 41(6): 1182-1192, 2021 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-33315830

RESUMO

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.


Assuntos
Neoplasias da Coroide/diagnóstico , Corioide/diagnóstico por imagem , Angiofluoresceinografia/métodos , Melanoma/diagnóstico , Nevo Pigmentado/diagnóstico , Tomografia de Coerência Óptica/métodos , Idoso , Corioide/irrigação sanguínea , Neoplasias da Coroide/irrigação sanguínea , Estudos Transversais , Feminino , Seguimentos , Fundo de Olho , Humanos , Masculino , Melanoma/irrigação sanguínea , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Estudos Retrospectivos , Ultrassonografia
11.
J Invest Dermatol ; 141(4): 840-851.e4, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32890629

RESUMO

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.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Receptor EphA2/metabolismo , Neoplasias Cutâneas/tratamento farmacológico , Animais , Comunicação Celular/genética , Linhagem Celular Tumoral , Células Endoteliais/patologia , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Melanoma/irrigação sanguínea , Melanoma/genética , Melanoma/patologia , Camundongos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Receptor EphA2/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Pele/irrigação sanguínea , Pele/patologia , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Migração Transendotelial e Transepitelial/genética , Microambiente Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Sci Rep ; 10(1): 20493, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33235311

RESUMO

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.


Assuntos
Células Endoteliais/metabolismo , Melanoma/irrigação sanguínea , Melanoma/patologia , Neovascularização Patológica/metabolismo , Fator de Transcrição YY1/metabolismo , Animais , Movimento Celular , Proliferação de Células , Colágeno/metabolismo , Combinação de Medicamentos , Células Endoteliais/patologia , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Laminina/metabolismo , Melanoma/genética , Camundongos Knockout , Neovascularização Patológica/patologia , Proteoglicanas/metabolismo , RNA Interferente Pequeno/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator de Transcrição YY1/genética
13.
Exp Dermatol ; 29(11): 1046-1054, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32998178

RESUMO

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.


Assuntos
Endotélio/imunologia , Imunoterapia , Linfócitos/imunologia , Melanoma/terapia , Neoplasias Cutâneas/terapia , Animais , Vasos Sanguíneos/imunologia , Movimento Celular , Modelos Animais de Doenças , Humanos , Melanoma/irrigação sanguínea , Melanoma/imunologia , Melanoma/secundário , Camundongos , Receptores de Retorno de Linfócitos , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/patologia , Evasão Tumoral/imunologia
14.
Sci Rep ; 10(1): 11372, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647211

RESUMO

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.


Assuntos
Hemoglobinas/farmacologia , Melanoma/tratamento farmacológico , Oxigênio/metabolismo , Polímeros/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Animais , Hipóxia Celular/efeitos dos fármacos , Eritrócitos/química , Feminino , Hemoglobinas/química , Hemoglobinas/isolamento & purificação , Hemoglobinas/uso terapêutico , Humanos , Infusões Intravenosas , Melanoma/irrigação sanguínea , Melanoma/patologia , Camundongos , Microcirculação/efeitos dos fármacos , Peso Molecular , Oxigênio/análise , Polimerização , Polímeros/química , Polímeros/uso terapêutico , Neoplasias Cutâneas/irrigação sanguínea , Neoplasias Cutâneas/patologia , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Nat Commun ; 11(1): 2810, 2020 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-32499572

RESUMO

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.


Assuntos
Proteína Rica em Cisteína 61/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Patológica , Pericitos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Aorta Torácica/patologia , Carcinoma Pulmonar de Lewis/metabolismo , Adesão Celular , Proliferação de Células , Feminino , Quinase 1 de Adesão Focal/genética , Humanos , Linfocinas/metabolismo , Masculino , Melanoma/irrigação sanguínea , Melanoma/metabolismo , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/patologia , Fator de Crescimento Placentário/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis/metabolismo , Transdução de Sinais , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor Tirosina Quinase Axl
16.
Cancer Res ; 80(16): 3345-3358, 2020 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-32586981

RESUMO

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.


Assuntos
Movimento Celular , Endotélio Vascular/metabolismo , Pericitos/fisiologia , Proteínas Proto-Oncogênicas c-sis/fisiologia , Animais , Vasos Sanguíneos , Neoplasias do Colo/irrigação sanguínea , Transição Epitelial-Mesenquimal , Matriz Extracelular , Técnicas de Inativação de Genes , Hibridização Genética , Neoplasias Hepáticas/secundário , Neoplasias Pulmonares/secundário , Melanoma/irrigação sanguínea , Melanoma/secundário , Camundongos , Células Neoplásicas Circulantes , Neoplasias Pancreáticas , Pericitos/metabolismo , Ativação Plaquetária/fisiologia , Proteínas Proto-Oncogênicas c-sis/deficiência , Proteínas Proto-Oncogênicas c-sis/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Trombocitopenia , Hipóxia Tumoral , Microambiente Tumoral
17.
Biomed Pharmacother ; 127: 110135, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32334374

RESUMO

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.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Melanoma/irrigação sanguínea , Neovascularização Patológica/etiologia , Humanos , Linfangiogênese , Melanoma/tratamento farmacológico , Melanoma/secundário
19.
Matrix Biol ; 87: 11-25, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31422156

RESUMO

Multimerin-2 is an extracellular matrix glycoprotein and member of the elastin microfibril interface-located (EMILIN) family of proteins. Multimerin-2 is deposited along blood vessels and we previously demonstrated that it regulates the VEGFA/VEGFR2 signaling axis and angiogenesis. However, its role in modulating vascular homeostasis remains largely unexplored. Here we identified Multimerin-2 as a key molecule required to maintain vascular stability. RNAi knockdown of Multimerin-2 in endothelial cells led to cell-cell junctional instability and increased permeability. Mechanistically cell-cell junction dismantlement occurred through the phosphorylation of VEGFR2 at Tyr951, activation of Src and phosphorylation of VE-cadherin. To provide an in vivo validation for these in vitro effects, we generated Multimerin-2-/- (Mmrn2-/-) mice. Although Mmrn2-/- mice developed normally and displayed no gross abnormalities, endothelial cells displayed cell junctional defects associated with increased levels of VEGFR2 phospho-Tyr949 (the murine counterpart of human Tyr951), impaired pericyte recruitment and increased vascular leakage. Of note, tumor associated vessels were defective in Mmrn2-/- mice, with increased number of small and often collapsed vessels, concurrent with a significant depletion of pericytic coverage. Consequently, the Mmrn2-/- vessels were less perfused and leakier, leading to increased tumor hypoxia. Chemotherapy efficacy was markedly impaired in Mmrn2-/- mice and this was associated with poor drug delivery to the tumor xenografts. Collectively, our findings demonstrate that Multimerin-2 is required for proper vessel homeostasis and stabilization, and unveil the possibility to utilize expression levels of this glycoprotein in predicting chemotherapy efficacy.


Assuntos
Antígenos CD/metabolismo , Antígenos de Superfície/genética , Caderinas/metabolismo , Proteínas da Matriz Extracelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Melanoma/irrigação sanguínea , Glicoproteínas de Membrana/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Antígenos de Superfície/metabolismo , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Cisplatino/farmacologia , Tratamento Farmacológico , Proteínas da Matriz Extracelular/metabolismo , Técnicas de Inativação de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Junções Intercelulares , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Transplante de Neoplasias , Fosforilação , Hipóxia Tumoral/efeitos dos fármacos
20.
Eur J Ophthalmol ; 30(4): 723-729, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31210061

RESUMO

PURPOSE: To describe the imaging features of choroidal nevus and melanoma using optical coherence tomography angiography, and evaluate the ability of this technique to establish the differential diagnosis based on the display of the tumor's intrinsic vasculature. METHODS: Comparative analysis of optical coherence tomography angiography findings in consecutive patients diagnosed with choroidal nevus or choroidal melanoma following a complete ophthalmic evaluation, including best-corrected visual acuity and several imaging studies: color fundus photography, B-scan ultrasound, spectral-domain optical coherence tomography, and optical coherence tomography angiography. Optical coherence tomography angiography was used to investigate qualitative differences in the tumor vasculature. RESULTS: Thirty-six eyes (18 cases of choroidal nevus and 18 cases of choroidal melanoma) from 36 consecutive patients were included in the study. Only cases located posterior to equator were included to enable performance of all tests. On optical coherence tomography angiography, choroidal nevus showed well-delimited margins (78%), hyperreflective choroid capillary vasculature (83%), fewer avascular areas (17%), and neovascular membrane in one case (6%). Choroidal melanoma showed imprecise margins (72%), hyporeflective choroidal capillary vasculature (72%), multiple avascular areas (78%), and choroidal vascular changes (e.g. thick vascular networks or vascular loops; 45%). CONCLUSION: Optical coherence tomography angiography can provide useful information for assessing and differentiating between choroidal nevi and small melanomas. Significant differences between these conditions were found for the pattern of reflectivity, and presence/absence of avascular zones and vascular anomalies, which could be helpful for supporting the diagnosis.


Assuntos
Doenças da Coroide/diagnóstico , Neoplasias da Coroide/diagnóstico , Melanoma/diagnóstico , Nevo Pigmentado/diagnóstico , Adulto , Idoso , Neoplasias da Coroide/irrigação sanguínea , Diagnóstico Diferencial , Feminino , Angiofluoresceinografia/métodos , Humanos , Masculino , Melanoma/irrigação sanguínea , Pessoa de Meia-Idade , Nevo Pigmentado/irrigação sanguínea , Estudos Retrospectivos , Tomografia de Coerência Óptica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA