38 research outputs found

    Molecular structure and function of biological barriers

    Get PDF
    Biological barriers are indispensable for the integrity and function of many vertebrate organs. The barrier function is based on intercellular protein complexes of the plasma membrane which form paracellular diffusion barriers and separate internal and external fluid compartments, an indispensable prerequisite for every organ development and function. The review summarizes key characteristics and molecular structure of intercellular junctions (tight junctions and adherens junctions) responsible for cellular barrier formation. One of the most important such cellular barriers is the blood-brain barrier (BBB) which forms an active interface between the circulation and neural tissue. Its principal cellular components are cerebral endothelial cells, pericytes and astrocytes, whose finely tuned interactions are needed for a proper function. The review highlights the most important functions of the BBB including some novel regulatory aspects as well

    CB2 Receptor Activation Inhibits Melanoma Cell Transmigration through the Blood-Brain Barrier

    Get PDF
    During parenchymal brain metastasis formation tumor cells need to migrate through cerebral endothelial cells, which form the morphological basis of the blood-brain barrier (BBB). The mechanisms of extravasation of tumor cells are highly uncharacterized, but in some aspects recapitulate the diapedesis of leukocytes. Extravasation of leukocytes through the BBB is decreased by the activation of type 2 cannabinoid receptors (CB2); therefore, in the present study we sought to investigate the role of CB2 receptors in the interaction of melanoma cells with the brain endothelium. First, we identified the presence of CB1, CB2(A), GPR18 (transcriptional variant 1) and GPR55 receptors in brain endothelial cells, while melanoma cells expressed CB1, CB2(A), GPR18 (transcriptional variants 1 and 2), GPR55 and GPR119. We observed that activation of CB2 receptors with JWH-133 reduced the adhesion of melanoma cells to the layer of brain endothelial cells. JWH-133 decreased the transendothelial migration rate of melanoma cells as well. Our results suggest that changes induced in endothelial cells are critical in the mediation of the effect of CB2 agonists. Our data identify CB2 as a potential target in reducing the number of brain metastastes originating from melanoma

    Downregulation of circulating miR 802-5p and miR 194-5p and upregulation of brain MEF2C along breast cancer brain metastasization

    Get PDF
    Breast cancer brain metastases (BCBM) have been under-investigated despite their high incidence and poor outcome. Micro RNAs (miRNAs), and particularly circulating miRNAs, regulate multiple cellular functions and their deregulation has been reported in different types of cancer and metastasis. However, their signature in plasma along brain metastasis development and their relevant targets remain undetermined. Here, we used a mouse model of BCBM and Next-Generation Sequencing (NGS) to establish the alterations in circulating miRNAs during brain metastasis formation and development. We further performed bioinformatics analysis to identify their targets with relevance in the metastatic process. We additionally analyzed human resected brain metastasis samples of breast cancer patients for target expression validation. Breast cancer cells were injected in the carotid artery of mice to preferentially induce metastasis in the brain, and samples were collected at different time points (5 h, 3, 7 and 10 days) to follow metastasis development. Metastases were detected from 7 days onwards, mainly in the brain. NGS revealed a deregulation of circulating miRNA profile during BCBM progression, raising from 18% at 3 days to 30% at 10 days following malignant cells' injection. Work was focused on those altered prior to metastasis detection, among which were miR-802-5p and miR-194-5p, whose downregulation was validated by qPCR. Using TargetScan and DIANA Tools the transcription factor myocyte enhancer factor 2C (MEF2C) was identified as a target for both miRNAs, and its expression was increasingly observed in malignant cells along brain metastasis development. Its upregulation was also observed in peritumoral astrocytes pointing to a role of MEF2C in the crosstalk between tumor cells and astrocytes. MEF2C expression was also observed in human BCBM, validating the observation in mouse. Collectively, downregulation of circulating miR-802-5p and miR-194-5p appears as a precocious event in BCBM and MEF2C emerges as a new player in brain metastasis development

    Picturing Breast Cancer Brain Metastasis Development to Unravel Molecular Players and Cellular Crosstalk

    Get PDF
    Simple Summary Breast cancer is a devastating disorder affecting millions of women worldwide. With improved therapeutics for the primary tumor, the appearance of metastasis has been increasing. Breast cancer frequently metastasizes to the brain, constituting a major hurdle without cure and with a poor survival. It is imperative to better understand the mechanisms involved in malignant cell transposition of the brain microvasculature and parenchymal colonization by deciphering the alterations occurring in the tumor and microvascular cells, as well as the occurrence of intercellular communication during the process. We aimed to profile the process of the formation of breast cancer brain metastasis and the timeline of events governing it. We used a specific mouse model of the disease to perform extensive microscopic analyses. We identified phenotypic changes and the activation of relevant molecular players in tumorigenesis, together with vascular alterations, and the occurrence of crosstalk. Our findings unravel putative therapeutic targets to tackle breast cancer brain metastasis. With breast cancer (BC) therapy improvements, the appearance of brain metastases has been increasing, representing a life-threatening condition. Brain metastasis formation involves BC cell (BCC) extravasation across the blood-brain barrier (BBB) and brain colonization by unclear mechanisms. We aimed to disclose the actors involved in BC brain metastasis formation, focusing on BCCs' phenotype, growth factor expression, and signaling pathway activation, correlating with BBB alterations and intercellular communication. Hippocampi of female mice inoculated with 4T1 BCCs were examined over time by hematoxylin-eosin, immunohistochemistry and immunofluorescence. Well-established metastases were observed at seven days, increasing thereafter. BCCs entering brain parenchyma presented mesenchymal, migratory, and proliferative features; however, with time, they increasingly expressed epithelial markers, reflecting a mesenchymal-epithelial transition. BCCs also expressed platelet-derived growth factor-B, beta(4) integrin, and focal adhesion kinase, suggesting autocrine and/or paracrine regulation with adhesion signaling activation, while balance between Rac1 and RhoA was associated with the motility status. Intercellular communication via gap junctions was clear among BCCs, and between BCCs and endothelial cells. Thrombin accumulation, junctional protein impairment, and vesicular proteins increase reflect BBB alterations related with extravasation. Expression of plasmalemma vesicle-associated protein was increased in BCCs, along with augmented vascularization, whereas pericyte contraction indicated mural cells' activation. Our results provide further understanding of BC brain metastasis formation, disclosing potential therapeutic targets

    Paracellular and transcellular migration of metastatic cells through the cerebral endothelium

    Get PDF
    Breast cancer and melanoma are among the most frequent cancer types leading to brain metastases. Despite the unquestionable clinical significance, important aspects of the development of secondary tumours of the central nervous system are largely uncharacterized, including extravasation of metastatic cells through the blood-brain barrier. By using transmission electron microscopy, here we followed interactions of cancer cells and brain endothelial cells during the adhesion, intercalation/incorporation and transendothelial migration steps. We observed that brain endothelial cells were actively involved in the initial phases of the extravasation by extending filopodia-like membrane protrusions towards the tumour cells. Melanoma cells tended to intercalate between endothelial cells and to transmigrate by utilizing the paracellular route. On the other hand, breast cancer cells were frequently incorporated into the endothelium and were able to migrate through the transcellular way from the apical to the basolateral side of brain endothelial cells. When co-culturing melanoma cells with cerebral endothelial cells, we observed N-cadherin enrichment at melanoma-melanoma and melanoma-endothelial cell borders. However, for breast cancer cells N-cadherin proved to be dispensable for the transendothelial migration both in vitro and in vivo. Our results indicate that breast cancer cells are more effective in the transcellular type of migration than melanoma cells
    corecore