Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Nat Commun ; 13(1): 3620, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35750885

ABSTRACT

Pigs are valuable large animal models for biomedical and genetic research, but insights into the tissue- and cell-type-specific transcriptome and heterogeneity remain limited. By leveraging single-cell RNA sequencing, we generate a multiple-organ single-cell transcriptomic map containing over 200,000 pig cells from 20 tissues/organs. We comprehensively characterize the heterogeneity of cells in tissues and identify 234 cell clusters, representing 58 major cell types. In-depth integrative analysis of endothelial cells reveals a high degree of heterogeneity. We identify several functionally distinct endothelial cell phenotypes, including an endothelial to mesenchymal transition subtype in adipose tissues. Intercellular communication analysis predicts tissue- and cell type-specific crosstalk between endothelial cells and other cell types through the VEGF, PDGF, TGF-ß, and BMP pathways. Regulon analysis of single-cell transcriptome of microglia in pig and 12 other species further identifies MEF2C as an evolutionally conserved regulon in the microglia. Our work describes the landscape of single-cell transcriptomes within diverse pig organs and identifies the heterogeneity of endothelial cells and evolutionally conserved regulon in microglia.


Subject(s)
Endothelial Cells , Microglia , Animals , Microglia/metabolism , Phenotype , Regulon/genetics , Single-Cell Analysis , Swine , Transcriptome
3.
Biomolecules ; 13(1)2022 12 22.
Article in English | MEDLINE | ID: mdl-36671408

ABSTRACT

Primary endothelial cells (ECs), especially human umbilical vein endothelial cells (HUVECs), are broadly used in vascular biology. Gene editing of primary endothelial cells is known to be challenging, due to the low DNA transfection efficiency and the limited proliferation capacity of ECs. We report the establishment of a highly efficient and selection-free CRISPR gene editing approach for primary endothelial cells (HUVECs) with ribonucleoprotein (RNP) complex. We first optimized an efficient and cost-effective protocol for messenger RNA (mRNA) delivery into primary HUVECs by nucleofection. Nearly 100% transfection efficiency of HUVECs was achieved with EGFP mRNA. Using this optimized DNA-free approach, we tested RNP-mediated CRISPR gene editing of primary HUVECs with three different gRNAs targeting the HIF1A gene. We achieved highly efficient (98%) and biallelic HIF1A knockout in HUVECs without selection. The effects of HIF1A knockout on ECs' angiogenic characteristics and response to hypoxia were validated by functional assays. Our work provides a simple method for highly efficient gene editing of primary endothelial cells (HUVECs) in studies and manipulations of ECs functions.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Humans , Gene Editing/methods , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , DNA , Human Umbilical Vein Endothelial Cells/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
4.
Int J Mol Sci ; 22(10)2021 May 14.
Article in English | MEDLINE | ID: mdl-34069135

ABSTRACT

Triple negative breast cancer presents higher mortality and poorer survival rates than other breast cancer (BC) types, due to the proneness to brain metastases formation, which are usually diagnosed at advanced stages. Therefore, the discovery of BC brain metastases (BCBM) biomarkers appears pivotal for a timely intervention. With this work, we aimed to disclose microRNAs (miRNAs) and extracellular vesicles (EVs) in the circulation as biomarkers of BCBM formation. Using a BCBM animal model, we analyzed EVs in plasma by nanoparticle tracking analysis and ascertained their blood-brain barrier (BBB) origin by flow cytometry. We further evaluated circulating miRNAs by RT-qPCR and their brain expression by in situ hybridization. In parallel, a cellular model of BCBM formation, combining triple negative BC cells and BBB endothelial cells, was used to differentiate the origin of biomarkers. Established metastases were associated with an increased content of circulating EVs, particularly of BBB origin. Interestingly, deregulated miRNAs in the circulation were observed prior to BCBM detection, and their brain origin was suggested by matching alterations in brain parenchyma. In vitro studies indicated that miR-194-5p and miR-205-5p are expressed and released by BC cells, endothelial cells and during their interaction. These results highlight miRNAs and EVs as biomarkers of BCBM in early and advanced stages, respectively.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Circulating MicroRNA/blood , Extracellular Vesicles/pathology , Animals , Blood-Brain Barrier , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Cell Line, Tumor , Circulating MicroRNA/genetics , Endothelium, Vascular/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , MicroRNAs/genetics , Xenograft Model Antitumor Assays
5.
Cancers (Basel) ; 13(4)2021 Feb 22.
Article in English | MEDLINE | ID: mdl-33671551

ABSTRACT

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, ß4 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.

6.
Sci Rep ; 10(1): 12237, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32699247

ABSTRACT

This study demonstrates a role for the extracellular matrix protein nephronectin (NPNT) in promoting experimental breast cancer brain metastasis, possibly through enhanced binding to- and migration through brain endothelial cells. With the introduction of more targeted breast cancer treatments, a prolonged survival has resulted during the last decade. Consequently, an increased number of patients develop metastasis in the brain, a challenging organ to treat. We recently reported that NPNT was highly expressed in primary breast cancer and associated with unfavourable prognosis. The current study addresses our hypothesis that NPNT promotes brain metastases through its integrin-binding motifs. SAGE-sequencing revealed that NPNT was significantly up-regulated in human breast cancer tissue compared to pair-matched normal breast tissue. Human brain metastatic breast cancers expressed both NPNT and its receptor, integrin α8ß1. Using an open access repository; BreastMark, we found a correlation between high NPNT mRNA levels and poor prognosis for patients with the luminal B subtype. The 66cl4 mouse cell line was used for expression of wild-type and mutant NPNT, which is unable to bind α8ß1. Using an in vivo model of brain metastatic colonization, 66cl4-NPNT cells showed an increased ability to form metastatic lesions compared to cells with mutant NPNT, possibly through reduced endothelial adhesion and transmigration.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Extracellular Matrix Proteins/metabolism , Integrins/metabolism , Animals , Brain/metabolism , Brain/pathology , Breast/metabolism , Breast/pathology , Cell Differentiation/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Prognosis , RNA, Messenger/metabolism
7.
Mol Oncol ; 14(9): 2040-2057, 2020 09.
Article in English | MEDLINE | ID: mdl-32534480

ABSTRACT

Brain metastases are life-threatening complications of triple-negative breast cancer, melanoma, and a few other tumor types. Poor outcome of cerebral secondary tumors largely depends on the microenvironment formed by cells of the neurovascular unit, among which pericytes are the least characterized. By using in vivo and in vitro techniques and human samples, here we show that pericytes play crucial role in the development of metastatic brain tumors by directly influencing key steps of the development of the disease. Brain pericytes had a prompt chemoattractant effect on breast cancer cells and established direct contacts with them. By secreting high amounts of extracellular matrix proteins, pericytes enhanced adhesion of both melanoma and triple-negative cancer cells, which might be particularly important in the exclusive perivascular growth of these tumor cells. In addition, pericytes secreted insulin-like growth factor 2 (IGF2), which had a very significant pro-proliferative effect on mammary carcinoma, but not on melanoma cells. By inhibiting IGF2 signaling using silencing or picropodophyllin (PPP), we could block the proliferation-increasing effect of pericytes on breast cancer cells. Administration of PPP (a blood-brain barrier-permeable substance) significantly decreased the size of brain tumors in mice inoculated with triple-negative breast cancer cells. Taken together, our results indicate that brain pericytes have significant pro-metastatic features, especially in breast cancer. Our study underlines the importance of targeting pericytes and the IGF axis as potential strategies in brain metastatic diseases.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Insulin-Like Growth Factor II/metabolism , Pericytes/metabolism , Animals , Brain/pathology , Cell Adhesion , Cell Communication , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Insulin-Like Growth Factor II/genetics , Mice , Pericytes/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Mol Oncol ; 14(3): 520-538, 2020 03.
Article in English | MEDLINE | ID: mdl-31930767

ABSTRACT

Breast cancer brain metastases (BCBMs) have been underinvestigated despite their high incidence and poor outcome. MicroRNAs (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 timepoints (5 h, 3, 7, and 10 days) to follow metastasis development in the brain and in peripheral organs. Metastases were detected from 7 days onwards, mainly in the brain. NGS revealed a deregulation of circulating miRNA profile during BCBM progression, rising 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.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Mammary Neoplasms, Animal/blood , MicroRNAs/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Computational Biology , Disease Progression , Down-Regulation , Female , Gene Expression Regulation, Neoplastic/genetics , High-Throughput Nucleotide Sequencing , Humans , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/secondary , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Up-Regulation , Xenograft Model Antitumor Assays
9.
Acta Neuropathol Commun ; 7(1): 133, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31426859

ABSTRACT

Therapeutic resistance of cerebral secondary tumours largely depends on unique aspects linked to the neurovascular unit, especially cerebral endothelial cells and astrocytes. By using advanced microscopy techniques, here we explored novel mechanisms related to the neurovascular unit during extravasation and proliferation of triple negative breast cancer cells in the brain. Metastatic mammary carcinoma cells arrested and elongated within one hour in cerebral microvessels, but their number decreased by almost 80% in the first two days. Interestingly, malignant cells induced vasoconstriction and development of intraluminal endothelial plugs, which isolated invading cells from the circulation. During diapedesis - which usually took place on day four and five after inoculation of the tumour cells - continuity of cerebral endothelial tight junctions remained intact, indicating migration of cancer cells through the transcellular pathway. In addition, metastatic cells induced formation of multiluminal vessels and claudin-5-positive endothelial blebs. However, even severe endothelial blebbing could be reversed and the vessel morphology was restored shortly after the tumour cells completed transendothelial migration. Similar to neuro-inflammatory leukocytes, tumour cells migrated not only through the endothelial layer, but through the glia limitans perivascularis as well. Nevertheless, along with the growth of metastatic lesions by co-option of pre-existing capillaries, astrocytes and astrocyte end-feet were gradually expelled from the vessels to the border of the tumour. Taken together, we identified previously unknown mechanisms involved in the reaction of brain resident cells to invading breast cancer cells. Our results contribute to a better understanding of the complex cross-talk between tumour cells and host cells in the brain, which is essential for the identification of new therapeutic targets in this devastating disease.


Subject(s)
Brain/blood supply , Brain/pathology , Breast Neoplasms/pathology , Cell Movement/physiology , Endothelial Cells/pathology , Animals , Brain/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Breast Neoplasms/diagnostic imaging , Cells, Cultured , Cerebral Cortex/blood supply , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Coculture Techniques , Female , Mice , Mice, Inbred BALB C
10.
J Cell Mol Med ; 23(4): 2619-2631, 2019 04.
Article in English | MEDLINE | ID: mdl-30712288

ABSTRACT

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.


Subject(s)
Brain Neoplasms/secondary , Breast Neoplasms/pathology , Cerebral Cortex/pathology , Melanoma, Experimental/pathology , Skin Neoplasms/pathology , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Coculture Techniques , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Gene Expression , Humans , Melanocytes/metabolism , Melanocytes/pathology , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Transplantation , Organ Specificity , Primary Cell Culture , Skin Neoplasms/blood supply , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tumor Cells, Cultured
11.
J Cereb Blood Flow Metab ; 38(4): 563-587, 2018 04.
Article in English | MEDLINE | ID: mdl-28920514

ABSTRACT

Despite the potential obstacle represented by the blood-brain barrier for extravasating malignant cells, metastases are more frequent than primary tumors in the central nervous system. Not only tightly interconnected endothelial cells can hinder metastasis formation, other cells of the brain microenvironment (like astrocytes and microglia) can also be very hostile, destroying the large majority of metastatic cells. However, malignant cells that are able to overcome these harmful mechanisms may benefit from the shielding and even support provided by cerebral endothelial cells, astrocytes and microglia, rendering the brain a sanctuary site against anti-tumor strategies. Thus, cells of the neurovascular unit have a Janus-faced attitude towards brain metastatic cells, being both destructive and protective. In this review, we present the main mechanisms of brain metastasis formation, including those involved in extravasation through the brain vasculature and survival in the cerebral environment.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Endothelial Cells/pathology , Neurons/pathology , Animals , Blood-Brain Barrier , Cerebrovascular Circulation , Humans , Microglia/pathology
12.
Brain Behav Immun ; 64: 220-231, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28432035

ABSTRACT

Cerebral pericytes are mural cells embedded in the basement membrane of capillaries. Increasing evidence suggests that they play important role in controlling neurovascular functions, i.e. cerebral blood flow, angiogenesis and permeability of the blood-brain barrier. These cells can also influence neuroinflammation which is highly regulated by the innate immune system. Therefore, we systematically tested the pattern recognition receptor expression of brain pericytes. We detected expression of NOD1, NOD2, NLRC5, NLRP1-3, NLRP5, NLRP9, NLRP10 and NLRX mRNA in non-treated cells. Among the ten known human TLRs, TLR2, TLR4, TLR5, TLR6 and TLR10 were found to be expressed. Inflammatory mediators induced the expression of NLRA, NLRC4 and TLR9 and increased the levels of NOD2, TLR2, inflammasome-forming caspases and inflammasome-cleaved interleukins. Oxidative stress, on the other hand, upregulated expression of TLR10 and NLRP9. Activation of selected pattern recognition receptors can lead to inflammasome assembly and caspase-dependent secretion of IL-1ß. TNF-α and IFN-γ increased the levels of pro-IL-1ß and pro-caspase-1 proteins; however, no canonical activation of NLRP1, NLRP2, NLRP3 or NLRC4 inflammasomes could be observed in human brain vascular pericytes. On the other hand, we could demonstrate secretion of active IL-1ß in response to non-canonical inflammasome activation, i.e. intracellular LPS or infection with E. coli bacteria. Our in vitro results indicate that pericytes might have an important regulatory role in neuroinflammation.


Subject(s)
Brain/metabolism , Inflammasomes/metabolism , Pericytes/metabolism , Receptors, Pattern Recognition/metabolism , Cells, Cultured , Gene Expression Regulation , Humans , Interleukin-1beta/metabolism , Signal Transduction
13.
Curr Pharm Des ; 22(35): 5442-5462, 2016.
Article in English | MEDLINE | ID: mdl-27464716

ABSTRACT

Besides being indispensable for the protection and nutrition of the central nervous system (CNS), blood-brain barrier (BBB)-forming cerebral endothelial cells (CECs) have a major role in hampering drugs to reach therapeutically relevant concentrations in the brain. In this respect, the most important defense systems of CECs are tight junctions (TJs) sealing the paracellular way of transport, efflux pumps (ABC transporters) and metabolic enzymes. Here we review current strategies aiming at overcoming the BBB with the purpose of effectively delivering drugs to the CNS. Besides chemical modification of drug candidates to improve CNS availability, the main strategies include: bypassing the BBB (intracranial or nasal routes), reversible opening of TJs (using hyperosmotic mannitol, ultrasounds, peptides and other physical methods or chemical agents), vector-mediated drug delivery systems (nanocarriers, exploitation of receptor- and carrier-mediated transport) and inhibition of efflux transporters. We discuss the main advantages, disadvantages and clinical relevance of each strategy. Special emphasis will be given to the description of the chemical characteristics of nanoparticles (lipidic, polymeric, inorganic, etc.) and the main strategies of targeting them to the CNS.


Subject(s)
Blood-Brain Barrier/drug effects , Brain/drug effects , Central Nervous System/drug effects , Drug Delivery Systems , Animals , Humans
14.
Am J Physiol Heart Circ Physiol ; 310(11): H1702-14, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27059078

ABSTRACT

The blood-brain barrier (BBB) is the main interface controlling molecular and cellular traffic between the central nervous system (CNS) and the periphery. It consists of cerebral endothelial cells (CECs) interconnected by continuous tight junctions, and closely associated pericytes and astrocytes. Different parts of the CNS have diverse functions and structures and may be subject of different pathologies, in which the BBB is actively involved. It is largely unknown, however, what are the cellular and molecular differences of the BBB in different regions of the brain. Using in silico, in vitro, and ex vivo techniques we compared the expression of BBB-associated genes and proteins (i.e., markers of CECs, brain pericytes, and astrocytes) in the cortical grey matter and white matter. In silico human database analysis (obtained from recalculated data of the Allen Brain Atlas), qPCR, Western blot, and immunofluorescence studies on porcine and mouse brain tissue indicated an increased expression of glial fibrillary acidic protein in astrocytes in the white matter compared with the grey matter. We have also found increased expression of genes of the junctional complex of CECs (occludin, claudin-5, and α-catenin) in the white matter compared with the cerebral cortex. Accordingly, occludin, claudin-5, and α-catenin proteins showed increased expression in CECs of the white matter compared with endothelial cells of the cortical grey matter. In parallel, barrier properties of white matter CECs were superior as well. These differences might be important in the pathogenesis of diseases differently affecting distinct regions of the brain.


Subject(s)
Blood-Brain Barrier/metabolism , Cell Adhesion Molecules/metabolism , Cerebral Cortex/metabolism , Gray Matter/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, Growth Factor/metabolism , Tight Junction Proteins/metabolism , White Matter/metabolism , Animals , Astrocytes/metabolism , Computer Simulation , Female , Humans , Male , Mice , Molecular Structure , Pericytes/metabolism , Swine , Tight Junctions/metabolism
15.
Cell Adh Migr ; 10(3): 269-81, 2016 05 03.
Article in English | MEDLINE | ID: mdl-26645485

ABSTRACT

Brain metastases are common and devastating complications of both breast cancer and melanoma. Although mammary carcinoma brain metastases are more frequent than those originating from melanoma, this latter has the highest tropism to the brain. Using static and dynamic in vitro approaches, here we show that melanoma cells have increased adhesion to the brain endothelium in comparison to breast cancer cells. Moreover, melanoma cells can transmigrate more rapidly and in a higher number through brain endothelial monolayers than breast cancer cells. In addition, melanoma cells have increased ability to impair tight junctions of cerebral endothelial cells. We also show that inhibition of Rac or PI3K impedes adhesion of breast cancer cells and melanoma cells to the brain endothelium. In addition, inhibition of Rac or PI3K inhibits the late phase of transmigration of breast cancer cells and the early phase of transmigration of melanoma cells. On the other hand, the Rac inhibitor EHT1864 impairs the junctional integrity of the brain endothelium, while the PI3K inhibitor LY294002 has no damaging effect on interendothelial junctions. We suggest that targeting the PI3K/Akt pathway may represent a novel opportunity in preventing the formation of brain metastases of melanoma and breast cancer.


Subject(s)
Brain/pathology , Breast Neoplasms/pathology , Endothelium, Vascular/pathology , Melanoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Transendothelial and Transepithelial Migration , rac1 GTP-Binding Protein/metabolism , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Endothelial Cells , Female , Humans , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrones/pharmacology , Quinolines/pharmacology , Rats , Tight Junctions/drug effects , Tight Junctions/metabolism
16.
J Neurochem ; 135(3): 551-64, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26083549

ABSTRACT

Cerebral endothelial cells (CECs) forming the blood-brain barrier are at the interface of the immune and the central nervous systems and thus may play an important role in the functional integration of the two systems. Here, we investigated how CECs recognize and respond to pathogen- and damage-associated molecular patterns to regulate the functions of the neurovascular unit. First we detected the expression of several NOD-like receptors (NLRs) - including NOD1, NOD2, NLRC4, NLRC5, NLRP1, NLRP3, NLRP5, NLRP9, NLRP10, NLRP12, NLRA, and NLRX - in human brain endothelial cells. Inflammatory cytokines, such as interferon-γ, tumor necrosis factor-α, and IL-1ß had stimulatory effects on the transcription of many of these receptors. Expression of key inflammasome components (NOD2, NLRP3, and caspase 1) along with caspase-cleaved interleukins IL-1ß and IL-33 could be induced by priming with lipopolysaccharide and activation with muramyl dipeptide. In addition, combined treatment with lipopolysaccharide and muramyl dipeptide resulted in IL-1ß secretion in a caspase- and ERK1/2 kinase-dependent manner. Our findings demonstrate that NLRs and inflammasomes can be activated in cerebral endothelial cells, which may confer a yet unexplored role to the blood-brain barrier in neuroimmune and neuroinflammatory processes.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Inflammasomes/metabolism , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Animals , Cells, Cultured , Humans , Rats
17.
J Nat Prod ; 77(12): 2641-50, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25479041

ABSTRACT

Two new arylbenzofuran-type neolignans, 1"-dehydroegonol 3"-methyl ether (1) and egonol 3"-methyl ether (2), and four known lignan derivatives, namely, helioxanthin (3), (7E)-7,8-dehydroheliobuphthalmin (4), heliobuphthalmin (5), and 7-acetoxyhinokinin (6), were isolated from a chloroform-soluble partition of the methanol extract of the fresh roots of Heliopsis helianthoides var. scabra. These six compounds were evaluated in vitro in terms of their ability to inhibit the various steps involved in brain tumor metastasis formation. Compounds 3 and 4 inhibited the migration of both melanoma and brain endothelial cells, and 3 also reduced the adhesion of melanoma cells to the brain endothelium. Furthermore, 3 and 4 additionally enhanced the barrier function of the blood-brain barrier and the expression of the tight junction protein ZO-1 at the junctions of the endothelial cells. These findings suggest that 3 and 4 may have the potential to interfere with different steps of brain metastasis formation and to enhance the barrier function of cerebral endothelial cells.


Subject(s)
Asteraceae/chemistry , Brain/drug effects , Lignans/isolation & purification , Lignans/pharmacology , Actin Cytoskeleton , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Cell Survival/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Hungary , Lignans/chemistry , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Plant Roots/chemistry , Rats , Zonula Occludens-1 Protein/drug effects
18.
Int J Mol Sci ; 15(5): 8063-74, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24815068

ABSTRACT

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.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Neoplasms/secondary , Cell Movement , Melanoma/metabolism , Melanoma/pathology , Receptor, Cannabinoid, CB2/metabolism , Animals , Blood-Brain Barrier/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Adhesion , Cell Line , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Rats
19.
Pigment Cell Melanoma Res ; 27(1): 113-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24148763

ABSTRACT

We have investigated the role of the Rho/ROCK signaling pathway in the interaction of metastatic melanoma cells with the brain endothelium. ROCK inhibition induced a shift of melanoma cells to the mesenchymal phenotype, increased the number of melanoma cells attached to the brain endothelium, and strengthened the adhesion force between melanoma and endothelial cells. Inhibition of ROCK raised the number of melanoma cells migrating through the brain endothelial monolayer and promoted the formation of parenchymal brain metastases in vivo. We have shown that inhibition of the Rho/ROCK pathway in melanoma, but not in brain endothelial cells, is responsible for this phenomenon. Our results indicate that the mesenchymal type of tumor cell movement is primordial in the transmigration of melanoma cells through the blood-brain barrier.


Subject(s)
Blood-Brain Barrier/enzymology , Cell Communication , Cell Movement , Endothelial Cells/enzymology , Melanoma/enzymology , Neoplasm Proteins/metabolism , Signal Transduction , rho-Associated Kinases/metabolism , Animals , Blood-Brain Barrier/pathology , Cell Line, Tumor , Endothelial Cells/pathology , Melanoma/genetics , Melanoma/pathology , Mice , Neoplasm Proteins/genetics , rho-Associated Kinases/genetics
20.
Int J Mol Sci ; 14(1): 1383-411, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23344048

ABSTRACT

The majority of brain metastases originate from lung cancer, breast cancer and malignant melanoma. In order to reach the brain, parenchyma metastatic cells have to transmigrate through the endothelial cell layer of brain capillaries, which forms the morphological basis of the blood-brain barrier (BBB). The BBB has a dual role in brain metastasis formation: it forms a tight barrier protecting the central nervous system from entering cancer cells, but it is also actively involved in protecting metastatic cells during extravasation and proliferation in the brain. The mechanisms of interaction of cancer cells and cerebral endothelial cells are largely uncharacterized. Here, we provide a comprehensive review on our current knowledge about the role of junctional and adhesion molecules, soluble factors, proteolytic enzymes and signaling pathways mediating the attachment of tumor cells to brain endothelial cells and the transendothelial migration of metastatic cells. Since brain metastases represent a great therapeutic challenge, it is indispensable to understand the mechanisms of the interaction of tumor cells with the BBB in order to find targets of prevention of brain metastasis formation.


Subject(s)
Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Breast Neoplasms/metabolism , Lung Neoplasms/metabolism , Melanoma/metabolism , Blood-Brain Barrier/physiopathology , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Melanoma/genetics , Melanoma/pathology , Models, Biological , Signal Transduction , Transendothelial and Transepithelial Migration
SELECTION OF CITATIONS
SEARCH DETAIL
...