Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
EBioMedicine ; 99: 104914, 2024 Jan.
Article En | MEDLINE | ID: mdl-38113759

BACKGROUND: Cerebral Cavernous Malformation (CCM) is a rare cerebrovascular disease, characterized by the presence of multiple vascular malformations that may result in intracerebral hemorrhages (ICHs), seizure(s), or focal neurological deficits (FND). Familial CCM (fCCM) is due to loss of function mutations in one of the three independent genes KRIT1 (CCM1), Malcavernin (CCM2), or Programmed Cell death 10 (PDCD10/CCM3). The aim of this study was to identify plasma protein biomarkers of fCCM to assess the severity of the disease and predict its progression. METHODS: Here, we have investigated plasma samples derived from n = 71 symptomatic fCCM patients (40 female/31 male) and n = 17 healthy donors (HD) (9 female/8 male) of the Phase 1/2 Treat_CCM trial, using multiplexed protein profiling approaches. FINDINGS: Biomarkers as sCD14 (p = 0.00409), LBP (p = 0.02911), CXCL4 (p = 0.038), ICAM-1 (p = 0.02013), ANG2 (p = 0.026), CCL5 (p = 0.00403), THBS1 (p = 0.0043), CRP (p = 0.0092), and HDL (p = 0.027), were significantly different in fCCM compared to HDs. Of note, sENG (p = 0.011), THBS1 (p = 0.011) and CXCL4 (p = 0.011), were correlated to CCM genotype. sROBO4 (p = 0.014), TM (p = 0.026) and CRP (p = 0.040) were able to predict incident adverse clinical events, such as ICH, FND or seizure. GDF-15, FLT3L, CXCL9, FGF-21 and CDCP1, were identified as predictors of the formation of new MRI-detectable lesions over 2-year follow-up. Furthermore, the functional relevance of ang2, thbs1, robo4 and cdcp1 markers was validated by zebrafish pre-clinical model of fCCM. INTERPRETATION: Overall, our study identifies a set of biochemical parameters to predict CCM progression, suggesting biological interpretations and potential therapeutic approaches to CCM disease. FUNDING: Italian Medicines Agency, Associazione Italiana per la Ricerca sul Cancro (AIRC), ERC, Leducq Transatlantic Network of Excellence, Swedish Research Council.


Hemangioma, Cavernous, Central Nervous System , Animals , Humans , Male , Female , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/genetics , Proto-Oncogene Proteins/genetics , Microtubule-Associated Proteins/genetics , Zebrafish/metabolism , Biomarkers , Seizures , Antigens, Neoplasm , Cell Adhesion Molecules
2.
Cell Mol Life Sci ; 79(6): 340, 2022 Jun 04.
Article En | MEDLINE | ID: mdl-35661927

Cerebral cavernous malformations (CCM) are low-flow vascular lesions prone to cause severe hemorrhage-associated neurological complications. Pathogenic germline variants in CCM1, CCM2, or CCM3 can be identified in nearly 100% of CCM patients with a positive family history. In line with the concept that tumor-like mechanisms are involved in CCM formation and growth, we here demonstrate an abnormally increased proliferation rate of CCM3-deficient endothelial cells in co-culture with wild-type cells and in mosaic human iPSC-derived vascular organoids. The observation that NSC59984, an anticancer drug, blocked the abnormal proliferation of mutant endothelial cells further supports this intriguing concept. Fluorescence-activated cell sorting and RNA sequencing revealed that co-culture induces upregulation of proangiogenic chemokine genes in wild-type endothelial cells. Furthermore, genes known to be significantly downregulated in CCM3-/- endothelial cell mono-cultures were upregulated back to normal levels in co-culture with wild-type cells. These results support the hypothesis that wild-type ECs facilitate the formation of a niche that promotes abnormal proliferation of mutant ECs. Thus, targeting the cancer-like features of CCMs is a promising new direction for drug development.


Endothelial Cells , Hemangioma, Cavernous, Central Nervous System , Apoptosis Regulatory Proteins/genetics , Cell Proliferation , Coculture Techniques , Endothelial Cells/pathology , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/pathology , Humans , Proto-Oncogene Proteins/genetics
3.
STAR Protoc ; 3(2): 101448, 2022 06 17.
Article En | MEDLINE | ID: mdl-35712011

In the study of cerebral cavernous malformations (CCMs), the quantification of lesion burden is the main parameter for evaluation of disease severity and efficacy of drugs. We describe a reliable and cost-effective protocol to evaluate the number and the size of vascular malformations in the murine brain. This approach is based on histology and confocal imaging and can be performed with standard laboratory equipment. We detail the preparation of brain sections followed by image acquisition and analysis. For complete details on the use and execution of this protocol, please refer to Maderna et al. (2022).


Hemangioma, Cavernous, Central Nervous System , Animals , Brain/diagnostic imaging , Hemangioma, Cavernous, Central Nervous System/diagnostic imaging , Histological Techniques , Mice
4.
Genes (Basel) ; 13(6)2022 05 27.
Article En | MEDLINE | ID: mdl-35741725

Cerebral cavernous malformations (CCM) are capillary malformations affecting the central nervous system and commonly present with headaches, epilepsy and stroke. Treatment of CCM is symptomatic, and its prevention is limited. CCM are often sporadic but sometimes may be multifocal and/or affect multiple family members. Heterozygous pathogenic variants in PDCD10 cause the rarest and apparently most severe genetic variant of familial CCM. We carried out an RNA-Seq and a Q-PCR validation analysis in Pdcd10-silenced and wild-type mouse endothelial cells in order to better elucidate CCM molecular pathogenesis. Ninety-four differentially expressed genes presented an FDR-corrected p-value < 0.05. A functionally clustered dendrogram showed that differentially expressed genes cluster in cell proliferation, oxidative stress, vascular processes and immune response gene-ontology functions. Among differentially expressed genes, the major cluster fell in signaling related to inflammation and pathogen recognition, including HIF1α and Nos2 signaling and immune regulation. Validation analysis performed on wild-type, Pdcd10-null and Pdcd10-null reconstituted cell lines was consistent with RNA-Seq data. This work confirmed previous mouse transcriptomic data in endothelial cells, which are recognized as a critical tissue for CCM formation and expands the potential molecular signatures of PDCD10-related familial CCM to alterations in inflammation and pathogen recognition pathways.


Endothelial Cells , Inflammation , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Endothelial Cells/metabolism , Gene Expression Profiling , Hemangioma, Cavernous, Central Nervous System , Hypoxia/metabolism , Inflammation/genetics , Inflammation/metabolism , Mice
6.
Cell Mol Life Sci ; 79(4): 206, 2022 Mar 25.
Article En | MEDLINE | ID: mdl-35333979

Cerebral Cavernous Malformation (CCM) is a brain vascular disease with various neurological symptoms. In this study, we describe the inflammatory profile in CCM and show for the first time the formation of neutrophil extracellular traps (NETs) in rodents and humans with CCM. Through RNA-seq analysis of cerebellum endothelial cells from wild-type mice and mice with an endothelial cell-specific ablation of the Ccm3 gene (Ccm3iECKO), we show that endothelial cells from Ccm3iECKO mice have an increased expression of inflammation-related genes. These genes encode proinflammatory cytokines and chemokines, as well as adhesion molecules, which promote recruitment of inflammatory and immune cells. Similarly, immunoassays showed elevated levels of these cytokines and chemokines in the cerebellum of the Ccm3iECKO mice. Consistently, both flow cytometry and immunofluorescence analysis showed infiltration of different subsets of leukocytes into the CCM lesions. Neutrophils, which are known to fight against infection through different strategies, including the formation of NETs, represented the leukocyte subset within the most pronounced increase in CCM. Here, we detected elevated levels of NETs in the blood and the deposition of NETs in the cerebral cavernomas of Ccm3iECKO mice. Degradation of NETs by DNase I treatment improved the vascular barrier. The deposition of NETs in the cavernomas  of patients with CCM confirms the clinical relevance of NETs in CCM.


Extracellular Traps , Hemangioma, Cavernous, Central Nervous System , Animals , Apoptosis Regulatory Proteins/genetics , Endothelial Cells/metabolism , Extracellular Traps/metabolism , Hemangioma, Cavernous, Central Nervous System/genetics , Hemangioma, Cavernous, Central Nervous System/metabolism , Hemangioma, Cavernous, Central Nervous System/pathology , Humans , Inflammation/pathology , Membrane Proteins/metabolism , Mice
7.
iScience ; 25(3): 103943, 2022 Mar 18.
Article En | MEDLINE | ID: mdl-35265815

Cavernomas are multi-lumen and blood-filled vascular malformations which form in the brain and the spinal cord. They lead to hemorrhage, epileptic seizures, neurological deficits, and paresthesia. An effective medical treatment is still lacking, and the available murine models for cavernomas have several limitations for preclinical studies. These include disease phenotypes that differ from human diseases, such as restriction of the lesions to the cerebellum, and absence of acute hemorrhage. Additional limitations of current murine models include rapid development of lesions, which are lethal before the first month of age. Here, we have characterized a murine model that recapitulates features of the human disease: lesions develop after weaning throughout the entire CNS, including the spinal cord, and undergo acute hemorrhage. This provides a preclinical model to develop new drugs for treatment of acute hemorrhage in the brain and spinal cord, as an unmet medical emergency for patients with cavernomas.

8.
Genes Dis ; 8(6): 798-813, 2021 Nov.
Article En | MEDLINE | ID: mdl-34522709

The programmed cell death 10 (PDCD10) gene was originally identified as an apoptosis-related gene, although it is now usually known as CCM3, as the third causative gene of cerebral cavernous malformation (CCM). CCM is a neurovascular disease that is characterized by vascular malformations and is associated with headaches, seizures, focal neurological deficits, and cerebral hemorrhage. The PDCD10/CCM3 protein has multiple subcellular localizations and interacts with several multi-protein complexes and signaling pathways. Thus PDCD10/CCM3 governs many cellular functions, which include cell-to-cell junctions and cytoskeleton organization, cell proliferation and apoptosis, and exocytosis and angiogenesis. Given its central role in the maintenance of homeostasis of the cell, dysregulation of PDCD10/CCM3 can result in a wide range of altered cell functions. This can lead to severe diseases, including CCM, cognitive disability, and several types of cancers. Here, we review the multifaceted roles of PDCD10/CCM3 in physiology and pathology, with a focus on its functions beyond CCM.

9.
J Cell Sci ; 134(15)2021 08 01.
Article En | MEDLINE | ID: mdl-34338295

Endothelial-to-mesenchymal transition (EndMT) is the biological process through which endothelial cells transdifferentiate into mesenchymal cells. During embryo development, EndMT regulates endocardial cushion formation via TGFß/BMP signaling. In adults, EndMT is mainly activated during pathological conditions. Hence, it is necessary to characterize molecular regulators cooperating with TGFß signaling in driving EndMT, to identify potential novel therapeutic targets to treat these pathologies. Here, we studied YAP, a transcriptional co-regulator involved in several biological processes, including epithelial-to-mesenchymal transition (EMT). As EndMT is the endothelial-specific form of EMT, and YAP (herein referring to YAP1) and TGFß signaling cross-talk in other contexts, we hypothesized that YAP contributes to EndMT by modulating TGFß signaling. We demonstrate that YAP is required to trigger TGFß-induced EndMT response, specifically contributing to SMAD3-driven EndMT early gene transcription. We provide novel evidence that YAP acts as SMAD3 transcriptional co-factor and prevents GSK3ß-mediated SMAD3 phosphorylation, thus protecting SMAD3 from degradation. YAP is therefore emerging as a possible candidate target to inhibit pathological TGFß-induced EndMT at early stages.


Endothelial Cells , Transforming Growth Factor beta , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition , Phosphorylation , Transforming Growth Factor beta/metabolism
11.
Elife ; 92020 11 03.
Article En | MEDLINE | ID: mdl-33138917

Cerebral cavernous malformation (CCM) is a rare neurovascular disease that is characterized by enlarged and irregular blood vessels that often lead to cerebral hemorrhage. Loss-of-function mutations to any of three genes results in CCM lesion formation; namely, KRIT1, CCM2, and PDCD10 (CCM3). Here, we report for the first time in-depth single-cell RNA sequencing, combined with spatial transcriptomics and immunohistochemistry, to comprehensively characterize subclasses of brain endothelial cells (ECs) under both normal conditions and after deletion of Pdcd10 (Ccm3) in a mouse model of CCM. Integrated single-cell analysis identifies arterial ECs as refractory to CCM transformation. Conversely, a subset of angiogenic venous capillary ECs and respective resident endothelial progenitors appear to be at the origin of CCM lesions. These data are relevant for the understanding of the plasticity of the brain vascular system and provide novel insights into the molecular basis of CCM disease at the single cell level.


Endothelial Cells/cytology , Hemangioma, Cavernous, Central Nervous System/physiopathology , Animals , Apoptosis Regulatory Proteins/metabolism , Arteries/pathology , Brain/blood supply , Brain/pathology , Cell Differentiation , Disease Models, Animal , Gene Deletion , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Mitosis , Neovascularization, Pathologic , Phenotype , RNA-Seq , Sequence Analysis, RNA , Signal Transduction/genetics , Single-Cell Analysis , Tamoxifen/pharmacology , Transcriptome
12.
Circ Res ; 127(8): 1056-1073, 2020 09 25.
Article En | MEDLINE | ID: mdl-32673519

RATIONALE: Intercellular tight junctions are crucial for correct regulation of the endothelial barrier. Their composition and integrity are affected in pathological contexts, such as inflammation and tumor growth. JAM-A (junctional adhesion molecule A) is a transmembrane component of tight junctions with a role in maintenance of endothelial barrier function, although how this is accomplished remains elusive. OBJECTIVE: We aimed to understand the molecular mechanisms through which JAM-A expression regulates tight junction organization to control endothelial permeability, with potential implications under pathological conditions. METHODS AND RESULTS: Genetic deletion of JAM-A in mice significantly increased vascular permeability. This was associated with significantly decreased expression of claudin-5 in the vasculature of various tissues, including brain and lung. We observed that C/EBP-α (CCAAT/enhancer-binding protein-α) can act as a transcription factor to trigger the expression of claudin-5 downstream of JAM-A, to thus enhance vascular barrier function. Accordingly, gain-of-function for C/EBP-α increased claudin-5 expression and decreased endothelial permeability, as measured by the passage of fluorescein isothiocyanate (FITC)-dextran through endothelial monolayers. Conversely, C/EBP-α loss-of-function showed the opposite effects of decreased claudin-5 levels and increased endothelial permeability. Mechanistically, JAM-A promoted C/EBP-α expression through suppression of ß-catenin transcriptional activity, and also through activation of EPAC (exchange protein directly activated by cAMP). C/EBP-α then directly binds the promoter of claudin-5 to thereby promote its transcription. Finally, JAM-A-C/EBP-α-mediated regulation of claudin-5 was lost in blood vessels from tissue biopsies from patients with glioblastoma and ovarian cancer. CONCLUSIONS: We describe here a novel role for the transcription factor C/EBP-α that is positively modulated by JAM-A, a component of tight junctions that acts through EPAC to up-regulate the expression of claudin-5, to thus decrease endothelial permeability. Overall, these data unravel a regulatory molecular pathway through which tight junctions limit vascular permeability. This will help in the identification of further therapeutic targets for diseases associated with endothelial barrier dysfunction. Graphic Abstract: An graphic abstract is available for this article.


CCAAT-Enhancer-Binding Proteins/metabolism , Capillary Permeability , Cell Adhesion Molecules/metabolism , Claudin-5/metabolism , Endothelial Cells/metabolism , Receptors, Cell Surface/metabolism , Tight Junctions/metabolism , Adult , Aged , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CCAAT-Enhancer-Binding Proteins/genetics , Cell Adhesion Molecules/genetics , Cell Line , Claudin-5/genetics , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neovascularization, Pathologic , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Receptors, Cell Surface/genetics , Signal Transduction , Tight Junctions/genetics , Up-Regulation
13.
Nat Commun ; 10(1): 2761, 2019 06 24.
Article En | MEDLINE | ID: mdl-31235698

Cerebral cavernous malformation (CCM) is a neurovascular familial or sporadic disease that is characterised by capillary-venous cavernomas, and is due to loss-of-function mutations to any one of three CCM genes. Familial CCM follows a two-hit mechanism similar to that of tumour suppressor genes, while in sporadic cavernomas only a small fraction of endothelial cells shows mutated CCM genes. We reported that in mouse models and in human patients, endothelial cells lining the lesions have different features from the surrounding endothelium, as they express mesenchymal/stem-cell markers. Here we show that cavernomas originate from clonal expansion of few Ccm3-null endothelial cells that express mesenchymal/stem-cell markers. These cells then attract surrounding wild-type endothelial cells, inducing them to express mesenchymal/stem-cell markers and to contribute to cavernoma growth. These characteristics of Ccm3-null cells are reminiscent of the tumour-initiating cells that are responsible for tumour growth. Our data support the concept that CCM has benign tumour characteristics.


Apoptosis Regulatory Proteins/genetics , Central Nervous System Neoplasms/pathology , Endothelial Cells/pathology , Hemangioma, Cavernous, Central Nervous System/pathology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Biomarkers/metabolism , Brain/blood supply , Brain/cytology , Brain/pathology , Cell Differentiation/genetics , Cell Line , Central Nervous System Neoplasms/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Female , Gene Knockout Techniques , Hemangioma, Cavernous, Central Nervous System/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Loss of Function Mutation , Membrane Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins/metabolism
14.
Elife ; 82019 03 04.
Article En | MEDLINE | ID: mdl-30829570

The biological players involved in angiogenesis are only partially defined. Here, we report that endothelial cells (ECs) express a novel isoform of the cell-surface adhesion molecule L1CAM, termed L1-ΔTM. The splicing factor NOVA2, which binds directly to L1CAM pre-mRNA, is necessary and sufficient for the skipping of L1CAM transmembrane domain in ECs, leading to the release of soluble L1-ΔTM. The latter exerts high angiogenic function through both autocrine and paracrine activities. Mechanistically, L1-ΔTM-induced angiogenesis requires fibroblast growth factor receptor-1 signaling, implying a crosstalk between the two molecules. NOVA2 and L1-ΔTM are overexpressed in the vasculature of ovarian cancer, where L1-ΔTM levels correlate with tumor vascularization, supporting the involvement of NOVA2-mediated L1-ΔTM production in tumor angiogenesis. Finally, high NOVA2 expression is associated with poor outcome in ovarian cancer patients. Our results point to L1-ΔTM as a novel, EC-derived angiogenic factor which may represent a target for innovative antiangiogenic therapies.


Alternative Splicing , Angiogenic Proteins/metabolism , Endothelial Cells/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Protein Isoforms/metabolism , RNA-Binding Proteins/metabolism , Cells, Cultured , Humans , Neuro-Oncological Ventral Antigen
15.
Mod Pathol ; 31(9): 1361-1366, 2018 09.
Article En | MEDLINE | ID: mdl-29713042

We hypothesized that in glioblastoma recurring after radiotherapy, a condition whereby the brain endothelium undergoes radiation-induced senescence, tumor cells with endothelial phenotype may be relevant for tumor neovascularization. Matched glioblastoma samples obtained at primary surgery and at surgery for tumor recurrence after radiotherapy, all expressing epidermal growth factor receptor variant III (EGFRvIII), were assessed by a technique that combines fluorescent in situ hybridization (FISH) for the EGFR/CEP7 chromosomal probe with immunostaining for endothelial cells (CD31) and activated pericytes (α Smooth Muscle Actin). Five EGFRvIII-expressing paired primary/recurrent glioblastoma samples, in which the tumor cells showed EGFR/CEP7 amplification, were then assessed by CD31 and α Smooth Muscle Actin immunofluorescence. In glomeruloid bodies, the ratio between CD31+ cells with amplified EGFR/CEP7 signal and the total CD31+ cells was 0.23 ± 0.09 (mean ± sem) and 0.63 ± 0.07 in primary tumors and in recurrent ones, respectively (p < 0.002, Student-t test). In capillaries, the ratio of CD31+ cells with amplified EGFR/CEP7 over the total CD31+ cells lining the capillary lumen was 0.21 ± 0.06 (mean ± sem) and 0.42 ± 0.07 at primary surgery and at recurrence, respectively (p < 0.005, Student-t test). Expression of α Smooth Muscle Actin by cells with EGFR/CEP7 amplification was not observed. Then, in glioblastoma recurring after radiotherapy, where the brain endothelium suffers from radiation-induced cell senescence, tumor-derived endothelium plays a role in neo-vascularization.


Brain Neoplasms/pathology , Cell Transdifferentiation/physiology , Endothelial Cells/pathology , Glioblastoma/pathology , Neoplasm Recurrence, Local/pathology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Endothelial Cells/metabolism , Endothelial Cells/radiation effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glioblastoma/genetics , Glioblastoma/radiotherapy , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/radiation effects , Humans , In Situ Hybridization, Fluorescence , Mice , Neoplasm Recurrence, Local/genetics
16.
Arterioscler Thromb Vasc Biol ; 38(2): 353-362, 2018 02.
Article En | MEDLINE | ID: mdl-29284606

OBJECTIVE: The assembly of a functional vascular system requires a coordinated and dynamic transition from activation to maturation. High vascular endothelial growth factor activity promotes activation, including junction destabilization and cell motility. Maturation involves junctional stabilization and formation of a functional endothelial barrier. The identity and mechanism of action of prostabilization signals are still mostly unknown. Bone morphogenetic protein receptors and their ligands have important functions during embryonic vessel assembly and maturation. Previous work has suggested a role for growth differentiation factor 6 (GDF6; bone morphogenetic protein 13) in vascular integrity although GDF6's mechanism of action was not clear. Therefore, we sought to further explore the requirement for GDF6 in vascular stabilization. APPROACH AND RESULTS: We investigated the role of GDF6 in promoting endothelial vascular integrity in vivo in zebrafish and in cultured human umbilical vein endothelial cells in vitro. We report that GDF6 promotes vascular integrity by counteracting vascular endothelial growth factor activity. GDF6-deficient endothelium has increased vascular endothelial growth factor signaling, increased vascular endothelial-cadherin Y658 phosphorylation, vascular endothelial-cadherin delocalization from cell-cell interfaces, and weakened endothelial cell adherence junctions that become prone to vascular leak. CONCLUSIONS: Our results suggest that GDF6 promotes vascular stabilization by restraining vascular endothelial growth factor signaling. Understanding how GDF6 affects vascular integrity may help to provide insights into hemorrhage and associated vascular pathologies in humans.


Capillary Permeability , Embryo, Nonmammalian/blood supply , Endothelial Cells/metabolism , Growth Differentiation Factor 6/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Gene Expression Regulation, Developmental , Growth Differentiation Factor 6/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mitogen-Activated Protein Kinase 3/metabolism , Neovascularization, Physiologic , Phosphorylation , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
17.
Curr Opin Hematol ; 24(3): 256-264, 2017 May.
Article En | MEDLINE | ID: mdl-28212190

PURPOSE OF REVIEW: Endothelial cells dysfunctions are crucial determinants of several human diseases. We review here the most recent reports on endothelial cell defects in cerebral cavernous malformations (CCMs), particularly focusing on adherens junctions. CCM is a vascular disease that affects specifically the venous microvessels of the central nervous system and which is caused by loss-of-function mutation in any one of the three CCM genes (CCM1, 2 or 3) in endothelial cells. The phenotypic result of these mutations are focal vascular malformations that are permeable and fragile causing neurological symptoms and occasionally haemorrhagic stroke. RECENT FINDINGS: CCM is still an incurable disease, as no pharmacological treatment is available, besides surgery. The definition of the molecular alterations ensuing loss of function mutation of CCM genes is contributing to orientate the testing of targeted pharmacological tools.Several signalling pathways are altered in the three genotypes in a similar way and concur in the acquisition of mesenchymal markers in endothelial cells. However, also genotype-specific defects are reported, in particular for the CCM1 and CCM3 mutation. SUMMARY: Besides the specific CCM disease, the characterization of endothelial alterations in CCM has the potentiality to shed light on basic molecular regulations as the acquisition and maintenance of organ and vascular site specificity of endothelial cells.


Endothelial Cells/metabolism , Hemangioma, Cavernous, Central Nervous System/etiology , Hemangioma, Cavernous, Central Nervous System/metabolism , Adherens Junctions/metabolism , Angiopoietins/metabolism , Animals , Biomarkers , Exocytosis , Hemangioma, Cavernous, Central Nervous System/pathology , Humans , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Translational Research, Biomedical
18.
Stem Cells ; 35(5): 1328-1340, 2017 05.
Article En | MEDLINE | ID: mdl-28090691

Vascular associated endothelial cell (ECs) progenitors are still poorly studied and their role in the newly forming vasculature at embryonic or postnatal stage remains elusive. In the present work, we first defined a set of genes highly expressed during embryo development and strongly downregulated in the adult mouse. In this group, we then concentrated on the progenitor cell marker Peg3/PW1. By in vivo staining of the vasculature we found that only a subset of cells coexpressed endothelial markers and PW1. These cells were quite abundant in the embryo vasculature but declined in number at postnatal and adult stages. Using a reporter mouse for PW1 expression, we have been able to isolate PW1-positive (PW1posECs) and negative endothelial cells (PW1negECs). PW1-positive cells were highly proliferative in comparison to PW1negECs and were able to form colonies when seeded at clonal dilution. Furthermore, by RNAseq analysis, PW1posECs expressed endothelial cell markers together with mesenchymal and stem cell markers. When challenged by endothelial growth factors in vitro, PW1posECs were able to proliferate more than PW1negECs and to efficiently form new vessels in vivo. Taken together these data identify a subset of vessel associated endothelial cells with characteristics of progenitor cells. Considering their high proliferative potential these cells may be of particular importance to design therapies to improve the perfusion of ischemic tissues or to promote vascular repair. Stem Cells 2017;35:1328-1340.


Blood Vessels/cytology , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Kruppel-Like Transcription Factors/metabolism , Animals , Biomarkers/metabolism , Blood Vessels/embryology , Blood Vessels/metabolism , Cell Proliferation/drug effects , Cell Separation , Embryonic Development/drug effects , Endothelial Progenitor Cells/drug effects , Gene Expression Profiling , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Phenotype
19.
Stroke ; 47(3): 886-90, 2016 Mar.
Article En | MEDLINE | ID: mdl-26839352

BACKGROUND AND PURPOSE: Cerebral cavernous malformation (CCM) is characterized by multiple lumen vascular malformations in the central nervous system that can cause neurological symptoms and brain hemorrhages. About 20% of CCM patients have an inherited form of the disease with ubiquitous loss-of-function mutation in any one of 3 genes CCM1, CCM2, and CCM3. The rest of patients develop sporadic vascular lesions histologically similar to those of the inherited form and likely mediated by a biallelic acquired mutation of CCM genes in the brain vasculature. However, the molecular phenotypic features of endothelial cells in CCM lesions in sporadic patients are still poorly described. This information is crucial for a targeted therapy. METHODS: We used immunofluorescence microscopy and immunohistochemistry to analyze the expression of endothelial-to-mesenchymal transition markers in the cavernoma of sporadic CCM patients in parallel with human familial cavernoma as a reference control. RESULTS: We report here that endothelial cells, a cell type critically involved in CCM development, undergo endothelial-to-mesenchymal transition in the lesions of sporadic patients. This switch in endothelial phenotype has been described only in genetic CCM patients and in murine models of the disease. In addition, TGF-ß/p-Smad- and ß-catenin-dependent signaling pathways seem activated in sporadic cavernomas as in familial ones. CONCLUSIONS: Our findings support the use of common therapeutic strategies for both sporadic and genetic CCM malformations.


Central Nervous System Neoplasms/pathology , Endothelium, Vascular/pathology , Epithelial-Mesenchymal Transition , Hemangioma, Cavernous, Central Nervous System/pathology , Adolescent , Adult , Aged , Central Nervous System Neoplasms/surgery , Child , Epithelial-Mesenchymal Transition/physiology , Female , Hemangioma, Cavernous, Central Nervous System/surgery , Humans , Male , Middle Aged , Young Adult
20.
J Cell Sci ; 125(Pt 6): 1401-6, 2012 Mar 15.
Article En | MEDLINE | ID: mdl-22328515

The neuropeptide pituitary adenylate cyclase-activating polypeptide 38 (PACAP38) has been implicated in the induction of synaptic plasticity at the excitatory glutamatergic synapse. In particular, recent studies have shown that it is involved in the regulation of N-methyl-D-aspartate (NMDA) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor activation. Here we demonstrate the effect of PACAP38 on the modulation of dendritic spine morphology through a disintegrin and metalloproteinase 10 (ADAM10)-N-cadherin-AMPA receptor signaling pathway. Treatment of primary hippocampal neurons with PACAP38 induced an accumulation of ADAM10 at the postsynaptic membrane. This event led to a significant decrease of dendritic spine head width and to a concomitant reduction of GluR1 colocalization with postsynaptic markers. The PACAP38-induced effect on dendritic spine head width was prevented by either treatment with the ADAM10-specific inhibitor or transfection of a cleavage-defective N-cadherin construct mutated in the ADAM10 cleavage site. Overall, our findings reveal that PACAP38 is involved in the modulation of dendritic spine morphology in hippocampal neurons, and assign to the ADAM10-N-cadherin signaling pathway a crucial role in this modification of the excitatory glutamatergic synapse.


ADAM Proteins/physiology , Cadherins/physiology , Dendritic Spines/metabolism , Hippocampus/metabolism , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Pituitary Adenylate Cyclase-Activating Polypeptide/physiology , Signal Transduction/physiology , ADAM Proteins/antagonists & inhibitors , ADAM10 Protein , Animals , Cadherins/chemistry , Cadherins/genetics , Glutamic Acid/physiology , Hippocampus/cytology , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Primary Cell Culture , Rats
...