Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 28
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Angiogenesis ; 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38580869

RÉSUMÉ

In European countries, nearly 10% of all hospital admissions are related to respiratory diseases, mainly chronic life-threatening diseases such as COPD, pulmonary hypertension, IPF or lung cancer. The contribution of blood vessels and angiogenesis to lung regeneration, remodeling and disease progression has been increasingly appreciated. The vascular supply of the lung shows the peculiarity of dual perfusion of the pulmonary circulation (vasa publica), which maintains a functional blood-gas barrier, and the bronchial circulation (vasa privata), which reveals a profiled capacity for angiogenesis (namely intussusceptive and sprouting angiogenesis) and alveolar-vascular remodeling by the recruitment of endothelial precursor cells. The aim of this review is to outline the importance of vascular remodeling and angiogenesis in a variety of non-neoplastic and neoplastic acute and chronic respiratory diseases such as lung infection, COPD, lung fibrosis, pulmonary hypertension and lung cancer.

2.
Biomedicines ; 12(2)2024 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-38397861

RÉSUMÉ

Intussusceptive angiogenesis (IA) and intussusceptive lymphangiogenesis (IL) play a key role in the growth and morphogenesis of vessels. However, there are very few studies in this regard in vessel tumors (VTs). Our objective is to assess the presence, characteristics, and possible mechanisms of the formation of intussusceptive structures in a broad spectrum of VTs. For this purpose, examples of benign and malignant blood and lymphatic VTs were studied via conventional procedures, semithin sections, and immunochemistry and immunofluorescence microscopy. The results demonstrated intussusceptive structures (pillars, meshes, and folds) in benign (lobular capillary hemangioma or pyogenic granuloma, intravascular papillary endothelial hyperplasia or Masson tumor, sinusoidal hemangioma, cavernous hemangioma, glomeruloid hemangioma, angiolipoma, and lymphangiomas), low-grade malignancy (retiform hemangioendothelioma and Dabska tumor), and malignant (angiosarcoma and Kaposi sarcoma) VTs. Intussusceptive structures showed an endothelial cover and a core formed of connective tissue components and presented findings suggesting an origin through vessel loops, endothelialized thrombus, interendothelial bridges, and/or splitting and fusion, and conditioned VT morphology. In conclusion, the findings support the participation of IA and IL, in association with sprouting angiogenesis, in VTs, and therefore in their growth and morphogenesis, which is of pathophysiological interest and lays the groundwork for in-depth molecular studies with therapeutic purposes.

3.
Int J Mol Sci ; 24(23)2023 Nov 24.
Article de Anglais | MEDLINE | ID: mdl-38069025

RÉSUMÉ

Intussusceptive pillars, regarded as a hallmark of intussusceptive angiogenesis, have been described in developing vasculature of many organs and organisms. The aim of this study was to resolve the question about pillar formation and their further maturation employing zebrafish caudal vein plexus (CVP). The CVP development was monitored by in vivo confocal microscopy in high spatio-temporal resolution using the transgenic zebrafish model Fli1a:eGPF//Gata1:dsRed. We tracked back the formation of pillars (diameter ≤ 4 µm) and intercapillary meshes (diameter > 4 µm) and analysed their morphology and behaviour. Transluminal pillars in the CVP arose via a combination of sprouting, lumen expansion, and/or the creation of intraluminal folds, and those mechanisms were not associated directly with blood flow. The follow-up of pillars indicated that one-third of them disappeared between 28 and 48 h post fertilisation (hpf), and of the remaining ones, only 1/17 changed their cross-section area by >50%. The majority of the bigger meshes (39/62) increased their cross-section area by >50%. Plexus simplification and the establishment of hierarchy were dominated by the dynamics of intercapillary meshes, which formed mainly via sprouting angiogenesis. These meshes were observed to grow, reshape, and merge with each other. Our observations suggested an alternative view on intussusceptive angiogenesis in the CVP.


Sujet(s)
Intussusception , Danio zébré , Animaux , Morphogenèse , Hémodynamique , Microscopie intravitale , Néovascularisation physiologique/physiologie
4.
Int J Mol Sci ; 24(4)2023 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-36835203

RÉSUMÉ

Kaposi sarcoma (KS) is an angioproliferative lesion in which two main KS cell sources are currently sustained: endothelial cells (ECs) and mesenchymal/stromal cells. Our objective is to establish the tissue location, characteristics and transdifferentiation steps to the KS cells of the latter. For this purpose, we studied specimens of 49 cases of cutaneous KS using immunochemistry and confocal and electron microscopy. The results showed that delimiting CD34+ stromal cells/Telocytes (CD34+SCs/TCs) in the external layer of the pre-existing blood vessels and around skin appendages form small convergent lumens, express markers for ECs of blood and lymphatic vessels, share ultrastructural characteristics with ECs and participate in the origin of two main types of neovessels, the evolution of which gives rise to lymphangiomatous or spindle-cell patterns-the substrate of the main KS histopathological variants. Intraluminal folds and pillars (papillae) are formed in the neovessels, which suggests they increase by vessel splitting (intussusceptive angiogenesis and intussusceptive lymphangiogenesis). In conclusion, delimiting CD34+SCs/TCs are mesenchymal/stromal cells that can transdifferentiate into KS ECs, participating in the formation of two types of neovessels. The subsequent growth of the latter involves intussusceptive mechanisms, originating several KS variants. These findings are of histogenic, clinical and therapeutic interest.


Sujet(s)
Sarcome de Kaposi , Tumeurs cutanées , Cellules stromales , Télocytes , Humains , Antigènes CD34/métabolisme , Cellules endothéliales/métabolisme , Sarcome de Kaposi/anatomopathologie , Tumeurs cutanées/anatomopathologie , Cellules stromales/métabolisme , Cellules stromales/anatomopathologie , Télocytes/métabolisme , Télocytes/anatomopathologie , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie
5.
Angiogenesis ; 26(2): 233-248, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36371548

RÉSUMÉ

A wide range of cardiac symptoms have been observed in COVID-19 patients, often significantly influencing the clinical outcome. While the pathophysiology of pulmonary COVID-19 manifestation has been substantially unraveled, the underlying pathomechanisms of cardiac involvement in COVID-19 are largely unknown. In this multicentre study, we performed a comprehensive analysis of heart samples from 24 autopsies with confirmed SARS-CoV-2 infection and compared them to samples of age-matched Influenza H1N1 A (n = 16), lymphocytic non-influenza myocarditis cases (n = 8), and non-inflamed heart tissue (n = 9). We employed conventional histopathology, multiplexed immunohistochemistry (MPX), microvascular corrosion casting, scanning electron microscopy, X-ray phase-contrast tomography using synchrotron radiation, and direct multiplexed measurements of gene expression, to assess morphological and molecular changes holistically. Based on histopathology, none of the COVID-19 samples fulfilled the established diagnostic criteria of viral myocarditis. However, quantification via MPX showed a significant increase in perivascular CD11b/TIE2 + -macrophages in COVID-19 over time, which was not observed in influenza or non-SARS-CoV-2 viral myocarditis patients. Ultrastructurally, a significant increase in intussusceptive angiogenesis as well as multifocal thrombi, inapparent in conventional morphological analysis, could be demonstrated. In line with this, on a molecular level, COVID-19 hearts displayed a distinct expression pattern of genes primarily coding for factors involved in angiogenesis and epithelial-mesenchymal transition (EMT), changes not seen in any of the other patient groups. We conclude that cardiac involvement in COVID-19 is an angiocentric macrophage-driven inflammatory process, distinct from classical anti-viral inflammatory responses, and substantially underappreciated by conventional histopathologic analysis. For the first time, we have observed intussusceptive angiogenesis in cardiac tissue, which we previously identified as the linchpin of vascular remodeling in COVID-19 pneumonia, as a pathognomic sign in affected hearts. Moreover, we identified CD11b + /TIE2 + macrophages as the drivers of intussusceptive angiogenesis and set forward a putative model for the molecular regulation of vascular alterations.


Sujet(s)
COVID-19 , Sous-type H1N1 du virus de la grippe A , Myocardite , Humains , Remodelage vasculaire , SARS-CoV-2 , Inflammation
6.
EBioMedicine ; 85: 104296, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36206625

RÉSUMÉ

BACKGROUND: COVID-19 is characterized by a heterogeneous clinical presentation, ranging from mild symptoms to severe courses of disease. 9-20% of hospitalized patients with severe lung disease die from COVID-19 and a substantial number of survivors develop long-COVID. Our objective was to provide comprehensive insights into the pathophysiology of severe COVID-19 and to identify liquid biomarkers for disease severity and therapy response. METHODS: We studied a total of 85 lungs (n = 31 COVID autopsy samples; n = 7 influenza A autopsy samples; n = 18 interstitial lung disease explants; n = 24 healthy controls) using the highest resolution Synchrotron radiation-based hierarchical phase-contrast tomography, scanning electron microscopy of microvascular corrosion casts, immunohistochemistry, matrix-assisted laser desorption ionization mass spectrometry imaging, and analysis of mRNA expression and biological pathways. Plasma samples from all disease groups were used for liquid biomarker determination using ELISA. The anatomic/molecular data were analyzed as a function of patients' hospitalization time. FINDINGS: The observed patchy/mosaic appearance of COVID-19 in conventional lung imaging resulted from microvascular occlusion and secondary lobular ischemia. The length of hospitalization was associated with increased intussusceptive angiogenesis. This was associated with enhanced angiogenic, and fibrotic gene expression demonstrated by molecular profiling and metabolomic analysis. Increased plasma fibrosis markers correlated with their pulmonary tissue transcript levels and predicted disease severity. Plasma analysis confirmed distinct fibrosis biomarkers (TSP2, GDF15, IGFBP7, Pro-C3) that predicted the fatal trajectory in COVID-19. INTERPRETATION: Pulmonary severe COVID-19 is a consequence of secondary lobular microischemia and fibrotic remodelling, resulting in a distinctive form of fibrotic interstitial lung disease that contributes to long-COVID. FUNDING: This project was made possible by a number of funders. The full list can be found within the Declaration of interests / Acknowledgements section at the end of the manuscript.


Sujet(s)
COVID-19 , Pneumopathies interstitielles , Humains , Poumon/imagerie diagnostique , Poumon/anatomopathologie , Pneumopathies interstitielles/anatomopathologie , Fibrose , Marqueurs biologiques/analyse , Ischémie/anatomopathologie , Syndrome de post-COVID-19
7.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article de Anglais | MEDLINE | ID: mdl-36012273

RÉSUMÉ

Perivascular cells in the pericytic microvasculature, pericytes and CD34+ stromal cells/telocytes (CD34+SCs/TCs), have an important role in angiogenesis. We compare the behavior of these cells depending on whether the growth of endothelial cells (ECs) from the pre-existing microvasculature is toward the interstitium with vascular bud and neovessel formation (sprouting angiogenesis) or toward the vascular lumen with intravascular pillar development and vessel division (intussusceptive angiogenesis). Detachment from the vascular wall, mobilization, proliferation, recruitment, and differentiation of pericytes and CD34+SCs/TCs, as well as associated changes in vessel permeability and functionality, and modifications of the extracellular matrix are more intense, longer lasting over time, and with a greater energy cost in sprouting angiogenesis than in intussusceptive angiogenesis, in which some of the aforementioned events do not occur or are compensated for by others (e.g., sparse EC and pericyte proliferation by cell elongation and thinning). The governing mechanisms involve cell-cell contacts (e.g., peg-and-socket junctions between pericytes and ECs), multiple autocrine and paracrine signaling molecules and pathways (e.g., vascular endothelial growth factor, platelet-derived growth factor, angiopoietins, transforming growth factor B, ephrins, semaphorins, and metalloproteinases), and other factors (e.g., hypoxia, vascular patency, and blood flow). Pericytes participate in vessel development, stabilization, maturation and regression in sprouting angiogenesis, and in interstitial tissue structure formation of the pillar core in intussusceptive angiogenesis. In sprouting angiogenesis, proliferating perivascular CD34+SCs/TCs are an important source of stromal cells during repair through granulation tissue formation and of cancer-associated fibroblasts (CAFs) in tumors. Conversely, CD34+SCs/TCs have less participation as precursor cells in intussusceptive angiogenesis. The dysfunction of these mechanisms is involved in several diseases, including neoplasms, with therapeutic implications.


Sujet(s)
Péricytes , Télocytes , Antigènes CD34/métabolisme , Cellules endothéliales/métabolisme , Néovascularisation physiologique/physiologie , Péricytes/métabolisme , Cellules stromales/métabolisme , Télocytes/métabolisme , Facteur de croissance endothéliale vasculaire de type A/analyse
8.
Cardiovasc Res ; 118(5): 1262-1275, 2022 03 25.
Article de Anglais | MEDLINE | ID: mdl-33909875

RÉSUMÉ

AIMS: Hepatic capillaries are lined with specialized liver sinusoidal endothelial cells (LSECs) which support macromolecule passage to hepatocytes and prevent fibrosis by keeping hepatic stellate cells (HSCs) quiescent. LSEC specialization is co-determined by transcription factors. The zinc-finger E-box-binding homeobox (Zeb)2 transcription factor is enriched in LSECs. Here, we aimed to elucidate the endothelium-specific role of Zeb2 during maintenance of the liver and in liver fibrosis. METHODS AND RESULTS: To study the role of Zeb2 in liver endothelium we generated EC-specific Zeb2 knock-out (ECKO) mice. Sequencing of liver EC RNA revealed that deficiency of Zeb2 results in prominent expression changes in angiogenesis-related genes. Accordingly, the vascular area was expanded and the presence of pillars inside ECKO liver vessels indicated that this was likely due to increased intussusceptive angiogenesis. LSEC marker expression was not profoundly affected and fenestrations were preserved upon Zeb2 deficiency. However, an increase in continuous EC markers suggested that Zeb2-deficient LSECs are more prone to dedifferentiation, a process called 'capillarization'. Changes in the endothelial expression of ligands that may be involved in HSC quiescence together with significant changes in the expression profile of HSCs showed that Zeb2 regulates LSEC-HSC communication and HSC activation. Accordingly, upon exposure to the hepatotoxin carbon tetrachloride (CCl4), livers of ECKO mice showed increased capillarization, HSC activation, and fibrosis compared to livers from wild-type littermates. The vascular maintenance and anti-fibrotic role of endothelial Zeb2 was confirmed in mice with EC-specific overexpression of Zeb2, as the latter resulted in reduced vascularity and attenuated CCl4-induced liver fibrosis. CONCLUSION: Endothelial Zeb2 preserves liver angioarchitecture and protects against liver fibrosis. Zeb2 and Zeb2-dependent genes in liver ECs may be exploited to design novel therapeutic strategies to attenuate hepatic fibrosis.


Sujet(s)
Cellules endothéliales , Cirrhose du foie , Animaux , Marqueurs biologiques/métabolisme , Cellules endothéliales/métabolisme , Endothélium , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Foie/métabolisme , Cirrhose du foie/induit chimiquement , Cirrhose du foie/génétique , Cirrhose du foie/prévention et contrôle , Souris
9.
Cytokine Growth Factor Rev ; 63: 69-77, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34728151

RÉSUMÉ

Coronavirus disease-2019 (COVID-19), the disease caused by severe acute respiratory syndrome-coronavirus-2, has claimed more than 4.4 million lives worldwide (as of 20 August 2021). Severe cases of the disease often result in respiratory distress due to cytokine storm, and mechanical ventilation is required. Although, the lungs are the primary organs affected by the disease, more evidence on damage to the heart, kidney, and liver is emerging. A common link in these connections is the cardiovascular network. Inner lining of the blood vessels, called endothelium, is formed by a single layer of endothelial cells. Several clinical manifestations involving the endothelium have been reported, such as its activation via immunomodulation, endotheliitis, thrombosis, vasoconstriction, and distinct intussusceptive angiogenesis (IA), a unique and rapid process of blood-vessel formation by splitting a vessel into two lumens. In fact, the virus directly infects the endothelium via TMPRSS2 spike glycoprotein priming to facilitate ACE-2-mediated viral entry. Recent studies have indicated a significant increase in remodeling of the pulmonary vascular bed via intussusception in patients with COVID-19. However, the lack of circulatory biomarkers for IA limits its detection in COVID-19 pathogenesis. In this review, we describe the implications of angiogenesis in COVID-19, unique features of the pulmonary vascular bed and its remodeling, and a rapid and non-invasive assessment of IA to overcome the technical limitations in patients with COVID-19.


Sujet(s)
COVID-19 , Cellules endothéliales , Endothélium , Endothélium vasculaire/anatomopathologie , Humains , Poumon/anatomopathologie , SARS-CoV-2 , Remodelage vasculaire
10.
Int J Mol Sci ; 22(23)2021 Nov 30.
Article de Anglais | MEDLINE | ID: mdl-34884806

RÉSUMÉ

Platelets in atherosclerosis, bypass stenosis, and restenosis have been extensively assessed. However, a sequential ultrastructural study of platelets in angiogenesis during the early phases of these lesions has received less attention. Our objective was the study of platelets in angiogenesis and vessel regression during intimal thickening (IT) formation, a precursor process of these occlusive vascular diseases. For this purpose, we used an experimental model of rat occluded arteries and procedures for ultrastructural observation. The results show (a) the absence of platelet adhesion in the de-endothelialized occluded arterial segment isolated from the circulation, (b) that intraarterial myriad platelets contributed from neovessels originated by sprouting angiogenesis from the periarterial microvasculature, (c) the association of platelets with blood components (fibrin, neutrophils, macrophages, and eosinophils) and non-polarized endothelial cells (ECs) forming aggregates (spheroids) in the arterial lumen, (d) the establishment of peg-and-socket junctions between platelets and polarized Ecs during intussusceptive angiogenesis originated from the EC aggregates, with the initial formation of IT, and (e) the aggregation of platelets in regressing neovessels ('transitory paracrine organoid') and IT increases. In conclusion, in sprouting and intussusceptive angiogenesis and vessel regression during IT formation, we contribute sequential ultrastructural findings on platelet behavior and relationships, which can be the basis for further studies using other procedures.


Sujet(s)
Artères/anatomopathologie , Plaquettes/métabolisme , Néovascularisation pathologique/anatomopathologie , Adhésivité plaquettaire/physiologie , Tunique intime/anatomopathologie , Animaux , Artères/ultrastructure , Athérosclérose/anatomopathologie , Resténose coronaire/anatomopathologie , Rats , Rat Sprague-Dawley , Tunique intime/ultrastructure , Remodelage vasculaire/physiologie
11.
Cells ; 10(10)2021 10 01.
Article de Anglais | MEDLINE | ID: mdl-34685606

RÉSUMÉ

Glioblastoma (GBM) is the most malignant tumor in the brain. In addition to the vascular pattern with thin-walled vessels and findings of sprouting angiogenesis, GBM presents a bizarre microvasculature (BM) formed by vascular clusters, vascular garlands, and glomeruloid bodies. The mechanisms in BM morphogenesis are not well known. Our objective was to assess the role of pericyte/endothelial proliferation and intussusceptive angiogenic mechanisms in the formation of the BM. For this purpose, we studied specimens of 66 GBM cases using immunochemistry and confocal microscopy. In the BM, the results showed (a) transitional forms between the BM patterns, mostly with prominent pericytes covering all the abluminal endothelial cell (EC) surface of the vessels, (b) a proliferation index high in the prominent pericytes and low in ECs (47.85 times higher in pericytes than in ECs), (c) intravascular pillars (hallmark of intussusceptive angiogenesis) formed by transcapillary interendothelial bridges, endothelial contacts of opposite vessel walls, and vessel loops, and (d) the persistence of these findings in complex glomeruloid bodies. In conclusion, disproportion in pericyte/EC proliferation and mechanisms of intussusceptive angiogenesis participate in BM formation. The contributions have morphogenic and clinical interest since pericytes and intussusceptive angiogenesis can condition antiangiogenic therapy in GBM.


Sujet(s)
Cellules endothéliales/anatomopathologie , Glioblastome/vascularisation , Glioblastome/anatomopathologie , Néovascularisation pathologique/anatomopathologie , Péricytes/anatomopathologie , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Prolifération cellulaire , Femelle , Humains , Mâle , Microvaisseaux/anatomopathologie , Adulte d'âge moyen , Névroglie/anatomopathologie , Jeune adulte
12.
Biochem Biophys Res Commun ; 570: 89-95, 2021 09 17.
Article de Anglais | MEDLINE | ID: mdl-34274851

RÉSUMÉ

Eribulin is a novel microtubule inhibitor that, similar to other types of microtubule inhibitors, induces apoptosis by inhibiting the mitotic division of cells. Besides this direct effect on tumor cells, previous studies have shown that eribulin has the potential to induce tumor vascular remodeling in several different cancers; however, the mechanisms underlying this phenomenon remain unclear. In the present study, we aimed to elucidate whether eribulin is effective against synovial sarcoma, a relatively rare sarcoma that often affects adolescents and young adults, and to histologically investigate the microstructure of tumor vessels after the administration of eribulin. We found that eribulin exhibits potent antitumor activity against synovial sarcoma in a tumor xenograft model and that tumor vessels frequently have intervascular pillars, a hallmark of intussusceptive angiogenesis (IA), after the administration of eribulin. IA is a distinct form of angiogenesis that is involved in normal developmental processes as well as pathological conditions. Our data indicate that IA is potentially involved in eribulin-induced vascular remodeling and thereby suggest previously unacknowledged role of IA in regulating the tumor vasculature after eribulin administration.


Sujet(s)
Furanes/usage thérapeutique , Intussusception/complications , Cétones/usage thérapeutique , Néovascularisation pathologique/traitement médicamenteux , Sarcomes/vascularisation , Sarcomes/traitement médicamenteux , Remodelage vasculaire , Animaux , Bévacizumab/pharmacologie , Bévacizumab/usage thérapeutique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Forme de la cellule/effets des médicaments et des substances chimiques , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/ultrastructure , Furanes/administration et posologie , Furanes/pharmacologie , Intussusception/traitement médicamenteux , Cétones/administration et posologie , Cétones/pharmacologie , Souris de lignée BALB C , Souris nude , Néovascularisation pathologique/complications , Péricytes/effets des médicaments et des substances chimiques , Péricytes/anatomopathologie , Péricytes/ultrastructure , Sarcomes/complications , Sarcomes/ultrastructure , Hypoxie tumorale/effets des médicaments et des substances chimiques , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Remodelage vasculaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
13.
Angiogenesis ; 24(1): 57-65, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33037487

RÉSUMÉ

Cirrhosis describes the development of excess fibrous tissue around regenerative nodules in response to chronic liver injury and usually leads to irreversible organ damage and end-stage liver disease. During the development of cirrhosis, the formation of collagenous scar tissue is paralleled by a reorganization and remodeling of the hepatic vascular system. To date, macrovascular remodeling in various cirrhosis models has been examined using three-dimensional (3D) imaging modalities, while microvascular changes have been studied mainly by two-dimensional (2D) light microscopic and electron microscopic imaging. Here, we report on the application of high-resolution 3D synchrotron radiation-based microtomography (SRµCT) for the study of the sinusoidal and capillary blood vessel system in three murine models of advanced parenchymal and biliary hepatic fibrosis. SRµCT facilitates the characterization of microvascular architecture and identifies features of intussusceptive angiogenesis in progressive liver fibrosis in a non-destructive 3D manner.


Sujet(s)
Imagerie tridimensionnelle , Cirrhose du foie/imagerie diagnostique , Microvaisseaux/imagerie diagnostique , Synchrotrons , Microtomographie aux rayons X , Animaux , Modèles animaux de maladie humaine , Souris de lignée C57BL
14.
Int J Mol Sci ; 21(21)2020 Oct 28.
Article de Anglais | MEDLINE | ID: mdl-33126763

RÉSUMÉ

Angiogenesis in arterial intimal thickening (AIT) has been considered mainly in late AIT stages and only refers to sprouting angiogenesis. We assess angiogenesis during early AIT development and the occurrence of the intussusceptive type. For this purpose, we studied AIT development in (a) human arteries with vasculitis in gallbladders with acute cholecystitis and urgent (n = 25) or delayed (n = 20) cholecystectomy, using immunohistochemical techniques and (b) experimentally occluded arterial segments (n = 56), using semithin and ultrathin sections and electron microscopy. The results showed transitory angiogenic phenomena, with formation of an important microvasculature, followed by vessel regression. In addition to the sequential description of angiogenic and regressive findings, we mainly contribute (a) formation of intravascular pillars (hallmarks of intussusception) during angiogenesis and vessel regression and (b) morphological interrelation between endothelial cells (ECs) in the arterial wall and vascular smooth muscle cells (VSMCs), which adopt a pericytic arrangement and establish peg-and-socket junctions with ECs. In conclusion, angiogenesis and vessel regression play an important role in AIT development in the conditions studied, with participation of intussusceptive angiogenesis during the formation and regression of a provisional microvasculature and with morphologic interrelation between ECs and VSMCs.


Sujet(s)
Artères/anatomopathologie , Cholécystite aigüe/anatomopathologie , Endothélium vasculaire/anatomopathologie , Vésicule biliaire/vascularisation , Myocytes du muscle lisse/anatomopathologie , Néovascularisation pathologique/anatomopathologie , Tunique intime/anatomopathologie , Adulte , Sujet âgé , Animaux , Phénomènes physiologiques cardiovasculaires , Cholécystite aigüe/chirurgie , Femelle , Artère fémorale/anatomopathologie , Vésicule biliaire/anatomopathologie , Humains , Mâle , Adulte d'âge moyen , Rats
15.
Life Sci ; 252: 117670, 2020 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-32298741

RÉSUMÉ

Deregulation of angiogenesis is a key reason for tumor growth and progression. Several anti-angiogenic drugs in clinical practice attempt to normalize abnormal tumor vasculature. Unfortunately, these drugs are ineffective due to the development of resistance in patients after drug holidays. A sizable literature suggests that resistance to these anti-angiogenic drugs occurs due to various compensatory mechanisms of tumor angiogenesis. Therefore, we describe different compensatory mechanisms of tumor angiogenesis, and explain why intussusceptive angiogenesis (IA), is a crucial mechanism of compensatory angiogenesis in tumors which resist anti-VEGF (vascular endothelial growth factor) therapies. IA is often overlooked due to the scarcity of experimental models. Therefore, we examine data from existing experimental models and our novel ex-ovo model of angiogenesis in chick embryos, and explain the important genes and signaling pathways driving IA. Using bio-informatic analyses of major genes regulating conventional sprouting angiogenesis (SA) and intussusceptive angiogenesis, we provide fresh insights on the 'angiogenic switch' which regulates the transition from SA to IA. Finally, we examine the interplay between molecules regulating SA, IA, and molecules known to promote tumor progression. Based on these analyses, we conclude that intussusceptive angiogenesis (IA) is a promising therapeutic target for developing effective anti-cancer treatment regimes.


Sujet(s)
Inhibiteurs de l'angiogenèse/pharmacologie , Tumeurs/traitement médicamenteux , Néovascularisation pathologique/traitement médicamenteux , Inhibiteurs de l'angiogenèse/administration et posologie , Animaux , Embryon de poulet , Évolution de la maladie , Résistance aux médicaments antinéoplasiques , Humains , Tumeurs/vascularisation , Néovascularisation pathologique/anatomopathologie , Facteur de croissance endothéliale vasculaire de type A/antagonistes et inhibiteurs
16.
Biomolecules ; 10(3)2020 03 23.
Article de Anglais | MEDLINE | ID: mdl-32210087

RÉSUMÉ

Vascular development is an orchestrated process of vessel formation from pre-existing vessels via sprouting and intussusceptive angiogenesis as well as vascular remodeling to generate the mature vasculature. Bone morphogenetic protein (BMP) signaling via intracellular SMAD1 and SMAD5 effectors regulates sprouting angiogenesis in the early mouse embryo, but its role in other processes of vascular development and in other vascular beds remains incompletely understood. Here, we investigate the function of SMAD1/5 during early postnatal retinal vascular development using inducible, endothelium-specific deletion of Smad1 and Smad5. We observe the formation of arterial-venous malformations in areas with high blood flow, and fewer and less functional tip cells at the angiogenic front. The vascular plexus region is remarkably hyperdense and this is associated with reduced vessel regression and aberrant vascular loop formation. Taken together, our results highlight important functions of SMAD1/5 during vessel formation and remodeling in the early postnatal retina.


Sujet(s)
Protéines morphogénétiques osseuses/métabolisme , Embryon de mammifère , Néovascularisation physiologique , Rétine/embryologie , Vaisseaux rétiniens/embryologie , Transduction du signal , Protéine Smad-1/métabolisme , Protéine Smad-5/métabolisme , Animaux , Protéines morphogénétiques osseuses/génétique , Embryon de mammifère/vascularisation , Embryon de mammifère/embryologie , Souris , Souris transgéniques , Protéine Smad-1/génétique , Protéine Smad-5/génétique
17.
EMBO Mol Med ; 12(2): e10862, 2020 02 07.
Article de Anglais | MEDLINE | ID: mdl-31793743

RÉSUMÉ

Pathological angiogenesis contributes to cancer progression and chronic inflammatory diseases. In inflammatory bowel disease, the microvasculature expands by intussusceptive angiogenesis (IA), a poorly characterized mechanism involving increased blood flow and splitting of pre-existing capillaries. In this report, mice lacking the protease MT1-MMP in endothelial cells (MT1iΔEC ) presented limited IA in the capillary plexus of the colon mucosa assessed by 3D imaging during 1% DSS-induced colitis. This resulted in better tissue perfusion, preserved intestinal morphology, and milder disease activity index. Combined in vivo intravital microscopy and lentiviral rescue experiments with in vitro cell culture demonstrated that MT1-MMP activity in endothelial cells is required for vasodilation and IA, as well as for nitric oxide production via binding of the C-terminal fragment of MT1-MMP substrate thrombospondin-1 (TSP1) to CD47/αvß3 integrin. Moreover, TSP1 levels were significantly higher in serum from IBD patients and in vivo administration of an anti-MT1-MMP inhibitory antibody or a nonamer peptide spanning the αvß3 integrin binding site in TSP1 reduced IA during mouse colitis. Our results identify MT1-MMP as a new actor in inflammatory IA and a promising therapeutic target for inflammatory bowel disease.


Sujet(s)
Colite , Matrix metalloproteinase 14 , Monoxyde d'azote/métabolisme , Thrombospondine-1 , Animaux , Colite/métabolisme , Colite/anatomopathologie , Cellules endothéliales , Humains , Intussusception , Matrix metalloproteinase 14/métabolisme , Souris , Souris de lignée C57BL , Néovascularisation pathologique , Thrombospondine-1/métabolisme
18.
J Cell Mol Med ; 23(6): 3916-3926, 2019 06.
Article de Anglais | MEDLINE | ID: mdl-30950188

RÉSUMÉ

The precise mechanisms of SDF-1 (CXCL12) in angiogenesis are not fully elucidated. Recently, we showed that Notch inhibition induces extensive intussusceptive angiogenesis by recruitment of mononuclear cells and it was associated with increased levels of SDF-1 and CXCR4. In the current study, we demonstrated SDF-1 expression in liver sinusoidal vessels of Notch1 knockout mice with regenerative hyperplasia by means of intussusception, but we did not detect any SDF-1 expression in wild-type mice with normal liver vessel structure. In addition, pharmacological inhibition of SDF-1/CXCR4 signalling by AMD3100 perturbs intussusceptive vascular growth and abolishes mononuclear cell recruitment in the chicken area vasculosa. In contrast, treatment with recombinant SDF-1 protein increased microvascular density by 34% through augmentation of pillar number compared to controls. The number of extravasating mononuclear cells was four times higher after SDF-1 application and two times less after blocking this pathway. Bone marrow-derived mononuclear cells (BMDC) were recruited to vessels in response to elevated expression of SDF-1 in endothelial cells. They participated in formation and stabilization of pillars. The current study is the first report to implicate SDF-1/CXCR4 signalling in intussusceptive angiogenesis and further highlights the stabilizing role of BMDC in the formation of pillars during vascular remodelling.


Sujet(s)
Chimiokine CXCL12/métabolisme , Intussusception/métabolisme , Néovascularisation pathologique/métabolisme , Récepteur Notch1/métabolisme , Récepteurs CXCR4/métabolisme , Animaux , Benzylamines , Cellules de la moelle osseuse/métabolisme , Adhérence cellulaire/génétique , Chimiokine CXCL12/génétique , Embryon de poulet , Cyclames , Cellules endothéliales/métabolisme , Cellules endothéliales/ultrastructure , Hépatocytes/métabolisme , Composés hétérocycliques/pharmacologie , Intussusception/génétique , Agranulocytes/métabolisme , Agranulocytes/ultrastructure , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Microscopie électronique à transmission , Néovascularisation pathologique/imagerie diagnostique , Néovascularisation pathologique/génétique , Récepteur Notch1/antagonistes et inhibiteurs , Récepteur Notch1/génétique , Récepteurs CXCR4/antagonistes et inhibiteurs , Récepteurs CXCR4/génétique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Transduction du signal/génétique
19.
Int J Biol Macromol ; 123: 1229-1240, 2019 Feb 15.
Article de Anglais | MEDLINE | ID: mdl-30468812

RÉSUMÉ

Angiogenesis is a distinct process which follows sprouting angiogenesis (SA) and intussusceptive angiogenesis (IA) forming the basis for various physiological and pathological scenarios. Angiogenesis is a double edged sword exerting both desirable and discernible effects owing to the referred microenvironment. Therapeutic interventions to promote angiogenesis in regenerative medicine is essential to achieve functional syncytium of tissue constructs while, angiogenic inhibition is a key therapeutic target to suppress tumor growth. In the recent years, clustered regularly interspaced short palindromic repeats associated 9 (CRISPR-Cas9) based gene editing approaches have been gaining considerable attention in the field of biomedical research owing to its ease in tailoring targeted genome in living organisms. The Zebrafish model, with adequately high-throughput fitness, is a likely option for genome editing and angiogenesis research. In this review, we focus on the implication of Zebrafish as a model to study IA and furthermore enumerate CRISPR/Cas9 based genome editing in Zebrafish as a candidate for modeling different types of angiogenesis and support its candidature as a model organism.


Sujet(s)
Protéine-9 associée à CRISPR/métabolisme , Systèmes CRISPR-Cas/génétique , Modèles biologiques , Néovascularisation physiologique , Danio zébré/métabolisme , Animaux , Animal génétiquement modifié
20.
Nitric Oxide ; 82: 48-58, 2019 01 01.
Article de Anglais | MEDLINE | ID: mdl-30439561

RÉSUMÉ

Angiogenesis is the formation of new blood vessels that occurs by two distinct processes following sprouting angiogenesis (SA) and intussusceptive angiogenesis (IA). Nitric oxide (NO) is known for its pro-angiogenic functions. However, no clear mechanisms are delineated on its role in promoting angiogenesis in reparative wound healing. We propose that NO regulates SA to IA transition and vice versa in wound milieu. We have used three models which include a new chick embryo extra-vasculature (CEV) burn wound model, adult Tie2-GFP transgenic Zebrafish caudal fin regeneration model and Zebrafish skin wound model to study the mechanisms underlying behind the role of NO in wound healing. Wounds created in CEV were treated with NO donor (Spermine NONOate (SPNO)), NOS inhibitor (L-nitro-l-arginine-methyl ester (l-NAME)), NaNO2, NaNO3, and beetroot juice, a nitrite-rich juice respectively and the pattern of wound healing was assessed. Morphological and histological techniques tracked the wound healing at the cellular level, and the molecular changes were investigated by using real-time RT-PCR gene expression analysis. The result concludes that NO donor promotes wound healing by activating SA at an early phase of healing while NOS inhibitor induces wound healing via IA. At the later phase of wound healing NO donor followed IA while NOS inhibitor failed to promote wound repair. The current work underpinned a differential regulation of NO on angiogenesis in wound milieu and this study would provide new insights in designing therapeutics for promoting wound repair.


Sujet(s)
Néovascularisation pathologique/traitement médicamenteux , Monoxyde d'azote/pharmacologie , Cicatrisation de plaie/effets des médicaments et des substances chimiques , Danio zébré/génétique , Danio zébré/physiologie , Animaux , Animal génétiquement modifié , Embryon de poulet , Modèles animaux de maladie humaine , Néovascularisation pathologique/métabolisme , Néovascularisation pathologique/anatomopathologie , Monoxyde d'azote/métabolisme , Danio zébré/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...