Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 56
2.
Cells Dev ; 174: 203841, 2023 06.
Article En | MEDLINE | ID: mdl-37060947

In the adult rodent brain, neural stem cells (NSCs) reside in the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the hippocampus. In these areas, NSCs and their progeny integrate intrinsic signals and extrinsic cues provided by their microenvironment that control their behavior. The vasculature in the SVZ and SGZ, and the choroid plexus (ChP) in the SVZ, have emerged as critical compartments of the neurogenic niches as they provide a rich repertoire of cues to regulate NSC quiescence, proliferation, self-renewal and differentiation. Physical contact between NSCs and blood vessels is also a feature within the niches and supports different processes such as quiescence, migration and vesicle transport. In this review, we provide a description of the brain and choroid plexus vasculature in both stem cell niches, highlighting the main properties and role of the vasculature in each niche. We also summarize the current understanding of how blood vessel- and ChP-derived signals influence the behavior of NSCs in physiological adulthood, as well as upon aging.


Neural Stem Cells , Neural Stem Cells/physiology , Neurogenesis/physiology , Brain , Lateral Ventricles/physiology , Cell Differentiation
3.
Neuron ; 110(24): 4074-4089.e6, 2022 12 21.
Article En | MEDLINE | ID: mdl-36549270

How the vascular and neural compartment cooperate to achieve such a complex and highly specialized structure as the central nervous system is still unclear. Here, we reveal a crosstalk between motor neurons (MNs) and endothelial cells (ECs), necessary for the coordinated development of MNs. By analyzing cell-to-cell interaction profiles of the mouse developing spinal cord, we uncovered semaphorin 3C (Sema3C) and PlexinD1 as a communication axis between MNs and ECs. Using cell-specific knockout mice and in vitro assays, we demonstrate that removal of Sema3C in MNs, or its receptor PlexinD1 in ECs, results in premature and aberrant vascularization of MN columns. Those vascular defects impair MN axon exit from the spinal cord. Impaired PlexinD1 signaling in ECs also causes MN maturation defects at later stages. This study highlights the importance of a timely and spatially controlled communication between MNs and ECs for proper spinal cord development.


Endothelial Cells , Motor Neurons , Animals , Mice , Motor Neurons/physiology , Spinal Cord , Signal Transduction , Axons , Mice, Knockout
4.
EMBO Mol Med ; 14(6): e14121, 2022 06 08.
Article En | MEDLINE | ID: mdl-35491615

The gut has a specific vascular barrier that controls trafficking of antigens and microbiota into the bloodstream. However, the molecular mechanisms regulating the maintenance of this vascular barrier remain elusive. Here, we identified Caspase-8 as a pro-survival factor in mature intestinal endothelial cells that is required to actively maintain vascular homeostasis in the small intestine in an organ-specific manner. In particular, we find that deletion of Caspase-8 in endothelial cells results in small intestinal hemorrhages and bowel inflammation, while all other organs remained unaffected. We also show that Caspase-8 seems to be particularly needed in lymphatic endothelial cells to maintain gut homeostasis. Our work demonstrates that endothelial cell dysfunction, leading to the breakdown of the gut-vascular barrier, is an active driver of chronic small intestinal inflammation, highlighting the role of the intestinal vasculature as a safeguard of organ function.


Caspase 8 , Endothelial Cells , Intestinal Mucosa , Animals , Caspase 8/metabolism , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enteritis/enzymology , Enteritis/pathology , Homeostasis , Intestinal Mucosa/enzymology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/enzymology , Intestine, Small/pathology , Mice
5.
Cell Rep ; 36(7): 109522, 2021 08 17.
Article En | MEDLINE | ID: mdl-34407407

Neuro-vascular communication is essential to synchronize central nervous system development. Here, we identify angiopoietin/Tie2 as a neuro-vascular signaling axis involved in regulating dendritic morphogenesis of Purkinje cells (PCs). We show that in the developing cerebellum Tie2 expression is not restricted to blood vessels, but it is also present in PCs. Its ligands angiopoietin-1 (Ang1) and angiopoietin-2 (Ang2) are expressed in neural cells and endothelial cells (ECs), respectively. PC-specific deletion of Tie2 results in reduced dendritic arborization, which is recapitulated in neural-specific Ang1-knockout and Ang2 full-knockout mice. Mechanistically, RNA sequencing reveals that Tie2-deficient PCs present alterations in gene expression of multiple genes involved in cytoskeleton organization, dendritic formation, growth, and branching. Functionally, mice with deletion of Tie2 in PCs present alterations in PC network functionality. Altogether, our data propose Ang/Tie2 signaling as a mediator of intercellular communication between neural cells, ECs, and PCs, required for proper PC dendritic morphogenesis and function.


Angiopoietin-2/metabolism , Dendrites/metabolism , Morphogenesis , Purkinje Cells/metabolism , Receptor, TIE-2/metabolism , Signal Transduction , Angiopoietin-1/metabolism , Animals , Cerebellum/blood supply , Cerebellum/growth & development , Gene Deletion , Gene Expression Regulation , Integrases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Organ Specificity
6.
Front Cell Dev Biol ; 9: 675562, 2021.
Article En | MEDLINE | ID: mdl-34055807

Angiogenesis is an essential process during development. Abnormal angiogenesis also contributes to many disease conditions such as tumor and retinal diseases. Previous studies have established the Hippo signaling pathway effector Yes-associated protein (YAP) as a crucial regulator of angiogenesis. In ECs, activated YAP promotes endothelial cell proliferation, migration and sprouting. YAP activity is regulated by vascular endothelial growth factor (VEGF) and mechanical cues such as extracellular matrix (ECM) stiffness. However, it is unclear how VEGF or ECM stiffness signal to YAP, especially how dephosphorylation of YAP occurs in response to VEGF stimulus or ECM stiffening. Here, we show that protein phosphatase 2A (PP2A) is required for this process. Blocking PP2A activity abolishes VEGF or ECM stiffening mediated YAP activation. Systemic administration of a PP2A inhibitor suppresses YAP activity in blood vessels in developmental and pathological angiogenesis mouse models. Consistently, PP2A inhibitor also inhibits sprouting angiogenesis. Mechanistically, PP2A directly interacts with YAP, and this interaction requires proper cytoskeleton dynamics. These findings identify PP2A as a crucial mediator of YAP activation in ECs and hence as an important regulator of angiogenesis.

7.
Cell Mol Life Sci ; 78(7): 3247-3264, 2021 Apr.
Article En | MEDLINE | ID: mdl-33783563

The formation of new blood vessels is driven by proliferation of endothelial cells (ECs), elongation of maturing vessel sprouts and ultimately vessel remodeling to create a hierarchically structured vascular system. Vessel regression is an essential process to remove redundant vessel branches in order to adapt the final vessel density to the demands of the surrounding tissue. How exactly vessel regression occurs and whether and to which extent cell death contributes to this process has been in the focus of several studies within the last decade. On top, recent findings challenge our simplistic view of the cell death signaling machinery as a sole executer of cellular demise, as emerging evidences suggest that some of the classic cell death regulators even promote blood vessel formation. This review summarizes our current knowledge on the role of the cell death signaling machinery with a focus on the apoptosis and necroptosis signaling pathways during blood vessel formation in development and pathology.


Blood Vessels/cytology , Cell Death , Endothelium, Vascular/cytology , Neovascularization, Pathologic , Neovascularization, Physiologic , Animals , Humans , Signal Transduction
8.
EMBO Mol Med ; 13(4): e13933, 2021 04 09.
Article En | MEDLINE | ID: mdl-33666362

Transmission of malaria-causing parasites to and by the mosquito relies on active parasite migration and constitutes bottlenecks in the Plasmodium life cycle. Parasite adaption to the biochemically and physically different environments must hence be a key evolutionary driver for transmission efficiency. To probe how subtle but physiologically relevant changes in environmental elasticity impact parasite migration, we introduce 2D and 3D polyacrylamide gels to study ookinetes, the parasite forms emigrating from the mosquito blood meal and sporozoites, the forms transmitted to the vertebrate host. We show that ookinetes adapt their migratory path but not their speed to environmental elasticity and are motile for over 24 h on soft substrates. In contrast, sporozoites evolved more short-lived rapid gliding motility for rapidly crossing the skin. Strikingly, sporozoites are highly sensitive to substrate elasticity possibly to avoid adhesion to soft endothelial cells on their long way to the liver. Hence, the two migratory stages of Plasmodium evolved different strategies to overcome the physical challenges posed by the respective environments and barriers they encounter.


Malaria , Parasites , Plasmodium , Animals , Elasticity , Endothelial Cells , Sporozoites
9.
Nat Neurosci ; 24(4): 478-488, 2021 04.
Article En | MEDLINE | ID: mdl-33510480

Neural-derived signals are crucial regulators of CNS vascularization. However, whether the vasculature responds to these signals by means of elongating and branching or in addition by building a feedback response to modulate neurodevelopmental processes remains unknown. In this study, we identified bidirectional crosstalk between the neural and the vascular compartment of the developing CNS required for oligodendrocyte precursor cell specification. Mechanistically, we show that neural progenitor cells (NPCs) express angiopoietin-1 (Ang1) and that this expression is regulated by Sonic hedgehog. We demonstrate that NPC-derived Ang1 signals to its receptor, Tie2, on endothelial cells to induce the production of transforming growth factor beta 1 (TGFß1). Endothelial-derived TGFß1, in turn, acts as an angiocrine molecule and signals back to NPCs to induce their commitment toward oligodendrocyte precursor cells. This work demonstrates a true bidirectional collaboration between NPCs and the vasculature as a critical regulator of oligodendrogenesis.


Cell Differentiation/physiology , Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , Neurogenesis/physiology , Oligodendrocyte Precursor Cells/cytology , Animals , Mice, Inbred C57BL , Neural Stem Cells/cytology , Oligodendrocyte Precursor Cells/metabolism
10.
Cell Rep ; 33(2): 108256, 2020 10 13.
Article En | MEDLINE | ID: mdl-33053356

Angiogenesis and neurogenesis are tightly coupled during embryonic brain development. However, little is known about how these two processes interact. We show that nascent blood vessels actively contact dividing neural stem cells by endothelial filopodia in the ventricular zone (VZ) of the murine ventral telencephalon; this association is conserved in the human ventral VZ. Using mouse mutants with altered vascular filopodia density, we show that this interaction leads to prolonged cell cycle of apical neural progenitors (ANPs) and favors early neuronal differentiation. Interestingly, pharmacological experiments reveal that ANPs induce vascular filopodia formation by upregulating vascular endothelial growth factor (VEGF)-A in a cell-cycle-dependent manner. This mutual relationship between vascular filopodia and ANPs works as a self-regulatory system that senses ANP proliferation rates and rapidly adjusts neuronal production levels. Our findings indicate a function of vascular filopodia in fine-tuning neural stem cell behavior, which is the basis for proper brain development.


Neural Stem Cells/metabolism , Neurogenesis , Pseudopodia/metabolism , Telencephalon/blood supply , Animals , Cell Cycle , Cell Differentiation , Cell Proliferation , Endothelium, Vascular/metabolism , Humans , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neurons/cytology , Pseudopodia/ultrastructure , Telencephalon/ultrastructure , Time-Lapse Imaging , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
11.
Cancer Cell ; 37(6): 800-817.e7, 2020 06 08.
Article En | MEDLINE | ID: mdl-32516590

Tumors are influenced by the mechanical properties of their microenvironment. Using patient samples and atomic force microscopy, we found that tissue stiffness is higher in liver metastases than in primary colorectal tumors. Highly activated metastasis-associated fibroblasts increase tissue stiffness, which enhances angiogenesis and anti-angiogenic therapy resistance. Drugs targeting the renin-angiotensin system, normally prescribed to treat hypertension, inhibit fibroblast contraction and extracellular matrix deposition, thereby reducing liver metastases stiffening and increasing the anti-angiogenic effects of bevacizumab. Patients treated with bevacizumab showed prolonged survival when concomitantly treated with renin-angiotensin inhibitors, highlighting the importance of modulating the mechanical microenvironment for therapeutic regimens.


Bevacizumab/pharmacology , Cancer-Associated Fibroblasts/drug effects , Colorectal Neoplasms/drug therapy , Drug Synergism , Liver Neoplasms/drug therapy , Neovascularization, Pathologic/prevention & control , Renin-Angiotensin System/drug effects , Angiogenesis Inhibitors/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cancer-Associated Fibroblasts/pathology , Captopril/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Losartan/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Tumor Microenvironment/drug effects
12.
Front Physiol ; 11: 599897, 2020.
Article En | MEDLINE | ID: mdl-33424624

During embryonic central nervous system (CNS) development, the neural and the vascular systems communicate with each other in order to give rise to a fully functional and mature CNS. The initial avascular CNS becomes vascularized by blood vessel sprouting from different vascular plexus in a highly stereotypical and controlled manner. This process is similar across different regions of the CNS. In particular for the developing spinal cord (SC), blood vessel ingression occurs from a perineural vascular plexus during embryonic development. In this review, we provide an updated and comprehensive description of the cellular and molecular mechanisms behind this stereotypical and controlled patterning of blood vessels in the developing embryonic SC, identified using different animal models. We discuss how signals derived from neural progenitors and differentiated neurons guide the SC growing vasculature. Lastly, we provide a perspective of how the molecular mechanisms identified during development could be used to better understand pathological situations.

13.
Elife ; 82019 12 23.
Article En | MEDLINE | ID: mdl-31868583

Axon branching is crucial for proper formation of neuronal networks. Although originally identified as an angiogenic factor, VEGF also signals directly to neurons to regulate their development and function. Here we show that VEGF and its receptor VEGFR2 (also known as KDR or FLK1) are expressed in mouse hippocampal neurons during development, with VEGFR2 locally expressed in the CA3 region. Activation of VEGF/VEGFR2 signaling in isolated hippocampal neurons results in increased axon branching. Remarkably, inactivation of VEGFR2 also results in increased axon branching in vitro and in vivo. The increased CA3 axon branching is not productive as these axons are less mature and form less functional synapses with CA1 neurons. Mechanistically, while VEGF promotes the growth of formed branches without affecting filopodia formation, loss of VEGFR2 increases the number of filopodia and enhances the growth rate of new branches. Thus, a controlled VEGF/VEGFR2 signaling is required for proper CA3 hippocampal axon branching during mouse hippocampus development.


Axons/physiology , Hippocampus/growth & development , Hippocampus/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Ephrin-B2/genetics , Gene Expression Regulation, Developmental , Hippocampus/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Neurogenesis/genetics , Neurogenesis/physiology , Neurons/cytology , Neurons/metabolism , Pseudopodia/metabolism , Signal Transduction/genetics , Synapses/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
14.
Elife ; 82019 12 23.
Article En | MEDLINE | ID: mdl-31868584

Vascular endothelial growth factor (VEGF) is an angiogenic factor that play important roles in the nervous system, although it is still unclear which receptors transduce those signals in neurons. Here, we show that in the developing hippocampus VEGFR2 (also known as KDR or FLK1) is expressed specifically in the CA3 region and it is required for dendritic arborization and spine morphogenesis in hippocampal neurons. Mice lacking VEGFR2 in neurons (Nes-cre Kdrlox/-) show decreased dendritic arbors and spines as well as a reduction in long-term potentiation (LTP) at the associational-commissural - CA3 synapses. Mechanistically, VEGFR2 internalization is required for VEGF-induced spine maturation. In analogy to endothelial cells, ephrinB2 controls VEGFR2 internalization in neurons. VEGFR2-ephrinB2 compound mice (Nes-cre Kdrlox/+ Efnb2lox/+) show reduced dendritic branching, reduced spine head size and impaired LTP. Our results demonstrate the functional crosstalk of VEGFR2 and ephrinB2 in vivo to control dendritic arborization, spine morphogenesis and hippocampal circuitry development.


Dendrites/metabolism , Ephrin-B2/metabolism , Hippocampus/metabolism , Neurogenesis/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , CA3 Region, Hippocampal , Dendritic Spines/metabolism , Endothelial Cells/metabolism , Ephrin-B2/genetics , Gene Expression Regulation, Developmental , Long-Term Potentiation/physiology , Mice , Neurogenesis/genetics , Neuronal Plasticity/physiology , Neurons/physiology , Synapses/physiology , Transcriptome , Vascular Endothelial Growth Factor Receptor-2/genetics
15.
J Clin Invest ; 129(12): 5092-5107, 2019 12 02.
Article En | MEDLINE | ID: mdl-31454332

During developmental angiogenesis, blood vessels grow and remodel to ultimately build a hierarchical vascular network. Whether, how, cell death signaling molecules contribute to blood vessel formation is still not well understood. Caspase-8 (Casp-8), a key protease in the extrinsic cell death-signaling pathway, regulates cell death via both apoptosis and necroptosis. Here, we show that expression of Casp-8 in endothelial cells (ECs) is required for proper postnatal retina angiogenesis. EC-specific Casp-8-KO pups (Casp-8ECKO) showed reduced retina angiogenesis, as the loss of Casp-8 reduced EC proliferation, sprouting, and migration independently of its cell death function. Instead, the loss of Casp-8 caused hyperactivation of p38 MAPK downstream of receptor-interacting serine/threonine protein kinase 3 (RIPK3) and destabilization of vascular endothelial cadherin (VE-cadherin) at EC junctions. In a mouse model of oxygen-induced retinopathy (OIR) resembling retinopathy of prematurity (ROP), loss of Casp-8 in ECs was beneficial, as pathological neovascularization was reduced in Casp-8ECKO pups. Taking these data together, we show that Casp-8 acts in a cell death-independent manner in ECs to regulate the formation of the retina vasculature and that Casp-8 in ECs is mechanistically involved in the pathophysiology of ROP.


Caspase 8/metabolism , Neovascularization, Pathologic , Neovascularization, Physiologic , Retina/embryology , Animals , Animals, Newborn , Antigens, CD/metabolism , Cadherins/metabolism , Cell Death , Cell Movement , Cell Proliferation , Endothelial Cells/metabolism , Female , Human Umbilical Vein Endothelial Cells , Humans , Lung/embryology , Mice , Mice, Knockout , Necroptosis , Oxygen/metabolism , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Clin Cancer Res ; 25(18): 5674-5685, 2019 09 15.
Article En | MEDLINE | ID: mdl-31239322

PURPOSE: To investigate the clinical relevance of macrophages in liver metastasis of colorectal cancer and their influence on angiogenesis and patient survival. Moreover to evaluate specific blood monocytes as markers of disease recurrence.Experimental design: In a mouse model with spontaneous liver metastasis, the angiogenic characteristics of tumor- and metastasis (MAM)-associated macrophages were evaluated. Macrophages and the vasculature from 130 primary tumor (pTU) and 123 patients with liver metastasis were assessed. In vivo and in human samples, the clinical relevance of macrophage VEGFR1 expression was analyzed. Blood samples from patients (n = 157, 80 pTU and 77 liver metastasis) were analyzed for assessing VEGFR1-positive (VEGFR1+) cells as suitable biomarkers of disease recurrence. RESULTS: The number of macrophages positively correlated with vascularization in metastasis. Both in the murine model as well as in primary isolated human cells, a subpopulation of MAMs expressing VEGFR1 were found highly angiogenic. While VEGFR1 expression in pTU patients did not predict prognosis; high percentage of VEGFR1+ cells in liver metastasis was associated with worse patient outcome. Interestingly, VEGFR1+-circulating monocytes in blood samples from patients with liver metastasis not only predicted progression but also site of recurrence. CONCLUSIONS: Our findings identify a new subset of proangiogenic VEGFR1+ MAMs in colorectal cancer that support metastatic growth and may become a liquid biomarker to predict disease recurrence in the liver.


Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Macrophages/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/etiology , Colorectal Neoplasms/mortality , Disease Models, Animal , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Liver Neoplasms/secondary , Macrophages/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Prognosis , Recurrence , Xenograft Model Antitumor Assays
17.
Int J Cancer ; 145(9): 2509-2520, 2019 11 01.
Article En | MEDLINE | ID: mdl-31034094

The immune microenvironment plays a crucial role in supporting tumor growth and metastasis. Tumor-associated macrophages (TAMs) and neutrophils (TANs) are essential components of this microenvironment and affect tumor growth and progression in almost all solid neoplasms. Furthermore, TAMs, TANs and tumor-infiltrating dendritic cells (TIDCs) are found to infiltrate specific distant organs to prepare them as a site for metastatic cell seeding, forming the pre-metastatic niche. The spleen was identified as a major reservoir and source of circulating and tumor infiltrating immune cells. However, discrepancies about its role in supporting tumor growth exist. Thus, here we investigated the role of splenectomy in primary tumor and metastatic growth, and in the formation of an inflammatory niche. In a murine 4T1 and E0771 breast and Panc02 pancreatic cancer model, our results show that while splenectomy reduces the number of infiltrating TAMs, TANs and TIDCs within primary tumors, it does not affect its growth. In line, fewer TAMs, TANs and TIDCs accumulate in the metastatic microenvironment after splenectomy. Interestingly though, this affected metastatic growth depending on the metastatic route/site. The number of hematogenous breast cancer lung metastases was reduced after splenectomy but no effect was observed in breast or pancreatic lymph node metastases. Moreover, we observed that the immune composition of the pre-metastatic niche in lungs of breast cancer bearing mice was altered, and that this could cause the reduction of metastases. Altogether, our results highlight that splenectomy affects the immune microenvironment not only of primary tumors but also of pre-metastatic and metastatic sites.


Inflammation/pathology , Lung Neoplasms/pathology , Lung/pathology , Lymphatic Metastasis/pathology , Spleen/surgery , Animals , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Female , Lymph Nodes/pathology , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neutrophils/pathology , Pancreatic Neoplasms/pathology , Spleen/pathology , Splenectomy/methods , Tumor Microenvironment/physiology
18.
Front Mol Neurosci ; 12: 85, 2019.
Article En | MEDLINE | ID: mdl-31031591

In the central nervous system (CNS), a precise communication between the vascular and neural compartments is essential for proper development and function. Recent studies demonstrate that certain neuronal populations secrete various molecular cues to regulate blood vessel growth and patterning in the spinal cord and brain during development. Interestingly, the vasculature is now emerging as a critical component that regulates stem cell niches during neocortical development, as well as during adulthood. In this review article, we will first provide an overview of blood vessel development and maintenance in embryonic and adult neurogenic niches. We will also summarize the current understanding of how blood vessel-derived signals influence the behavior of neural stem cells (NSCs) during early development as well as in adulthood, with a focus on their metabolism.

19.
J Cell Mol Med ; 22(11): 5244-5256, 2018 11.
Article En | MEDLINE | ID: mdl-30133118

Autophagy and exosome secretion play important roles in a variety of physiological and disease states, including the development of age-related macular degeneration. Previous studies have demonstrated that these cellular mechanisms share common pathways of activation. Low oxidative damage in ARPE-19 cells, alters both autophagy and exosome biogenesis. Moreover, oxidative stress modifies the protein and genetic cargo of exosomes, possibly affecting the fate of surrounding cells. In order to understand the connection between these two mechanisms and their impact on angiogenesis, stressed ARPE-19 cells were treated with a siRNA-targeting Atg7, a key protein for the formation of autophagosomes. Subsequently, we observed the formation of multivesicular bodies and the release of exosomes. Released exosomes contained VEGFR2 as part of their cargo. This receptor for VEGF-which is critical for the development of new blood vessels-was higher in exosome populations released from stressed ARPE-19. While stressed exosomes enhanced tube formation, exosomes became ineffective after silencing VEGFR2 in ARPE-19 cells and were, consequently, unable to influence angiogenesis. Moreover, vessel sprouting in the presence of stressed exosomes seems to follow a VEGF-independent pathway. We propose that abnormal vessel growth correlates with VEGFR2-expressing exosomes release from stressed ARPE-19 cells, and is directly linked to autophagy.


Autophagy/genetics , Macular Degeneration/genetics , Neovascularization, Physiologic/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Autophagosomes/metabolism , Cells, Cultured , Exosomes/genetics , Humans , Macular Degeneration/pathology , Oxidative Stress/genetics , Retinal Pigment Epithelium/metabolism
20.
Cell Rep ; 23(7): 2039-2055, 2018 05 15.
Article En | MEDLINE | ID: mdl-29768203

The progressive deposition of misfolded hyperphosphorylated tau is a pathological hallmark of tauopathies, including Alzheimer's disease. However, the underlying molecular mechanisms governing the intercellular spreading of tau species remain elusive. Here, we show that full-length soluble tau is unconventionally secreted by direct translocation across the plasma membrane. Increased secretion is favored by tau hyperphosphorylation, which provokes microtubule detachment and increases the availability of free protein inside cells. Using a series of binding assays, we show that free tau interacts with components enriched at the inner leaflet of the plasma membrane, finally leading to its translocation across the plasma membrane mediated by sulfated proteoglycans. We provide further evidence that secreted soluble tau species spread trans-cellularly and are sufficient for the induction of intracellular tau aggregation in adjacent cells. Our study demonstrates the mechanistic details of tau secretion and provides insights into the initiation and progression of tau pathology.


tau Proteins/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Chlorocebus aethiops , Cricetulus , Green Fluorescent Proteins/metabolism , Humans , Mice, Inbred C57BL , Neurons/metabolism , Phosphorylation , Protein Aggregates , Protein Binding , Protein Transport , Proteoglycans/metabolism
...