Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nature ; 625(7996): 768-777, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38200313

ABSTRACT

Cerebrospinal fluid (CSF) in the subarachnoid space around the brain has long been known to drain through the lymphatics to cervical lymph nodes1-17, but the connections and regulation have been challenging to identify. Here, using fluorescent CSF tracers in Prox1-GFP lymphatic reporter mice18, we found that the nasopharyngeal lymphatic plexus is a major hub for CSF outflow to deep cervical lymph nodes. This plexus had unusual valves and short lymphangions but no smooth-muscle coverage, whereas downstream deep cervical lymphatics had typical semilunar valves, long lymphangions and smooth muscle coverage that transported CSF to the deep cervical lymph nodes. α-Adrenergic and nitric oxide signalling in the smooth muscle cells regulated CSF drainage through the transport properties of deep cervical lymphatics. During ageing, the nasopharyngeal lymphatic plexus atrophied, but deep cervical lymphatics were not similarly altered, and CSF outflow could still be increased by adrenergic or nitric oxide signalling. Single-cell analysis of gene expression in lymphatic endothelial cells of the nasopharyngeal plexus of aged mice revealed increased type I interferon signalling and other inflammatory cytokines. The importance of evidence for the nasopharyngeal lymphatic plexus functioning as a CSF outflow hub is highlighted by its regression during ageing. Yet, the ageing-resistant pharmacological activation of deep cervical lymphatic transport towards lymph nodes can still increase CSF outflow, offering an approach for augmenting CSF clearance in age-related neurological conditions in which greater efflux would be beneficial.


Subject(s)
Cerebrospinal Fluid , Cervical Vertebrae , Drainage , Lymphatic Vessels , Animals , Mice , Aging/metabolism , Cerebrospinal Fluid/metabolism , Cervical Vertebrae/metabolism , Endothelial Cells/metabolism , Fluorescence , Genes, Reporter , Interferon Type I/immunology , Interferon Type I/metabolism , Lymphatic Vessels/physiology , Myocytes, Smooth Muscle/metabolism , Nitric Oxide/metabolism , Nose/physiology , Pharynx/metabolism , Receptors, Adrenergic, alpha/metabolism , Single-Cell Analysis , Signal Transduction
2.
Sci Immunol ; 8(88): eadg6155, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37801517

ABSTRACT

High neonatal susceptibility to meningitis has been attributed to the anatomical barriers that act to protect the central nervous system (CNS) from infection being immature and not fully developed. However, the mechanisms by which pathogens breach CNS barriers are poorly understood. Using the Armstrong strain of lymphocytic choriomeningitis virus (LCMV) to study virus propagation into the CNS during systemic infection, we demonstrate that mortality in neonatal, but not adult, mice is high after infection. Virus propagated extensively from the perivenous sinus region of the dura mater to the leptomeninges, choroid plexus, and cerebral cortex. Although the structural barrier of CNS border tissues is comparable between neonates and adults, immunofluorescence staining and single-cell RNA sequencing analyses revealed that the neonatal dural immune cells are immature and predominantly composed of CD206hi macrophages, with major histocompatibility complex class II (MHCII)hi macrophages being rare. In adults, however, perivenous sinus immune cells were enriched in MHCIIhi macrophages that are specialized for producing antiviral molecules and chemokines compared with CD206hi macrophages and protected the CNS against systemic virus invasion. Our findings clarify how systemic pathogens enter the CNS through its border tissues and how the immune barrier at the perivenous sinus region of the dura blocks pathogen access to the CNS.


Subject(s)
Encephalitis, Viral , Lymphocytic Choriomeningitis , Meningitis, Viral , Meningoencephalitis , Mice , Animals , Central Nervous System , Meninges , Lymphocytic choriomeningitis virus
3.
Nat Commun ; 14(1): 2754, 2023 05 13.
Article in English | MEDLINE | ID: mdl-37179330

ABSTRACT

Active thermogenesis in the brown adipose tissue (BAT) facilitating the utilization of lipids and glucose is critical for maintaining body temperature and reducing metabolic diseases, whereas inactive BAT accumulates lipids in brown adipocytes (BAs), leading to BAT whitening. Although cellular crosstalk between endothelial cells (ECs) and adipocytes is essential for the transport and utilization of fatty acid in BAs, the angiocrine roles of ECs mediating this crosstalk remain poorly understood. Using single-nucleus RNA sequencing and knock-out male mice, we demonstrate that stem cell factor (SCF) derived from ECs upregulates gene expressions and protein levels of the enzymes for de novo lipogenesis, and promotes lipid accumulation by activating c-Kit in BAs. In the early phase of lipid accumulation induced by denervation or thermoneutrality, transiently expressed c-Kit on BAs increases the protein levels of the lipogenic enzymes via PI3K and AKT signaling. EC-specific SCF deletion and BA-specific c-Kit deletion attenuate the induction of the lipogenic enzymes and suppress the enlargement of lipid droplets in BAs after denervation or thermoneutrality in male mice. These data provide insight into SCF/c-Kit signaling as a regulator that promotes lipid accumulation through the increase of lipogenic enzymes in BAT when thermogenesis is inhibited.


Subject(s)
Adipocytes, Brown , Hypercholesterolemia , Animals , Male , Mice , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Endothelial Cells/metabolism , Fatty Acids/metabolism , Hypercholesterolemia/metabolism , Lipogenesis/genetics , Mice, Knockout , Receptor Protein-Tyrosine Kinases/metabolism , Stem Cell Factor/genetics , Stem Cell Factor/metabolism , Thermogenesis/genetics , Proto-Oncogene Proteins c-kit
4.
Sci Adv ; 8(23): eabn2611, 2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35687678

ABSTRACT

In sprouting angiogenesis, the precise mechanisms underlying how intracellular vascular endothelial growth factor receptor 2 (VEGFR2) signaling is higher in one endothelial cell (EC) compared with its neighbor and acquires the tip EC phenotype under a similar external cue are elusive. Here, we show that Merlin, encoded by the neurofibromatosis type 2 (NF2) gene, suppresses VEGFR2 internalization depending on VE-cadherin density and inhibits tip EC induction. Accordingly, endothelial Nf2 depletion promotes tip EC induction with excessive filopodia by enhancing VEGFR2 internalization in both the growing and matured vessels. Mechanistically, Merlin binds to the VEGFR2-VE-cadherin complex at cell-cell junctions and reduces VEGFR2 internalization-induced downstream signaling during tip EC induction. As a consequence, nonfunctional excessive sprouting occurs during tumor angiogenesis in EC-specific Nf2-deleted mice, leading to delayed tumor growth. Together, Nf2/Merlin is a crucial molecular gatekeeper for tip EC induction, capillary integrity, and proper tumor angiogenesis by suppressing VEGFR2 internalization.

5.
Nat Commun ; 12(1): 4405, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34285232

ABSTRACT

Stimulator of interferon genes (STING) promotes anti-tumour immunity by linking innate and adaptive immunity, but it remains unclear how intratumoural treatment with STING agonists yields anti-tumour effects. Here we demonstrate that intratumoural injection of the STING agonist cGAMP induces strong, rapid, and selective apoptosis of tumour endothelial cells (ECs) in implanted LLC tumour, melanoma and breast tumour, but not in spontaneous breast cancer and melanoma. In both implanted and spontaneous tumours, cGAMP greatly increases TNFα from tumour-associated myeloid cells. However, compared to spontaneous tumour ECs, implanted tumour ECs are more vulnerable to TNFα-TNFR1 signalling-mediated apoptosis, which promotes effective anti-tumour activity. The spontaneous tumour's refractoriness to cGAMP is abolished by co-treatment with AKT 1/2 inhibitor (AKTi). Combined treatment with cGAMP and AKTi induces extensive tumour EC apoptosis, leading to extensive tumour apoptosis and marked growth suppression of the spontaneous tumour. These findings propose an advanced avenue for treating primary tumours that are refractory to single STING agonist therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Membrane Proteins/agonists , Neoplasms/drug therapy , Nucleotides, Cyclic/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Female , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells , Humans , Immunity, Innate/drug effects , Injections, Intralesional , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Neoplasms/blood supply , Neoplasms/immunology , Neoplasms/pathology , Nucleotides, Cyclic/therapeutic use , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/metabolism
6.
J Clin Invest ; 131(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34003804

ABSTRACT

The upper respiratory tract is compromised in the early period of COVID-19, but SARS-CoV-2 tropism at the cellular level is not fully defined. Unlike recent single-cell RNA-Seq analyses indicating uniformly low mRNA expression of SARS-CoV-2 entry-related host molecules in all nasal epithelial cells, we show that the protein levels are relatively high and that their localizations are restricted to the apical side of multiciliated epithelial cells. In addition, we provide evidence in patients with COVID-19 that SARS-CoV-2 is massively detected and replicated within the multiciliated cells. We observed these findings during the early stage of COVID-19, when infected ciliated cells were rapidly replaced by differentiating precursor cells. Moreover, our analyses revealed that SARS-CoV-2 cellular tropism was restricted to the nasal ciliated versus oral squamous epithelium. These results imply that targeting ciliated cells of the nasal epithelium during the early stage of COVID-19 could be an ideal strategy to prevent SARS-CoV-2 propagation.


Subject(s)
COVID-19/virology , Host Microbial Interactions , Nasal Mucosa/virology , SARS-CoV-2 , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/pathology , COVID-19/physiopathology , Cell Differentiation , Cilia/pathology , Cilia/physiology , Cilia/virology , Furin/genetics , Furin/metabolism , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Macaca , Models, Biological , Nasal Mucosa/pathology , Nasal Mucosa/physiopathology , Pandemics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Seq , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Stem Cells/pathology , Stem Cells/virology , Virus Internalization , Virus Replication/genetics , Virus Replication/physiology
7.
Nat Commun ; 11(1): 4102, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32796823

ABSTRACT

Emerging evidence suggests that intestinal stromal cells (IntSCs) play essential roles in maintaining intestinal homeostasis. However, the extent of heterogeneity within the villi stromal compartment and how IntSCs regulate the structure and function of specialized intestinal lymphatic capillary called lacteal remain elusive. Here we show that selective hyperactivation or depletion of YAP/TAZ in PDGFRß+ IntSCs leads to lacteal sprouting or regression with junctional disintegration and impaired dietary fat uptake. Indeed, mechanical or osmotic stress regulates IntSC secretion of VEGF-C mediated by YAP/TAZ. Single-cell RNA sequencing delineated novel subtypes of villi fibroblasts that upregulate Vegfc upon YAP/TAZ activation. These populations of fibroblasts were distributed in proximity to lacteal, suggesting that they constitute a peri-lacteal microenvironment. Our findings demonstrate the heterogeneity of IntSCs and reveal that distinct subsets of villi fibroblasts regulate lacteal integrity through YAP/TAZ-induced VEGF-C secretion, providing new insights into the dynamic regulatory mechanisms behind lymphangiogenesis and lymphatic remodeling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Fibroblasts/metabolism , Intestinal Mucosa/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor C/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Cluster Analysis , Enzyme-Linked Immunosorbent Assay , Fibroblasts/ultrastructure , Flow Cytometry , Fluorescent Antibody Technique , In Situ Hybridization, Fluorescence , Intestinal Mucosa/ultrastructure , Lymphangiogenesis/genetics , Lymphangiogenesis/physiology , Mice , Mice, Inbred C57BL , Microscopy, Electron , Neovascularization, Physiologic/genetics , Neovascularization, Physiologic/physiology , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Vascular Endothelial Growth Factor C/genetics , YAP-Signaling Proteins
8.
Nat Commun ; 11(1): 3866, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737287

ABSTRACT

Upon severe head injury (HI), blood vessels of the meninges and brain parenchyma are inevitably damaged. While limited vascular regeneration of the injured brain has been studied extensively, our understanding of meningeal vascular regeneration following head injury is quite limited. Here, we identify key pathways governing meningeal vascular regeneration following HI. Rapid and complete vascular regeneration in the meninges is predominantly driven by VEGFR2 signaling. Substantial increase of VEGFR2 is observed in both human patients and mouse models of HI, and endothelial cell-specific deletion of Vegfr2 in the latter inhibits meningeal vascular regeneration. We further identify the facilitating, stabilizing and arresting roles of Tie2, PDGFRß and Dll4 signaling, respectively, in meningeal vascular regeneration. Prolonged inhibition of this angiogenic process following HI compromises immunological and stromal integrity of the injured meninges. These findings establish a molecular framework for meningeal vascular regeneration after HI, and may guide development of wound healing therapeutics.


Subject(s)
Craniocerebral Trauma/genetics , Endothelial Cells/metabolism , Neovascularization, Physiologic/genetics , Regeneration/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebrovascular Circulation , Craniocerebral Trauma/metabolism , Craniocerebral Trauma/pathology , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation/genetics , Humans , Macrophages/metabolism , Macrophages/pathology , Meninges/injuries , Meninges/metabolism , Mice , Mice, Knockout , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing/genetics
9.
Nat Commun ; 11(1): 519, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980640

ABSTRACT

Fibroblastic reticular cells (FRCs) are immunologically specialized myofibroblasts of lymphoid organ, and FRC maturation is essential for structural and functional properties of lymph nodes (LNs). Here we show that YAP and TAZ (YAP/TAZ), the final effectors of Hippo signaling, regulate FRC commitment and maturation. Selective depletion of YAP/TAZ in FRCs impairs FRC growth and differentiation and compromises the structural organization of LNs, whereas hyperactivation of YAP/TAZ enhances myofibroblastic characteristics of FRCs and aggravates LN fibrosis. Mechanistically, the interaction between YAP/TAZ and p52 promotes chemokine expression that is required for commitment of FRC lineage prior to lymphotoxin-ß receptor (LTßR) engagement, whereas LTßR activation suppresses YAP/TAZ activity for FRC maturation. Our findings thus present YAP/TAZ as critical regulators of commitment and maturation of FRCs, and hold promise for better understanding of FRC-mediated pathophysiologic processes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Fibroblasts/metabolism , Lymph Nodes/cytology , Trans-Activators/metabolism , Adipocytes/metabolism , Animals , Chemokines/metabolism , Fibroblasts/ultrastructure , Lymph Nodes/ultrastructure , Lymphotoxin beta Receptor/metabolism , Mesoderm/metabolism , Mice, Inbred C57BL , Myofibroblasts/metabolism , YAP-Signaling Proteins
10.
Nat Commun ; 10(1): 838, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30783090

ABSTRACT

Hypoxia is a main driver of sprouting angiogenesis, but how tip endothelial cells are directed to hypoxic regions remains poorly understood. Here, we show that an endothelial MST1-FOXO1 cascade is essential for directional migration of tip cells towards hypoxic regions. In mice, endothelial-specific deletion of either MST1 or FOXO1 leads to the loss of tip cell polarity and subsequent impairment of sprouting angiogenesis. Mechanistically, MST1 is activated by reactive oxygen species (ROS) produced in mitochondria in response to hypoxia, and activated MST1 promotes the nuclear import of FOXO1, thus augmenting its transcriptional regulation of polarity and migration-associated genes. Furthermore, endothelial MST1-FOXO1 cascade is required for revascularization and neovascularization in the oxygen-induced retinopathy model. Together, the results of our study delineate a crucial coupling between extracellular hypoxia and an intracellular ROS-MST1-FOXO1 cascade in establishing endothelial tip cell polarity during sprouting angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Forkhead Box Protein O1/metabolism , Hepatocyte Growth Factor/metabolism , Neovascularization, Physiologic/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Hypoxia , Cell Polarity , Cells, Cultured , Forkhead Box Protein O1/genetics , Gene Expression Regulation , Hepatocyte Growth Factor/genetics , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins , Mice, Inbred C57BL , Mice, Mutant Strains , Neovascularization, Pathologic/metabolism , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Reactive Oxygen Species/metabolism , Retina/cytology , Retina/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...