Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.100
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 597-620, 2020 04 26.
Article in English | MEDLINE | ID: mdl-32340575

ABSTRACT

Neuroimmunology, albeit a relatively established discipline, has recently sparked numerous exciting findings on microglia, the resident macrophages of the central nervous system (CNS). This review addresses meningeal immunity, a less-studied aspect of neuroimmune interactions. The meninges, a triple layer of membranes-the pia mater, arachnoid mater, and dura mater-surround the CNS, encompassing the cerebrospinal fluid produced by the choroid plexus epithelium. Unlike the adjacent brain parenchyma, the meninges contain a wide repertoire of immune cells. These constitute meningeal immunity, which is primarily concerned with immune surveillance of the CNS, and-according to recent evidence-also participates in postinjury CNS recovery, chronic neurodegenerative conditions, and even higher brain function. Meningeal immunity has recently come under the spotlight owing to the characterization of meningeal lymphatic vessels draining the CNS. Here, we review the current state of our understanding of meningeal immunity and its effects on healthy and diseased brains.


Subject(s)
Central Nervous System/immunology , Central Nervous System/metabolism , Disease Susceptibility , Homeostasis , Immunity , Meninges/physiology , Animals , Humans , Lymphatic Vessels/immunology , Lymphatic Vessels/metabolism , Neuroimmunomodulation , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
Cell ; 186(2): 382-397.e24, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36669473

ABSTRACT

Blood and lymphatic vessels form a versatile transport network and provide inductive signals to regulate tissue-specific functions. Blood vessels in bone regulate osteogenesis and hematopoiesis, but current dogma suggests that bone lacks lymphatic vessels. Here, by combining high-resolution light-sheet imaging and cell-specific mouse genetics, we demonstrate presence of lymphatic vessels in mouse and human bones. We find that lymphatic vessels in bone expand during genotoxic stress. VEGF-C/VEGFR-3 signaling and genotoxic stress-induced IL6 drive lymphangiogenesis in bones. During lymphangiogenesis, secretion of CXCL12 from proliferating lymphatic endothelial cells is critical for hematopoietic and bone regeneration. Moreover, lymphangiocrine CXCL12 triggers expansion of mature Myh11+ CXCR4+ pericytes, which differentiate into bone cells and contribute to bone and hematopoietic regeneration. In aged animals, such expansion of lymphatic vessels and Myh11-positive cells in response to genotoxic stress is impaired. These data suggest lymphangiogenesis as a therapeutic avenue to stimulate hematopoietic and bone regeneration.


Subject(s)
Bone Regeneration , Lymphatic Vessels , Aged , Animals , Humans , Mice , Endothelial Cells , Lymphangiogenesis
3.
Cell ; 181(4): 905-913.e7, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32333836

ABSTRACT

We have previously provided the first genetic evidence that angiotensin converting enzyme 2 (ACE2) is the critical receptor for severe acute respiratory syndrome coronavirus (SARS-CoV), and ACE2 protects the lung from injury, providing a molecular explanation for the severe lung failure and death due to SARS-CoV infections. ACE2 has now also been identified as a key receptor for SARS-CoV-2 infections, and it has been proposed that inhibiting this interaction might be used in treating patients with COVID-19. However, it is not known whether human recombinant soluble ACE2 (hrsACE2) blocks growth of SARS-CoV-2. Here, we show that clinical grade hrsACE2 reduced SARS-CoV-2 recovery from Vero cells by a factor of 1,000-5,000. An equivalent mouse rsACE2 had no effect. We also show that SARS-CoV-2 can directly infect engineered human blood vessel organoids and human kidney organoids, which can be inhibited by hrsACE2. These data demonstrate that hrsACE2 can significantly block early stages of SARS-CoV-2 infections.


Subject(s)
Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Peptidyl-Dipeptidase A/pharmacology , Pneumonia, Viral/drug therapy , Recombinant Proteins/pharmacology , Angiotensin-Converting Enzyme 2 , Animals , Betacoronavirus/genetics , Betacoronavirus/isolation & purification , Betacoronavirus/ultrastructure , Blood Vessels/virology , COVID-19 , Chlorocebus aethiops , Humans , Kidney/cytology , Kidney/virology , Mice , Organoids/virology , Pandemics , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Receptors, Virus/metabolism , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells
4.
Immunity ; 57(10): 2328-2343.e8, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39217987

ABSTRACT

The precise neurophysiological changes prompted by meningeal lymphatic dysfunction remain unclear. Here, we showed that inducing meningeal lymphatic vessel ablation in adult mice led to gene expression changes in glial cells, followed by reductions in mature oligodendrocyte numbers and specific lipid species in the brain. These phenomena were accompanied by altered meningeal adaptive immunity and brain myeloid cell activation. During brain remyelination, meningeal lymphatic dysfunction provoked a state of immunosuppression that contributed to delayed spontaneous oligodendrocyte replenishment and axonal loss. The deficiencies in mature oligodendrocytes and neuroinflammation due to impaired meningeal lymphatic function were solely recapitulated in immunocompetent mice. Patients diagnosed with multiple sclerosis presented reduced vascular endothelial growth factor C in the cerebrospinal fluid, particularly shortly after clinical relapses, possibly indicative of poor meningeal lymphatic function. These data demonstrate that meningeal lymphatics regulate oligodendrocyte function and brain myelination, which might have implications for human demyelinating diseases.


Subject(s)
Brain , Lymphatic Vessels , Meninges , Multiple Sclerosis , Myelin Sheath , Oligodendroglia , Animals , Oligodendroglia/metabolism , Mice , Meninges/immunology , Brain/metabolism , Brain/immunology , Humans , Myelin Sheath/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Vascular Endothelial Growth Factor C/metabolism , Mice, Inbred C57BL , Cell Survival , Remyelination , Female , Male , Adaptive Immunity
5.
Cell ; 174(1): 143-155.e16, 2018 06 28.
Article in English | MEDLINE | ID: mdl-29779947

ABSTRACT

Neisseria meningitidis, a bacterium responsible for meningitis and septicemia, proliferates and eventually fills the lumen of blood capillaries with multicellular aggregates. The impact of this aggregation process and its specific properties are unknown. We first show that aggregative properties are necessary for efficient infection and study their underlying physical mechanisms. Micropipette aspiration and single-cell tracking unravel unique features of an atypical fluidized phase, with single-cell diffusion exceeding that of isolated cells. A quantitative description of the bacterial pair interactions combined with active matter physics-based modeling show that this behavior relies on type IV pili active dynamics that mediate alternating phases of bacteria fast mutual approach, contact, and release. These peculiar fluid properties proved necessary to adjust to the geometry of capillaries upon bacterial proliferation. Intermittent attractive forces thus generate a fluidized phase that allows for efficient colonization of the blood capillary network during infection.


Subject(s)
Bacterial Adhesion/physiology , Capillaries/microbiology , Fimbriae, Bacterial/physiology , Neisseria meningitidis/pathogenicity , Animals , Bacterial Load , Capillaries/pathology , Endothelium/metabolism , Endothelium/microbiology , Endothelium/pathology , Female , Fimbriae Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, SCID , Microscopy, Confocal , Neisseria meningitidis/physiology , Skin Transplantation , Surface Tension , Time-Lapse Imaging , Transplantation, Heterologous
6.
Annu Rev Neurosci ; 47(1): 323-344, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38648267

ABSTRACT

Since its recent discovery, the meningeal lymphatic system has reshaped our understanding of central nervous system (CNS) fluid exchange, waste clearance, immune cell trafficking, and immune privilege. Meningeal lymphatics have also been demonstrated to functionally modify the outcome of neurological disorders and their responses to treatment, including brain tumors, inflammatory diseases such as multiple sclerosis, CNS injuries, and neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. In this review, we discuss recent evidence of the contribution of meningeal lymphatics to neurological diseases, as well as the available experimental methods for manipulating meningeal lymphatics in these conditions. Finally, we also provide a discussion of the pressing questions and challenges in utilizing meningeal lymphatics as a prime target for CNS therapeutic intervention and possibly drug delivery for brain disorders.


Subject(s)
Central Nervous System Diseases , Meninges , Humans , Animals , Central Nervous System Diseases/physiopathology , Central Nervous System Diseases/pathology , Lymphatic System/physiology , Lymphatic System/physiopathology , Lymphatic Vessels/physiology
7.
Physiol Rev ; 104(2): 765-834, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37971403

ABSTRACT

Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.


Subject(s)
Cardiovascular Diseases , Diethylstilbestrol/analogs & derivatives , Phosphoric Diester Hydrolases , Humans , Phosphodiesterase Inhibitors/therapeutic use , Cyclic AMP , Cyclic GMP , Protein Isoforms
8.
Annu Rev Cell Dev Biol ; 32: 677-691, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27298093

ABSTRACT

The two vascular systems of our body are the blood and the lymphatic vasculature. Our understanding of the genes and molecular mechanisms controlling the development of the lymphatic vasculature network has significantly improved. The availability of novel animal models and better imaging tools led to the identification of lymphatics in tissues and organs previously thought to be devoid of them. Similarly, the classical textbook list of established functional roles of the lymphatic system has been expanded by the addition of novel findings. In this review we provide a historical perspective of some of the important landmarks that opened the doors to researchers working in this field. We also summarize some of the current views about embryonic lymphangiogenesis, particularly about the source(s), commitment, and differentiation of lymphatic endothelial cells.


Subject(s)
Cell Lineage , Lymphangiogenesis , Animals , Blood Vessels/physiology , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans
9.
Immunity ; 51(3): 561-572.e5, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31402260

ABSTRACT

Lymphatic vessels form a critical component in the regulation of human health and disease. While their functional significance is increasingly being recognized, the comprehensive heterogeneity of lymphatics remains uncharacterized. Here, we report the profiling of 33,000 lymphatic endothelial cells (LECs) in human lymph nodes (LNs) by single-cell RNA sequencing. Unbiased clustering revealed six major types of human LECs. LECs lining the subcapsular sinus (SCS) of LNs abundantly expressed neutrophil chemoattractants, whereas LECs lining the medullary sinus (MS) expressed a C-type lectin CD209. Binding of a carbohydrate Lewis X (CD15) to CD209 mediated neutrophil binding to the MS. The neutrophil-selective homing by MS LECs may retain neutrophils in the LN medulla and allow lymph-borne pathogens to clear, preventing their spread through LNs in humans. Our study provides a comprehensive characterization of LEC heterogeneity and unveils a previously undefined role for medullary LECs in human immunity.


Subject(s)
Endothelial Cells/immunology , Neutrophils/immunology , Animals , Cell Adhesion Molecules/immunology , Cells, Cultured , Humans , Lectins, C-Type/immunology , Lewis X Antigen/immunology , Lymph Nodes/immunology , Lymphatic Vessels/immunology , Mice, Inbred C57BL , Receptors, Cell Surface/immunology , Surveys and Questionnaires
10.
EMBO J ; 42(5): e109032, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36715213

ABSTRACT

Despite a growing catalog of secreted factors critical for lymphatic network assembly, little is known about the mechanisms that modulate the expression level of these molecular cues in blood vascular endothelial cells (BECs). Here, we show that a BEC-specific transcription factor, SOX7, plays a crucial role in a non-cell-autonomous manner by modulating the transcription of angiocrine signals to pattern lymphatic vessels. While SOX7 is not expressed in lymphatic endothelial cells (LECs), the conditional loss of SOX7 function in mouse embryos causes a dysmorphic dermal lymphatic phenotype. We identify novel distant regulatory regions in mice and humans that contribute to directly repressing the transcription of a major lymphangiogenic growth factor (Vegfc) in a SOX7-dependent manner. Further, we show that SOX7 directly binds HEY1, a canonical repressor of the Notch pathway, suggesting that transcriptional repression may also be modulated by the recruitment of this protein partner at Vegfc genomic regulatory regions. Our work unveils a role for SOX7 in modulating downstream signaling events crucial for lymphatic patterning, at least in part via the transcriptional repression of VEGFC levels in the blood vascular endothelium.


Subject(s)
Endothelial Cells , Lymphatic Vessels , Humans , Mice , Animals , Endothelial Cells/metabolism , Lymphatic Vessels/metabolism , Gene Expression Regulation , Endothelium, Vascular , Transcription Factors/metabolism , Lymphangiogenesis/genetics , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
11.
Development ; 151(19)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39289869

ABSTRACT

Skeletal muscle development is a complex process involving myoblast fusion to generate multinucleated fibers. Myonuclei first align in the center of the myotubes before migrating to the periphery of the myofiber. Blood vessels (BVs) are important contributors to the correct development of skeletal muscle, and myonuclei are found next to BVs in adult muscle. Here, we show that most myonuclear migration to the periphery occurs between embryonic day 17.5 and postnatal day 1 in mouse. Furthermore, myonuclear accretion after postnatal day 7 does not result in centrally nucleated myofibers as observed in the embryo. Instead, myonuclei remain at the periphery of the myofiber without moving to the center. Finally, we show that hypovascularization of skeletal muscle alters the interaction between myonuclei and BVs, suggesting that BVs may contribute to myonuclear positioning during skeletal muscle postnatal development. Overall, this work provides a comprehensive analysis of skeletal muscle development during the highly dynamic postnatal period, bringing new insights about myonuclear positioning and its interaction with BVs.


Subject(s)
Cell Nucleus , Muscle Development , Muscle, Skeletal , Animals , Muscle, Skeletal/blood supply , Muscle, Skeletal/embryology , Muscle, Skeletal/growth & development , Muscle Development/physiology , Mice , Cell Nucleus/metabolism , Blood Vessels/growth & development , Blood Vessels/embryology , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Animals, Newborn , Mice, Inbred C57BL
12.
Development ; 151(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39119717

ABSTRACT

Developing long bones alter their shape while maintaining uniform cortical thickness via coordinated activity of bone-forming osteoblasts and bone-resorbing osteoclasts at periosteal and endosteal surfaces, a process we designate trans-pairing. Two types of trans-pairing shift cortical bone in opposite orientations: peri-forming trans-pairing (peri-t-p) increases bone marrow space and endo-forming trans-pairing (endo-t-p) decreases it, via paired activity of bone resorption and formation across the cortex. Here, we focused on endo-t-p in growing bones. Analysis of endo-t-p activity in the cortex of mouse fibulae revealed osteoclasts under the periosteum compressed by muscles, and expression of RANKL in periosteal cells of the cambium layer. Furthermore, mature osteoblasts were localized on the endosteum, while preosteoblasts were at the periosteum and within cortical canals. X-ray tomographic microscopy revealed the presence of cortical canals more closely associated with endo- than with peri-t-p. Sciatic nerve transection followed by muscle atrophy and unloading induced circumferential endo-t-p with concomitant spread of cortical canals. Such canals likely supply the endosteum with preosteoblasts from the periosteum under endo-t-p, allowing bone shape to change in response to mechanical stress or nerve injury.


Subject(s)
Osteoblasts , Osteoclasts , Periosteum , Animals , Osteoblasts/metabolism , Osteoblasts/cytology , Periosteum/cytology , Periosteum/metabolism , Osteoclasts/metabolism , Osteoclasts/cytology , Mice , Bone Development , Osteogenesis/physiology , Bone Resorption/pathology , Cortical Bone , RANK Ligand/metabolism , Mice, Inbred C57BL
13.
Proc Natl Acad Sci U S A ; 121(28): e2402624121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38954543

ABSTRACT

The pial vasculature is the sole source of blood supply to the neocortex. The brain is contained within the skull, a vascularized bone marrow with a unique anatomical connection to the brain meninges. Recent developments in tissue clearing have enabled detailed mapping of the entire pial and calvarial vasculature. However, what are the absolute flow rate values of those vascular networks? This information cannot accurately be retrieved with the commonly used bioimaging methods. Here, we introduce Pia-FLOW, a unique approach based on large-scale transcranial fluorescence localization microscopy, to attain hemodynamic imaging of the whole murine pial and calvarial vasculature at frame rates up to 1,000 Hz and spatial resolution reaching 5.4 µm. Using Pia-FLOW, we provide detailed maps of flow velocity, direction, and vascular diameters which can serve as ground-truth data for further studies, advancing our understanding of brain fluid dynamics. Furthermore, Pia-FLOW revealed that the pial vascular network functions as one unit for robust allocation of blood after stroke.


Subject(s)
Connectome , Hemodynamics , Pia Mater , Animals , Mice , Hemodynamics/physiology , Pia Mater/blood supply , Cerebrovascular Circulation/physiology , Brain/blood supply , Brain/diagnostic imaging , Skull/diagnostic imaging , Skull/blood supply , Stroke/physiopathology , Stroke/diagnostic imaging , Male , Mice, Inbred C57BL
14.
Proc Natl Acad Sci U S A ; 121(30): e2404778121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39018197

ABSTRACT

Tumor blood vessels are highly leaky in structure and have poor blood perfusion, which hampers infiltration and function of CD8T cells within tumor. Normalizing tumor vessels is thus thought to be important in promoting the flux of immune T cells and enhancing ant-tumor immunity. However, how tumor vasculature is normalized is poorly understood. Metformin (Met) combined with ant-PD-1 therapy is known to stimulate proliferation of and to produce large amounts of IFNγ from tumor-infiltrating CD8T lymphocytes (CD8TILs). We found that the combination therapy promotes the pericyte coverage of tumor vascular endothelial cells (ECs) to improve blood perfusion and that it suppresses the hyperpermeability through the increase of VE-cadherin. Peripheral node addressin(PNAd) and vascular cell adhesion molecule (VCAM)-1, both implicated to promote tumor infiltration of CD8T cells, were also increased. Importantly, tumor vessel normalization, characterized as the reduced 70-kDa dextran leakage and the enhancement of VE-cadherin and VCAM-1, were canceled by anti-CD8 Ab or anti-IFNγ Ab injection to mice. The increased CD8TILs were also abrogated by anti-IFNγ Ab injection. In vascular ECs, flow cytometry analysis revealed that pSTAT1 expression was found to be associated with VE-cadherin expression. Moreover, in vitro treatment with Met and IFNγ enhanced VE-cadherin and VCAM-1 on human umbilical vein endothelial cells (HUVECs). The Kaplan-Meier method revealed a correlation of VE-cadherin or VCAM-1 levels with overall survival in patients treated with immune checkpoint inhibitors. These data indicate that IFNγ-mediated cross talk of CD8TILs with tumor vessels is important for creating a better tumor microenvironment and maintaining sustained antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Interferon-gamma , Metformin , Programmed Cell Death 1 Receptor , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Animals , Interferon-gamma/metabolism , Mice , Metformin/pharmacology , Metformin/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Cell Line, Tumor , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Vascular Cell Adhesion Molecule-1/metabolism , Mice, Inbred C57BL , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Cadherins/metabolism , Antigens, CD/metabolism , Drug Synergism
15.
Proc Natl Acad Sci U S A ; 121(18): e2317760121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38652741

ABSTRACT

The complex interplay between malignant cells and the cellular and molecular components of the tumor stroma is a key aspect of cancer growth and development. These tumor-host interactions are often affected by soluble bioactive molecules such as proteoglycans. Decorin, an archetypical small leucine-rich proteoglycan primarily expressed by stromal cells, affects cancer growth in its soluble form by interacting with several receptor tyrosine kinases (RTK). Overall, decorin leads to a context-dependent and protracted cessation of oncogenic RTK activity by attenuating their ability to drive a prosurvival program and to sustain a proangiogenic network. Through an unbiased transcriptomic analysis using deep RNAseq, we identified that decorin down-regulated a cluster of tumor-associated genes involved in lymphatic vessel (LV) development when systemically delivered to mice harboring breast carcinoma allografts. We found that Lyve1 and Podoplanin, two established markers of LVs, were markedly suppressed at both the mRNA and protein levels, and this suppression correlated with a significant reduction in tumor LVs. We further identified that soluble decorin, but not its homologous proteoglycan biglycan, inhibited LV sprouting in an ex vivo 3D model of lymphangiogenesis. Mechanistically, we found that decorin interacted with vascular endothelial growth factor receptor 3 (VEGFR3), the main lymphatic RTK, and its activity was required for the decorin-mediated block of lymphangiogenesis. Finally, we identified that Lyve1 was in part degraded via decorin-evoked autophagy in a nutrient- and energy-independent manner. These findings implicate decorin as a biological factor with antilymphangiogenic activity and provide a potential therapeutic agent for curtailing breast cancer growth and metastasis.


Subject(s)
Decorin , Lymphangiogenesis , Decorin/metabolism , Decorin/genetics , Animals , Mice , Humans , Female , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Cell Line, Tumor , Disease Progression , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Gene Expression Regulation, Neoplastic
16.
Circ Res ; 135(6): 671-684, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39092506

ABSTRACT

BACKGROUND: The elaborate patterning of coronary arteries critically supports the high metabolic activity of the beating heart. How coronary endothelial cells coordinate hierarchical vascular remodeling and achieve arteriovenous specification remains largely unknown. Understanding the molecular and cellular cues that pattern coronary arteries is crucial to develop innovative therapeutic strategies that restore functional perfusion within the ischemic heart. METHODS: Single-cell transcriptomics and histological validation were used to delineate heterogeneous transcriptional states of the developing and mature coronary endothelium with a focus on sprouting endothelium and arterial cell specification. Genetic lineage tracing and high-resolution 3-dimensional imaging were used to characterize the origin and mechanisms of coronary angiogenic sprouting, as well as to fate-map selective endothelial lineages. Integration of single-cell transcriptomic data from ischemic adult mouse hearts and human embryonic data served to assess the conservation of transcriptional states across development, disease, and species. RESULTS: We discover that coronary arteries originate from cells that have previously transitioned through a specific tip cell phenotype. We identify nonoverlapping intramyocardial and subepicardial tip cell populations with differential gene expression profiles and regulatory pathways. Esm1-lineage tracing confirmed that intramyocardial tip cells selectively contribute to coronary arteries and endocardial tunnels, but not veins. Notably, prearterial cells are detected from development stages to adulthood, increasingly in response to ischemic injury, and in human embryos, suggesting that tip cell-to-artery specification is a conserved mechanism. CONCLUSIONS: A tip cell-to-artery specification mechanism drives arterialization of the intramyocardial plexus and endocardial tunnels throughout life and is reactivated upon ischemic injury. Differential sprouting programs govern the formation and specification of the venous and arterial coronary plexus.


Subject(s)
Coronary Vessels , Neovascularization, Physiologic , Animals , Coronary Vessels/embryology , Coronary Vessels/metabolism , Humans , Mice , Single-Cell Analysis , Endothelial Cells/metabolism , Transcriptome , Cell Lineage , Mice, Inbred C57BL , Male , Female , Myocardium/metabolism , Myocardium/cytology
17.
Circ Res ; 134(11): 1465-1482, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38655691

ABSTRACT

BACKGROUND: Preclinical studies have shown the therapeutic potential of VEGF-B (vascular endothelial growth factor B) in revascularization of the ischemic myocardium, but the associated cardiac hypertrophy and adverse side effects remain a concern. To understand the importance of endothelial proliferation and migration for the beneficial versus adverse effects of VEGF-B in the heart, we explored the cardiac effects of autocrine versus paracrine VEGF-B expression in transgenic and gene-transduced mice. METHODS: We used single-cell RNA sequencing to compare cardiac endothelial gene expression in VEGF-B transgenic mouse models. Lineage tracing was used to identify the origin of a VEGF-B-induced novel endothelial cell population and adeno-associated virus-mediated gene delivery to compare the effects of VEGF-B isoforms. Cardiac function was investigated using echocardiography, magnetic resonance imaging, and micro-computed tomography. RESULTS: Unlike in physiological cardiac hypertrophy driven by a cardiomyocyte-specific VEGF-B transgene (myosin heavy chain alpha-VEGF-B), autocrine VEGF-B expression in cardiac endothelium (aP2 [adipocyte protein 2]-VEGF-B) was associated with septal defects and failure to increase perfused subendocardial capillaries postnatally. Paracrine VEGF-B led to robust proliferation and myocardial migration of a novel cardiac endothelial cell lineage (VEGF-B-induced endothelial cells) of endocardial origin, whereas autocrine VEGF-B increased proliferation of VEGF-B-induced endothelial cells but failed to promote their migration and efficient contribution to myocardial capillaries. The surviving aP2-VEGF-B offspring showed an altered ratio of secreted VEGF-B isoforms and developed massive pathological cardiac hypertrophy with a distinct cardiac vessel pattern. In the normal heart, we found a small VEGF-B-induced endothelial cell population that was only minimally expanded during myocardial infarction but not during physiological cardiac hypertrophy associated with mouse pregnancy. CONCLUSIONS: Paracrine and autocrine secretions of VEGF-B induce expansion of a specific endocardium-derived endothelial cell population with distinct angiogenic markers. However, autocrine VEGF-B signaling fails to promote VEGF-B-induced endothelial cell migration and contribution to myocardial capillaries, predisposing to septal defects and inducing a mismatch between angiogenesis and myocardial growth, which results in pathological cardiac hypertrophy.


Subject(s)
Cardiomegaly , Cell Lineage , Endocardium , Endothelial Cells , Mice, Transgenic , Vascular Endothelial Growth Factor B , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cardiomegaly/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor B/genetics , Mice , Endocardium/metabolism , Endocardium/pathology , Paracrine Communication , Cell Proliferation , Autocrine Communication , Mice, Inbred C57BL , Female , Male , Cell Movement
18.
Circ Res ; 134(10): 1259-1275, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38597112

ABSTRACT

BACKGROUND: GPCRs (G-protein-coupled receptors) play a central role in the regulation of smooth muscle cell (SMC) contractility, but the function of SMC-expressed orphan GPCR class C group 5 member C (GPRC5C) is unclear. The aim of this project is to define the role of GPRC5C in SMC in vitro and in vivo. METHODS: We studied the role of GPRC5C in the regulation of SMC contractility and differentiation in human and murine SMC in vitro, as well as in tamoxifen-inducible, SMC-specific GPRC5C knockout mice under basal conditions and in vascular disease in vivo. RESULTS: Mesenteric arteries from tamoxifen-inducible, SMC-specific GPRC5C knockout mice showed ex vivo significantly reduced angiotensin II (Ang II)-dependent calcium mobilization and contraction, whereas responses to other relaxant or contractile factors were normal. In vitro, the knockdown of GPRC5C in human aortic SMC resulted in diminished Ang II-dependent inositol phosphate production and lower myosin light chain phosphorylation. In line with this, tamoxifen-inducible, SMC-specific GPRC5C knockout mice showed reduced Ang II-induced arterial hypertension, and acute inactivation of GPRC5C was able to ameliorate established arterial hypertension. Mechanistically, we show that GPRC5C and the Ang II receptor AT1 dimerize, and knockdown of GPRC5C resulted in reduced binding of Ang II to AT1 receptors in HEK293 cells, human and murine SMC, and arteries from tamoxifen-inducible, SMC-specific GPRC5C knockout mice. CONCLUSIONS: Our data show that GPRC5C regulates Ang II-dependent vascular contraction by facilitating AT1 receptor-ligand binding and signaling.


Subject(s)
Angiotensin II , Muscle, Smooth, Vascular , Receptors, G-Protein-Coupled , Animals , Humans , Male , Mice , Angiotensin II/pharmacology , Cells, Cultured , Hypertension/metabolism , Hypertension/physiopathology , Hypertension/chemically induced , Hypertension/genetics , Mesenteric Arteries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle Contraction , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Vasoconstriction
19.
Semin Immunol ; 59: 101629, 2022 01.
Article in English | MEDLINE | ID: mdl-35753867

ABSTRACT

Traumatic injuries of the central nervous system (CNS) affect millions of people worldwide, and they can lead to severely damaging consequences such as permanent disability and paralysis. Multiple factors can obstruct recovery after CNS injury. One of the most significant is the progressive neuronal death that follows the initial mechanical impact, leading to the loss of undamaged cells via a process termed secondary neurodegeneration. Efforts to define treatments that limit the spread of damage, while important, have been largely ineffectual owing to gaps in the mechanistic understanding that underlies the persisting neuronal cell death. Inflammation, with its influx of immune cells that occurs shortly after injury, has been associated with secondary neurodegeneration. However, the role of the immune system after CNS injury is far more complex. Studies have indicated that the immune response after CNS injury is detrimental, owing to immune cell-produced factors (e.g., pro-inflammatory cytokines, free radicals, neurotoxic glutamate) that worsen tissue damage. Our lab and others have also demonstrated the beneficial immune response that occurs after CNS injury, with the release of growth factors such as brain-derived growth factor (BDNF) and interleukin (IL-10) and the clearance of apoptotic and myelin debris by immune cells1-4. In this review, we first discuss the multifaceted roles of the immune system after CNS injury. We then speculate on how advancements in single-cell RNA technologies can dramatically change our understanding of the immune response, how the spinal cord meninges serve as an important site for hosting immunological processes critical for recovery, and how the origin of peripherally recruited immune cells impacts their function in the injured CNS.


Subject(s)
Central Nervous System , Spinal Cord Injuries , Humans , Inflammation , Cytokines , Immune System , Immunity
20.
Proc Natl Acad Sci U S A ; 120(21): e2212933120, 2023 05 23.
Article in English | MEDLINE | ID: mdl-37186852

ABSTRACT

The glymphatic system is a perivascular fluid transport system for waste clearance. Glymphatic transport is believed to be driven by the perivascular pumping effect created by the pulsation of the arterial wall caused by the cardiac cycle. Ultrasound sonication of circulating microbubbles (MBs) in the cerebral vasculature induces volumetric expansion and contraction of MBs that push and pull on the vessel wall to generate a MB pumping effect. The objective of this study was to evaluate whether glymphatic transport can be mechanically manipulated by focused ultrasound (FUS) sonication of MBs. The glymphatic pathway in intact mouse brains was studied using intranasal administration of fluorescently labeled albumin as fluid tracers, followed by FUS sonication at a deep brain target (thalamus) in the presence of intravenously injected MBs. Intracisternal magna injection, the conventional technique used in studying glymphatic transport, was employed to provide a comparative reference. Three-dimensional confocal microscopy imaging of optically cleared brain tissue revealed that FUS sonication enhanced the transport of fluorescently labeled albumin tracer in the perivascular space (PVS) along microvessels, primarily the arterioles. We also obtained evidence of FUS-enhanced penetration of the albumin tracer from the PVS into the interstitial space. This study revealed that ultrasound combined with circulating MBs could mechanically enhance glymphatic transport in the brain.


Subject(s)
Glymphatic System , Microbubbles , Mice , Animals , Brain/diagnostic imaging , Brain/metabolism , Glymphatic System/diagnostic imaging , Glymphatic System/metabolism , Ultrasonography , Albumins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL