Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Biol Open ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38639409

ABSTRACT

Blood vessels serve as intermediate conduits for the extension of sympathetic axons towards target tissues, while also acting as crucial targets for their homeostatic processes encompassing the regulation of temperature, blood pressure, and oxygen availability. How sympathetic axons innervate not only blood vessels but also a wide array of target tissues is not clear. Here we show that in embryonic skin, after the establishment of co-branching between sensory nerves and blood vessels, sympathetic axons invade the skin alongside these sensory nerves and extend their branches towards these blood vessels covered by vascular smooth muscle cells (VSMCs). Our mosaic labeling technique for sympathetic axons shows that collateral branching predominantly mediates the innervation of VSMC-covered blood vessels by sympathetic axons. The expression of nerve growth factor (NGF), previously known to induce collateral axon branching in culture, can be detected in the vascular smooth muscle cell (VSMC)-covered blood vessels, as well as sensory nerves. Indeed, VSMC-specific Ngf knockout leads to a significant decrease of collateral branching of sympathetic axons innervating VSMC-covered blood vessels. These data suggest that VSMC-derived NGF serves as an inductive signal for collateral branching of sympathetic axons innervating blood vessels in the embryonic skin.

2.
Front Cell Dev Biol ; 11: 1150775, 2023.
Article in English | MEDLINE | ID: mdl-37091974

ABSTRACT

The central nervous system (CNS) is considered as an immune privilege organ, based on experiments in the mid 20th century showing that the brain fails to mount an efficient immune response against an allogeneic graft. This suggests that in addition to the presence of the blood-brain barrier (BBB), the apparent absence of classical lymphatic vasculature in the CNS parenchyma limits the capacity for an immune response. Although this view is partially overturned by the recent discovery of the lymphatic-like hybrid vessels in the Schlemm's canal in the eye and the lymphatic vasculature in the outmost layer of the meninges, the existence of lymphatic vessels in the CNS parenchyma has not been reported. Two potential mechanisms by which lymphatic vasculature may arise in the organs are: 1) sprouting and invasion of lymphatic vessels from the surrounding tissues into the parenchyma and 2) differentiation of blood endothelial cells into lymphatic endothelial cells in the parenchyma. Considering these mechanisms, we here discuss what causes the dearth of lymphatic vessels specifically in the CNS parenchyma.

3.
Biol Open ; 11(9)2022 09 15.
Article in English | MEDLINE | ID: mdl-36017733

ABSTRACT

Recent genetic lineage tracing studies reveal heterogeneous origins of vascular endothelial cells and pericytes in the developing brain vasculature, despite classical experimental evidence for a mesodermal origin. Here we provide evidence through a genetic lineage tracing experiment that cephalic paraxial mesodermal cells give rise to endothelial cells and pericytes in the developing mouse brain. We show that Hepatic leukemia factor (Hlf) is transiently expressed by cephalic paraxial mesenchyme at embryonic day (E) 8.0-9.0 and the genetically marked E8.0 Hlf-expressing cells mainly contribute to the developing brain vasculature. Interestingly, the genetically marked E10.5 Hlf-expressing cells, which have been previously reported to contain embryonic hematopoietic stem cells, fail to contribute to the vascular cells. Combined, our genetic lineage tracing data demonstrate that a transient expression of Hlf marks a cephalic paraxial mesenchyme contributing to the developing brain vasculature. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Endothelial Cells , Leukemia , Animals , Brain , Humans , Leukemia/metabolism , Mesoderm , Mice , Stem Cells
4.
Front Cell Dev Biol ; 10: 850645, 2022.
Article in English | MEDLINE | ID: mdl-35359438

ABSTRACT

Embryos devoid of autonomic innervation suffer sudden cardiac death. However, whether autonomic neurons have a role in heart development is poorly understood. To investigate if sympathetic neurons impact cardiomyocyte maturation, we co-cultured phenotypically immature cardiomyocytes derived from human induced pluripotent stem cells with mouse sympathetic ganglion neurons. We found that 1) multiple cardiac structure and ion channel genes related to cardiomyocyte maturation were up-regulated when co-cultured with sympathetic neurons; 2) sarcomere organization and connexin-43 gap junctions increased; 3) calcium imaging showed greater transient amplitudes. However, sarcomere spacing, relaxation time, and level of sarcoplasmic reticulum calcium did not show matured phenotypes. We further found that addition of endothelial and epicardial support cells did not enhance maturation to a greater extent beyond sympathetic neurons, while administration of isoproterenol alone was insufficient to induce changes in gene expression. These results demonstrate that sympathetic neurons have a significant and complex role in regulating cardiomyocyte development.

5.
Article in English | MEDLINE | ID: mdl-35379659

ABSTRACT

This review highlights new concepts in vascular patterning in the last 10 years, with emphasis on its beauty and complexity. Endothelial cell signaling pathways that respond to molecular or mechanical signals are described, and examples of vascular patterning that use these pathways in brain, skin, heart, and kidney are highlighted. The pathological consequences of patterning loss are discussed in the context of arteriovenous malformations (AVMs), and prospects for the next 10 years presented.


Subject(s)
Arteriovenous Malformations , Humans , Arteriovenous Malformations/metabolism , Arteriovenous Malformations/pathology , Signal Transduction , Brain/metabolism
6.
Angiogenesis ; 25(2): 205-224, 2022 05.
Article in English | MEDLINE | ID: mdl-34665379

ABSTRACT

In mice, embryonic dermal lymphatic development is well understood and used to study gene functions in lymphangiogenesis. Notch signaling is an evolutionarily conserved pathway that modulates cell fate decisions, which has been shown to both inhibit and promote dermal lymphangiogenesis. Here, we demonstrate distinct roles for Notch4 signaling versus canonical Notch signaling in embryonic dermal lymphangiogenesis. Actively growing embryonic dermal lymphatics expressed NOTCH1, NOTCH4, and DLL4 which correlated with Notch activity. In lymphatic endothelial cells (LECs), DLL4 activation of Notch induced a subset of Notch effectors and lymphatic genes, which were distinctly regulated by Notch1 and Notch4 activation. Treatment of LECs with VEGF-A or VEGF-C upregulated Dll4 transcripts and differentially and temporally regulated the expression of Notch1 and Hes/Hey genes. Mice nullizygous for Notch4 had an increase in the closure of the lymphangiogenic fronts which correlated with reduced vessel caliber in the maturing lymphatic plexus at E14.5 and reduced branching at E16.5. Activation of Notch4 suppressed LEC migration in a wounding assay significantly more than Notch1, suggesting a dominant role for Notch4 in regulating LEC migration. Unlike Notch4 nulls, inhibition of canonical Notch signaling by expressing a dominant negative form of MAML1 (DNMAML) in Prox1+ LECs led to increased lymphatic density consistent with an increase in LEC proliferation, described for the loss of LEC Notch1. Moreover, loss of Notch4 did not affect LEC canonical Notch signaling. Thus, we propose that Notch4 signaling and canonical Notch signaling have distinct functions in the coordination of embryonic dermal lymphangiogenesis.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels , Animals , Endothelial Cells/metabolism , Lymphangiogenesis/physiology , Lymphatic System/metabolism , Lymphatic Vessels/metabolism , Mice , Receptors, Notch/metabolism , Signal Transduction
7.
Sci Adv ; 7(49): eabh4181, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34851661

ABSTRACT

Neurons can regulate the development, pathogenesis, and regeneration of target organs. However, the role of neurons during heart development and regeneration remains unclear. We genetically inhibited sympathetic innervation in vivo, which resulted in heart enlargement with an increase in cardiomyocyte number. Transcriptomic and protein analysis showed down-regulation of the two clock gene homologs Period1/Period2 (Per1/Per2) accompanied by up-regulation of cell cycle genes. Per1/Per2 deletion increased heart size and cardiomyocyte proliferation, recapitulating sympathetic neuron­deficient hearts. Conversely, increasing sympathetic activity by norepinephrine treatment induced Per1/Per2 and suppressed cardiomyocyte proliferation. We further found that the two clock genes negatively regulate myocyte mitosis entry through the Wee1 kinase pathway. Our findings demonstrate a previously unknown link between cardiac neurons and clock genes in regulation of cardiomyocyte proliferation and heart size and provide mechanistic insights for developing neuromodulation strategies for cardiac regen5eration.

8.
Dev Biol ; 477: 70-84, 2021 09.
Article in English | MEDLINE | ID: mdl-34015362

ABSTRACT

The C-X-C chemokine receptor CXCR4 and its ligand CXCL12 play an important role in organ-specific vascular branching morphogenesis. CXCR4 is preferentially expressed by arterial endothelial cells, and local secretion of CXCL12 determines the organotypic pattern of CXCR4+ arterial branching. Previous loss-of-function studies clearly demonstrated that CXCL12-CXCR4 signaling is necessary for proper arterial branching in the developing organs such as the skin and heart. To further understand the role of CXCL12-CXCR4 signaling in organ-specific vascular development, we generated a mouse model carrying the Cre recombinase-inducible Cxcr4 transgene. Endothelial cell-specific Cxcr4 gain-of-function embryos exhibited defective vascular remodeling and formation of a hierarchical vascular branching network in the developing skin and heart. Ectopic expression of CXCR4 in venous endothelial cells, but not in lymphatic endothelial cells, caused blood-filled, enlarged lymphatic vascular phenotypes, accompanied by edema. These data suggest that CXCR4 expression is tightly regulated in endothelial cells for appropriate vascular development in an organ-specific manner.


Subject(s)
Blood Vessels/embryology , Endothelial Cells/physiology , Neovascularization, Physiologic/physiology , Receptors, CXCR4/physiology , Animals , Blood Vessels/anatomy & histology , Endothelial Cells/metabolism , Gain of Function Mutation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Receptors, CXCR4/biosynthesis , Vascular Remodeling/physiology
9.
Ultrasound Med Biol ; 47(3): 751-758, 2021 03.
Article in English | MEDLINE | ID: mdl-33293111

ABSTRACT

In vivo micro-imaging of mice is useful in studying the genetic basis of cardiac development in mutant embryos. We examined Phox2b-/- mutant mice, which lack autonomic innervation to the heart and die in utero, and investigated whether this lack of innervation causes cardiac dysfunction during embryogenesis. A VisualSonics Vevo 2100 ultrahigh-frequency linear array ultrasound machine with 30- and 40-MHz probes was used to analyze embryo size, gross characteristics, ventricular contractility and rhythm. Phox2b-/- mutant embryos underwent cessation of heartbeat and death at a greater rate than wild-type controls. We did not observe a hydrops phenotype or congenital heart defects in Phox2b-/- mutants. Analysis of heart rhythm revealed no significant correlation with genotype. Absent these signs of a progressive pathology, we suggest that Phox2b-/- mutant embryos likely die of sudden death secondary to acute arrhythmia. These data provide insight into the role of cardiac autonomic innervation during development.


Subject(s)
Echocardiography/methods , Embryo Loss/genetics , Heart/diagnostic imaging , Heart/embryology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Homozygote , Mice , Mice, Inbred C57BL , Phenotype
10.
Nat Commun ; 11(1): 6314, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33298956

ABSTRACT

Blood and lymphatic vessels structurally bear a strong resemblance but never share a lumen, thus maintaining their distinct functions. Although lymphatic vessels initially arise from embryonic veins, the molecular mechanism that maintains separation of these two systems has not been elucidated. Here, we show that genetic deficiency of Folliculin, a tumor suppressor, leads to misconnection of blood and lymphatic vessels in mice and humans. Absence of Folliculin results in the appearance of lymphatic-biased venous endothelial cells caused by ectopic expression of Prox1, a master transcription factor for lymphatic specification. Mechanistically, this phenotype is ascribed to nuclear translocation of the basic helix-loop-helix transcription factor Transcription Factor E3 (TFE3), binding to a regulatory element of Prox1, thereby enhancing its venous expression. Overall, these data demonstrate that Folliculin acts as a gatekeeper that maintains separation of blood and lymphatic vessels by limiting the plasticity of committed endothelial cells.


Subject(s)
Cell Plasticity , Lymphatic Vessels/embryology , Proto-Oncogene Proteins/deficiency , Tumor Suppressor Proteins/deficiency , Veins/embryology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Nucleus/metabolism , Embryo, Mammalian , Endothelial Cells/metabolism , Endothelium, Lymphatic/cytology , Endothelium, Lymphatic/embryology , Endothelium, Vascular/cytology , Endothelium, Vascular/embryology , Female , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Lymphatic Vessels/cytology , Male , Mice , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins/genetics , RNA Interference , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Veins/cytology
11.
Commun Biol ; 3(1): 734, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33277595

ABSTRACT

The preferential accumulation of vascular smooth muscle cells (vSMCs) on arteries versus veins during early development is a well-described phenomenon, but the molecular pathways underlying this polarization are not well understood. In zebrafish, the cxcr4a receptor (mammalian CXCR4) and its ligand cxcl12b (mammalian CXCL12) are both preferentially expressed on arteries at time points consistent with the arrival and differentiation of the first vSMCs during vascular development. We show that autocrine cxcl12b/cxcr4 activity leads to increased production of the vSMC chemoattractant ligand pdgfb by endothelial cells in vitro and increased expression of pdgfb by arteries of zebrafish and mice in vivo. Additionally, we demonstrate that expression of the blood flow-regulated transcription factor klf2a in primitive veins negatively regulates cxcr4/cxcl12 and pdgfb expression, restricting vSMC recruitment to the arterial vasculature. Together, this signalling axis leads to the differential acquisition of vSMCs at sites where klf2a expression is low and both cxcr4a and pdgfb are co-expressed, i.e. arteries during early development.


Subject(s)
Chemokines/metabolism , Muscle, Smooth, Vascular/cytology , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Mice , Mutation , Myocytes, Smooth Muscle , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Signal Transduction , Zebrafish
12.
Hum Mol Genet ; 29(20): 3350-3360, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33030203

ABSTRACT

Proteus syndrome is a progressive overgrowth disorder with vascular malformations caused by mosaic expression of the AKT1 c.49G > A, p.(E17K) activating variant which was predicted to cause lethality if expressed ubiquitously. To test that hypothesis, we used the ACTB-Cre gene to activate a conditional Akt1 p.(E17K) allele in the mouse. No offspring that was heterozygous for both Cre and the conditional allele (ßA-Akt1WT/flx) was viable. Fewer than expected numbers of ßA-Akt1WT/flx embryos were seen beginning at E11.5, but a few survived until E17.5. The phenotype ranged from mild to severe, but generally ßA-Akt1WT/flx embryos had fewer visible blood vessels and more hemorrhages than their wild-type littermates, which was suggestive of a vascular abnormality. Examination of E13.5 limb skin showed a primitive capillary network with increased branching complexity and abnormal patterning compared with wild-type skin. By E15.5, wild-type skin had undergone angiogenesis and formed a hierarchical network of remodeled vessels, whereas in ßA-Akt1WT/flx embryos, the capillary network failed to remodel. Mural cell coverage of the blood vessels was also reduced in ßA-Akt1WT/flx skin compared with that of wild type. Restricting expression of Akt1E17K to endothelial, cardiac or smooth muscle cells resulted in viable offspring and remodeled vasculature and did not recapitulate the ßA-Akt1WT/flx phenotype. We conclude that ubiquitous expression of Akt1E17K suppresses remodeling and inhibits the formation of a normal skin vasculature. We postulate that this failure prevents proper circulation necessary to support the growing embryo and that it is the result of interactions of multiple cell types with increased AKT signaling.


Subject(s)
Embryo Loss/pathology , Embryo, Mammalian/pathology , Neovascularization, Pathologic/pathology , Peripheral Vascular Diseases/pathology , Proteus Syndrome/pathology , Proto-Oncogene Proteins c-akt/metabolism , Animals , Embryo Loss/etiology , Embryo Loss/metabolism , Embryo, Mammalian/metabolism , Female , Mice , Mice, Transgenic , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Peripheral Vascular Diseases/etiology , Peripheral Vascular Diseases/metabolism , Proteus Syndrome/etiology , Proteus Syndrome/metabolism , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction
13.
Mol Biol Cell ; 31(18): 1974-1987, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32583739

ABSTRACT

Among the three nonmuscle myosin 2 (NM2) paralogs, NM 2A and 2B, but not 2C, are detected in endothelial cells. To study the role of NM2 in vascular formation, we ablate NM2 in endothelial cells in mice. Ablating NM2A, but not NM2B, results in reduced blood vessel coverage and increased vascular branching in the developing mouse skin and coronary vasculature. NM2B becomes essential for vascular formation when NM2A expression is limited. Mice ablated for NM2B and one allele of NM2A develop vascular abnormalities similar to those in NM2A ablated mice. Using the embryoid body angiogenic sprouting assay in collagen gels reveals that NM2A is required for persistent angiogenic sprouting by stabilizing the endothelial cell cortex, and thereby preventing excessive branching and ensuring persistent migration of the endothelial sprouts. Mechanistically, NM2 promotes focal adhesion formation and cortical protrusion retraction during angiogenic sprouting. Further studies demonstrate the critical role of Rho kinase-activated NM2 signaling in the regulation of angiogenic sprouting in vitro and in vivo.


Subject(s)
Neovascularization, Physiologic/physiology , Nonmuscle Myosin Type IIA/metabolism , Nonmuscle Myosin Type IIB/metabolism , Angiogenesis Inducing Agents , Animals , Collagen/metabolism , Cytoskeletal Proteins/metabolism , Endothelial Cells/metabolism , Mice , Mice, Knockout , Morphogenesis , Myosin Heavy Chains/metabolism , Myosin Type II/metabolism , Neovascularization, Physiologic/genetics , Signal Transduction , rho-Associated Kinases/metabolism
14.
J Cell Biol ; 218(10): 3506-3525, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31541017

ABSTRACT

Ischemic stroke causes blood-brain barrier (BBB) breakdown due to significant damage to the integrity of BBB components. Recent studies have highlighted the importance of pericytes in the repair process of BBB functions triggered by PDGFRß up-regulation. Here, we show that perlecan, a major heparan sulfate proteoglycan of basement membranes, aids in BBB maintenance and repair through pericyte interactions. Using a transient middle cerebral artery occlusion model, we found larger infarct volumes and more BBB leakage in conditional perlecan (Hspg2)-deficient (Hspg2 - / - -TG) mice than in control mice. Control mice showed increased numbers of pericytes in the ischemic lesion, whereas Hspg2 - / - -TG mice did not. At the mechanistic level, pericytes attached to recombinant perlecan C-terminal domain V (perlecan DV, endorepellin). Perlecan DV enhanced the PDGF-BB-induced phosphorylation of PDGFRß, SHP-2, and FAK partially through integrin α5ß1 and promoted pericyte migration. Perlecan therefore appears to regulate pericyte recruitment through the cooperative functioning of PDGFRß and integrin α5ß1 to support BBB maintenance and repair following ischemic stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Heparan Sulfate Proteoglycans/metabolism , Infarction, Middle Cerebral Artery/metabolism , Pericytes/metabolism , Animals , Blood-Brain Barrier/pathology , Disease Models, Animal , Heparan Sulfate Proteoglycans/administration & dosage , Heparan Sulfate Proteoglycans/deficiency , Infarction, Middle Cerebral Artery/pathology , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
J Exp Med ; 216(4): 900-915, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30846482

ABSTRACT

Microglia play a pivotal role in the coordination of brain development and have emerged as a critical determinant in the progression of neurodegenerative diseases; however, the role of microglia in the onset and progression of neurodevelopmental disorders is less clear. Here we show that conditional deletion of αVß8 from the central nervous system (Itgb8ΔCNS mice) blocks microglia in their normal stepwise development from immature precursors to mature microglia. These "dysmature" microglia appear to result from reduced TGFß signaling during a critical perinatal window, are distinct from microglia with induced reduction in TGFß signaling during adulthood, and directly cause a unique neurodevelopmental syndrome characterized by oligodendrocyte maturational arrest, interneuron loss, and spastic neuromotor dysfunction. Consistent with this, early (but not late) microglia depletion completely reverses this phenotype. Together, these data identify novel roles for αVß8 and TGFß signaling in coordinating microgliogenesis with brain development and implicate abnormally programmed microglia or their products in human neurodevelopmental disorders that share this neuropathology.


Subject(s)
Integrins/metabolism , Interneurons/metabolism , Microglia/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism , Animals , Brain/growth & development , Brain/metabolism , Female , Integrins/genetics , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurodevelopmental Disorders/metabolism , Oligodendroglia/metabolism , Phenotype , Receptor, Transforming Growth Factor-beta Type II/genetics , Transforming Growth Factor beta1/genetics
16.
J Mol Cell Cardiol ; 127: 270-276, 2019 02.
Article in English | MEDLINE | ID: mdl-30639412

ABSTRACT

Atherosclerosis is a chronic disorder of the vessel wall. One key regulator of disease progression is lipid handling in macrophages. However, the role of macrophage mitochondrial-dependent fatty acid ß-oxidation (FAO) in atherosclerosis is not well defined. To address this, we focused on carnitine palmitoyltransferase (CPT) 1 and 2, which play an essential role in the transport of long chain fatty acids (FAs) into the mitochondria. Using conditional alleles of these mitochondrial enzymes, we have generated myeloid-specific Cpt1a and Cpt2 knockout mutants (CPT1a M-KO and CPT2 M-KO). In culture, macrophages derived from CPT1a and CPT2 M-KO mice have impaired FAO, enhanced expression of the CD36 scavenger receptor, increased uptake of oxidized low-density lipoprotein (oxLDL), and augmented transformation into cholesterol-rich foam cells. In line with these in vitro observations, in the atherosclerosis-susceptible apolipoprotein E (ApoE) KO background, CPT2 M-KO mice demonstrated augmented atherosclerosis, accompanied by increased accumulation of aortic macrophages with elevated CD36 expression. These data suggest that macrophage FAO is athero-protective and that augmenting FAO may potentially slow atherosclerotic progression.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Progression , Fatty Acids/metabolism , Macrophages/metabolism , Animals , Mice , Mice, Knockout , Myeloid Cells/metabolism , Oxidation-Reduction
17.
Front Cardiovasc Med ; 5: 78, 2018.
Article in English | MEDLINE | ID: mdl-29998128

ABSTRACT

Pericytes are mural cells surrounding blood vessels, adjacent to endothelial cells. Pericytes play critical roles in maturation and maintenance of vascular branching morphogenesis. In the central nervous system (CNS), pericytes are necessary for the formation and regulation of the blood-brain barrier (BBB) and pericyte deficiency accompanies CNS diseases including multiple sclerosis, diabetic retinopathy, neonatal intraventricular hemorrhage, and neurodegenerative disorders. Despite the importance of pericytes, their developmental origins and phenotypic diversity remain incompletely understood. Pericytes express multiple markers and the origin of pericytes differs by tissue, which may cause difficulty for the identification and understanding of the ontogeny of pericytes. Also, pericytes have the potential to give rise to different tissues in vitro but this is not clear in vivo. These studies indicate that pericytes are heterogeneous in a tissue- and context- dependent manner. This short review focuses on recent studies about identification of pericytes, heterogeneous origin of pericytes during development and in adults, and the differentiation capacity of pericytes, and pericytes in pathological settings.

18.
J Vis Exp ; (133)2018 03 29.
Article in English | MEDLINE | ID: mdl-29658918

ABSTRACT

Here, we present a protocol of a whole-mount adult ear skin imaging technique to study comprehensive three-dimensional neuro-vascular branching morphogenesis and patterning, as well as immune cell distribution at a cellular level. The analysis of peripheral nerve and blood vessel anatomical structures in adult tissues provides some insights into the understanding of functional neuro-vascular wiring and neuro-vascular degeneration in pathological conditions such as wound healing. As a highly informative model system, we have focused our studies on adult ear skin, which is readily accessible for dissection. Our simple and reproducible protocol provides an accurate depiction of the cellular components in the entire skin, such as peripheral nerves (sensory axons, sympathetic axons, and Schwann cells), blood vessels (endothelial cells and vascular smooth muscle cells), and inflammatory cells. We believe this protocol will pave the way to investigate morphological abnormalities in peripheral nerves and blood vessels as well as the inflammation in the adult ear skin under different pathological conditions.


Subject(s)
Ear/diagnostic imaging , Microscopy, Confocal/methods , Morphogenesis/immunology , Skin/blood supply , Animals , Cell Differentiation , Ear/pathology , Humans , Immunohistochemistry , Mice
19.
Dev Dyn ; 247(5): 741-753, 2018 05.
Article in English | MEDLINE | ID: mdl-29493038

ABSTRACT

BACKGROUND: Traditionally, the central nervous system (CNS) has been viewed as an immune-privileged environment with no lymphatic vessels. This view was partially overturned by the discovery of lymphatic vessels in the dural membrane that surrounds the brain, in contact with the interior surface of the skull. We here examine the distribution and developmental timing of these lymphatic vessels. RESULTS: Using the Prox1-GFP BAC transgenic reporter and immunostaining with antibodies to lymphatic markers LYVE-1, Prox1, and Podoplanin, we have carried out whole-mount imaging of dural lymphatic vasculature at postnatal stages. We have found that between birth and postnatal day (P) 13, lymphatic vessels extend alongside dural blood vessels from the side of the skull toward the midline. Between P13 and P20, lymphatic vessels along the transverse sinuses reach the superior sagittal sinus (SSS) and extend along the SSS toward the olfactory bulb. CONCLUSIONS: Compared with the embryonic developmental timing of lymphatic vessels in other tissues, e.g. skin, dural lymphatic vessel development is dramatically delayed. This study provides useful anatomical data for continuing investigations of the fundamental mechanisms that underlie dural lymphatic vessel development. Developmental Dynamics 247:741-753, 2018. © 2018 Wiley Periodicals, Inc.


Subject(s)
Lymphatic Vessels/embryology , Animals , Brain/blood supply , Meninges/embryology , Mice , Mice, Transgenic , Skin/embryology
20.
Sci Rep ; 8(1): 430, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323138

ABSTRACT

Obesity and type 2 diabetes are frequently associated with peripheral neuropathy. Though there are multiple methods for diagnosis and analysis of morphological changes of peripheral nerves and blood vessels, three-dimensional high-resolution imaging is necessary to appreciate the pathogenesis with an anatomically recognizable branching morphogenesis and patterning. Here we established a novel technique for whole-mount imaging of adult mouse ear skin to visualize branching morphogenesis and patterning of peripheral nerves and blood vessels. Whole-mount immunostaining of adult mouse ear skin showed that peripheral sensory and sympathetic nerves align with large-diameter blood vessels. Diet-induced obesity (DIO) mice exhibit defective vascular smooth muscle cells (VSMCs) coverage, while there is no significant change in the amount of peripheral nerves. The leptin receptor-deficient db/db mice, a severe obese and type 2 diabetic mouse model, exhibit defective VSMC coverage and a large increase in the amount of smaller-diameter nerve bundles with myelin sheath and unmyelinated nerve fibers. Interestingly, an increase in the amount of myeloid immune cells was observed in the DIO but not db/db mouse skin. These data suggest that our whole-mount imaging method enables us to investigate the neuro-vascular and neuro-immune phenotypes in the animal models of obesity and diabetes.


Subject(s)
Blood Vessels/diagnostic imaging , Diabetes Mellitus, Type 2/complications , Ear/diagnostic imaging , Obesity/complications , Peripheral Nerves/diagnostic imaging , Peripheral Nervous System Diseases/diagnostic imaging , Animals , Disease Models, Animal , Ear/blood supply , Ear/innervation , Imaging, Three-Dimensional , Mice , Microscopy, Confocal/methods , Morphogenesis , Receptors, Leptin/genetics , Skin/blood supply , Skin/innervation
SELECTION OF CITATIONS
SEARCH DETAIL
...