Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Cell ; 185(14): 2523-2541.e30, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35738284

ABSTRACT

Stem cell research endeavors to generate specific subtypes of classically defined "cell types." Here, we generate >90% pure human artery or vein endothelial cells from pluripotent stem cells within 3-4Ā days. We specified artery cells by inhibiting vein-specifying signals and vice versa. These cells modeled viral infection of human vasculature by Nipah and Hendra viruses, which are extraordinarily deadly (Ć¢ĀˆĀ¼57%-59% fatality rate) and require biosafety-level-4 containment. Generating pure populations of artery and vein cells highlighted that Nipah and Hendra viruses preferentially infected arteries; arteries expressed higher levels of their viral-entry receptor. Virally infected artery cells fused into syncytia containing up to 23 nuclei, which rapidly died. Despite infecting arteries and occupying Ć¢ĀˆĀ¼6%-17% of their transcriptome, Nipah and Hendra largely eluded innate immune detection, minimally eliciting interferon signaling. We thus efficiently generate artery and vein cells, introduce stem-cell-based toolkits for biosafety-level-4 virology, and explore the arterial tropism and cellular effects of Nipah and Hendra viruses.


Subject(s)
Hendra Virus , Nipah Virus , Pluripotent Stem Cells , Arteries , Endothelial Cells , Hendra Virus/genetics , Humans , Tropism
2.
Cell ; 176(5): 1128-1142.e18, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30686582

ABSTRACT

Collateral arteries are an uncommon vessel subtype that can provide alternate blood flow to preserve tissue following vascular occlusion. Some patients with heart disease develop collateral coronary arteries, and this correlates with increased survival. However, it is not known how these collaterals develop or how to stimulate them. We demonstrate that neonatal mouse hearts use a novel mechanism to build collateral arteries in response to injury. Arterial endothelial cells (ECs) migrated away from arteries along existing capillaries and reassembled into collateral arteries, which we termed "artery reassembly". Artery ECs expressed CXCR4, and following injury, capillary ECs induced its ligand, CXCL12. CXCL12 or CXCR4 deletion impaired collateral artery formation and neonatal heart regeneration. Artery reassembly was nearly absent in adults but was induced by exogenous CXCL12. Thus, understanding neonatal regenerative mechanisms can identify pathways that restore these processes in adults and identify potentially translatable therapeutic strategies for ischemic heart disease.


Subject(s)
Collateral Circulation/physiology , Heart/growth & development , Regeneration/physiology , Animals , Animals, Newborn/growth & development , Chemokine CXCL12/metabolism , Coronary Vessels/growth & development , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Receptors, CXCR4/metabolism , Signal Transduction
3.
Nature ; 633(8031): 905-913, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39198643

ABSTRACT

Life-threatening thrombotic events and neurological symptoms are prevalent in COVID-19 and areĀ persistent in patients with long COVID experiencing post-acute sequelae of SARS-CoV-2 infection1-4. Despite the clinical evidence1,5-7, the underlying mechanisms of coagulopathy in COVID-19 and its consequences in inflammation and neuropathology remain poorly understood and treatment options are insufficient. Fibrinogen, the central structural component of blood clots, is abundantly deposited in the lungs and brains of patients with COVID-19, correlates with disease severity and is a predictive biomarker for post-COVID-19 cognitive deficits1,5,8-10. Here we show that fibrin binds to the SARS-CoV-2 spike protein, forming proinflammatory blood clots that drive systemic thromboinflammation and neuropathology in COVID-19. Fibrin, acting through its inflammatory domain, is required for oxidative stress and macrophage activation in the lungs, whereas itĀ suppresses natural killer cells, after SARS-CoV-2 infection. Fibrin promotes neuroinflammation and neuronal loss after infection, as well as innate immune activation in the brain and lungs independently of active infection. AĀ monoclonal antibody targeting the inflammatory fibrin domain providesĀ protection from microglial activation and neuronal injury, as well as from thromboinflammation in the lung after infection. Thus, fibrin drives inflammation and neuropathology in SARS-CoV-2 infection, and fibrin-targeting immunotherapy may represent a therapeutic intervention for patients with acute COVID-19 and long COVID.


Subject(s)
Brain , COVID-19 , Fibrin , Inflammation , Thrombosis , Animals , Female , Humans , Male , Mice , Brain/drug effects , Brain/immunology , Brain/pathology , Brain/virology , COVID-19/immunology , COVID-19/pathology , COVID-19/virology , COVID-19/complications , Fibrin/antagonists & inhibitors , Fibrin/metabolism , Fibrinogen/metabolism , Immunity, Innate , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Inflammation/virology , Killer Cells, Natural/immunology , Lung/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Macrophage Activation/drug effects , Microglia/immunology , Microglia/pathology , Neuroinflammatory Diseases/complications , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/virology , Neurons/pathology , Neurons/virology , Oxidative Stress , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/metabolism , Thrombosis/complications , Thrombosis/immunology , Thrombosis/pathology , Thrombosis/virology , Post-Acute COVID-19 Syndrome/immunology , Post-Acute COVID-19 Syndrome/virology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology
4.
EMBO Rep ; 24(10): e55043, 2023 10 09.
Article in English | MEDLINE | ID: mdl-37551717

ABSTRACT

The cardiac endothelium influences ventricular chamber development by coordinating trabeculation and compaction. However, the endothelial-specific molecular mechanisms mediating this coordination are not fully understood. Here, we identify the Sox7 transcription factor as a critical cue instructing cardiac endothelium identity during ventricular chamber development. Endothelial-specific loss of Sox7 function in mice results in cardiac ventricular defects similar to non-compaction cardiomyopathy, with a change in the proportions of trabecular and compact cardiomyocytes in the mutant hearts. This phenotype is paralleled by abnormal coronary artery formation. Loss of Sox7 function disrupts the transcriptional regulation of the Notch pathway and connexins 37 and 40, which govern coronary arterial specification. Upon Sox7 endothelial-specific deletion, single-nuclei transcriptomics analysis identifies the depletion of a subset of Sox9/Gpc3-positive endocardial progenitor cells and an increase in erythro-myeloid cell lineages. Fate mapping analysis reveals that a subset of Sox7-null endothelial cells transdifferentiate into hematopoietic but not cardiomyocyte lineages. Our findings determine that Sox7 maintains cardiac endothelial cell identity, which is crucial to the cellular cross-talk that drives ventricular compaction and coronary artery development.


Subject(s)
Coronary Vessels , Endothelial Cells , Animals , Mice , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Myocytes, Cardiac/metabolism , Gene Expression Regulation , Endothelium/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
5.
Genes Dev ; 31(13): 1308-1324, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28779009

ABSTRACT

Sufficient blood flow to tissues relies on arterial blood vessels, but the mechanisms regulating their development are poorly understood. Many arteries, including coronary arteries of the heart, form through remodeling of an immature vascular plexus in a process triggered and shaped by blood flow. However, little is known about how cues from fluid shear stress are translated into responses that pattern artery development. Here, we show that mice lacking endothelial Dach1 had small coronary arteries, decreased endothelial cell polarization, and reduced expression of the chemokine Cxcl12 Under shear stress in culture, Dach1 overexpression stimulated endothelial cell polarization and migration against flow, which was reversed upon CXCL12/CXCR4 inhibition. In vivo, DACH1 was expressed during early arteriogenesis but was down in mature arteries. Mature artery-type shear stress (high, uniform laminar) specifically down-regulated DACH1, while the remodeling artery-type flow (low, variable) maintained DACH1 expression. Together, our data support a model in which DACH1 stimulates coronary artery growth by activating Cxcl12 expression and endothelial cell migration against blood flow into developing arteries. This activity is suppressed once arteries reach a mature morphology and acquire high, laminar flow that down-regulates DACH1. Thus, we identified a mechanism by which blood flow quality balances artery growth and maturation.


Subject(s)
Coronary Vessels/growth & development , Eye Proteins/genetics , Eye Proteins/metabolism , Neovascularization, Physiologic/genetics , Signal Transduction/genetics , Animals , Blood Flow Velocity/physiology , Cell Movement/genetics , Cells, Cultured , Chemokine CXCL12/genetics , Coronary Vessels/physiopathology , Endothelial Cells/cytology , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Mice, Inbred C57BL , Mutation , Organ Culture Techniques , Receptors, CXCR4/genetics , Stress, Mechanical
6.
Dev Biol ; 498: 77-86, 2023 06.
Article in English | MEDLINE | ID: mdl-37037405

ABSTRACT

Outflow tract (OFT) develops from cardiac progenitor cells in the second heart field (SHF) domain. APJ, a G-Protein Coupled Receptor, is expressed by cardiac progenitors in the SHF. By lineage tracing APJ+SHF cells, we show that these cardiac progenitors contribute to the cells of OFT, which eventually give rise to aorta and pulmonary trunk/artery upon its morphogenesis. Furthermore, we show that early APJĀ Ć¢Ā€Ā‹+Ā Ć¢Ā€Ā‹cells give rise to both aorta and pulmonary cells but late APJĀ Ć¢Ā€Ā‹+Ā Ć¢Ā€Ā‹cells predominantly give rise to pulmonary cells. APJ is expressed by the outflow tract progenitors in the SHF but its role is unclear. We performed knockout studies to determine the role of APJ in SHF cell proliferation and survival. Our data suggested that APJ knockout in the SHF reduced the proliferation of SHF progenitors, while there was no significant impact on survival. In addition, we show that ectopic overexpression of WNT in these cells disrupted aorta and pulmonary morphogenesis from OFT. Overall, our study has identified APJĀ Ć¢Ā€Ā‹+Ā Ć¢Ā€Ā‹progenitor population within the SHF that give rise to aorta and pulmonary trunk/artery cells. Furthermore, we show that APJ signaling stimulates proliferation of these cells in the SHF.


Subject(s)
Heart , Signal Transduction , Stem Cells , Pulmonary Artery , Aorta , Myocardium , Gene Expression Regulation, Developmental
7.
Arterioscler Thromb Vasc Biol ; 43(8): 1455-1477, 2023 08.
Article in English | MEDLINE | ID: mdl-37345524

ABSTRACT

BACKGROUND: Collateral arteries act as natural bypasses which reroute blood flow to ischemic regions and facilitate tissue regeneration. In an injured heart, neonatal artery endothelial cells orchestrate a systematic series of cellular events, which includes their outward migration, proliferation, and coalescence into fully functional collateral arteries. This process, called artery reassembly, aids complete cardiac regeneration in neonatal hearts but is absent in adults. The reason for this age-dependent disparity in artery cell response is completely unknown. In this study, we investigated if regenerative potential of coronary arteries is dictated by their ability to dedifferentiate. METHODS: Single-cell RNA sequencing of coronary endothelial cells was performed to identify differences in molecular profiles of neonatal and adult endothelial cells in mice. Findings from this in silico analyses were confirmed with in vivo experiments using genetic lineage tracing, whole organ immunostaining, confocal imaging, and cardiac functional assays in mice. RESULTS: Upon coronary occlusion, neonates showed a significant increase in actively cycling artery cells and expressed prominent dedifferentiation markers. Data from in silico pathway analyses and in vivo experiments suggested that upon myocardial infarction, cell cycle reentry of preexisting neonatal artery cells, the subsequent collateral artery formation, and recovery of cardiac function are dependent on arterial VegfR2 (vascular endothelial growth factor receptor-2). This subpopulation of dedifferentiated and proliferating artery cells was absent in nonregenerative postnatal day 7 or adult hearts. CONCLUSIONS: These data indicate that adult artery endothelial cells fail to drive collateral artery development due to their limited ability to dedifferentiate and proliferate.


Subject(s)
Endothelial Cells , Vascular Endothelial Growth Factor A , Animals , Mice , Endothelial Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Collateral Circulation/physiology , Coronary Vessels/metabolism , Cell Proliferation
8.
Nature ; 559(7714): 356-362, 2018 07.
Article in English | MEDLINE | ID: mdl-29973725

ABSTRACT

Arteries and veins are specified by antagonistic transcriptional programs. However, during development and regeneration, new arteries can arise from pre-existing veins through a poorly understood process of cell fate conversion. Here, using single-cell RNA sequencing and mouse genetics, we show that vein cells of the developing heart undergo an early cell fate switch to create a pre-artery population that subsequently builds coronary arteries. Vein cells underwent a gradual and simultaneous switch from venous to arterial fate before a subset of cells crossed a transcriptional threshold into the pre-artery state. Before the onset of coronary blood flow, pre-artery cells appeared in the immature vessel plexus, expressed mature artery markers, and decreased cell cycling. The vein-specifying transcription factor COUP-TF2 (also known as NR2F2) prevented plexus cells from overcoming the pre-artery threshold by inducing cell cycle genes. Thus, vein-derived coronary arteries are built by pre-artery cells that can differentiate independently of blood flow upon the release of inhibition mediated by COUP-TF2 and cell cycle factors.


Subject(s)
Arteries/cytology , Coronary Vessels/cytology , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism , Veins/cytology , Animals , Arteries/metabolism , COUP Transcription Factor II/metabolism , Cell Cycle/genetics , Cell Differentiation , Cell Lineage , Coronary Vessels/metabolism , Female , Male , Mice , Sequence Analysis, RNA , Veins/metabolism
10.
Bioessays ; 43(7): e2100036, 2021 07.
Article in English | MEDLINE | ID: mdl-34145927

ABSTRACT

The establishment of distinct cellular identities was pivotal during the evolution of Metazoa, enabling the emergence of an array of specialized tissues with different functions. In most animals including vertebrates, cell specialization occurs in response to a combination of intrinsic (e.g., cellular ontogeny) and extrinsic (e.g., local environment) factors that drive the acquisition of unique characteristics at the single-cell level. The first functional organ system to form in vertebrates is the cardiovascular system, which is lined by a network of endothelial cells whose organ-specific characteristics have long been recognized. Recent genetic analyses at the single-cell level have revealed that heterogeneity exists not only at the organ level but also between neighboring endothelial cells. Thus, how endothelial heterogeneity is established has become a key question in vascular biology. Drawing upon evidence from multiple organ systems, here we will discuss the role that lineage history may play in establishing endothelial heterogeneity.


Subject(s)
Endothelial Cells , Vertebrates , Animals , Cell Proliferation , Vertebrates/genetics
11.
Eur Heart J ; 42(41): 4264-4276, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34279605

ABSTRACT

AIMS: Non-compaction cardiomyopathy is a devastating genetic disease caused by insufficient consolidation of ventricular wall muscle that can result in inadequate cardiac performance. Despite being the third most common cardiomyopathy, the mechanisms underlying the disease, including the cell types involved, are poorly understood. We have previously shown that endothelial cell-specific deletion of the chromatin remodeller gene Ino80 results in defective coronary vessel development that leads to ventricular non-compaction in embryonic mouse hearts. We aimed to identify candidate angiocrines expressed by endocardial and endothelial cells (ECs) in wildtype and LVNC conditions in Tie2Cre;Ino80fl/fltransgenic embryonic mouse hearts, and test the effect of these candidates on cardiomyocyte proliferation and maturation. METHODS AND RESULTS: We used single-cell RNA-sequencing to characterize endothelial and endocardial defects in Ino80-deficient hearts. We observed a pathological endocardial cell population in the non-compacted hearts and identified multiple dysregulated angiocrine factors that dramatically affected cardiomyocyte behaviour. We identified Col15a1 as a coronary vessel-secreted angiocrine factor, downregulated by Ino80-deficiency, that functioned to promote cardiomyocyte proliferation. Furthermore, mutant endocardial and endothelial cells up-regulated expression of secreted factors, such as Tgfbi, Igfbp3, Isg15, and Adm, which decreased cardiomyocyte proliferation and increased maturation. CONCLUSIONS: These findings support a model where coronary endothelial cells normally promote myocardial compaction through secreted factors, but that endocardial and endothelial cells can secrete factors that contribute to non-compaction under pathological conditions.


Subject(s)
Endothelial Cells , Myocytes, Cardiac , Animals , Endocardium , Heart Ventricles , Mice , Myocardium
12.
Curr Opin Hematol ; 28(3): 214-220, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33741761

ABSTRACT

PURPOSE OF REVIEW: There have been tremendous advances in the tools available for surveying blood vessels within whole organs and tissues. Here, we summarize some of the recent developments in methods for immunolabeling and imaging whole organs and provide a protocol optimized for the heart. RECENT FINDINGS: Multiple protocols have been established for chemically clearing large organs and variations are compatible with cell type-specific labeling. Heart tissue can be successfully cleared to reveal the three-dimensional structure of the entire coronary vasculature in neonatal and adult mice. Obtaining vascular reconstructions requires exceptionally large imaging files and new computational methods to process the data for accurate vascular quantifications. This is a continually advancing field that has revolutionized our ability to acquire data on larger samples as a faster rate. SUMMARY: Historically, cardiovascular research has relied heavily on histological analyses that use tissue sections, which usually sample cellular phenotypes in small regions and lack information on whole tissue-level organization. This approach can be modified to survey whole organs but image acquisition and analysis time can become unreasonable. In recent years, whole-organ immunolabeling and clearing methods have emerged as a workable solution, and new microscopy modalities, such as light-sheet microscopy, significantly improve image acquisition times. These innovations make studying the vasculature in the context of the whole organ widely available and promise to reveal fascinating new cellular behaviors in adult tissues and during repair.


Subject(s)
Cardiovascular Physiological Phenomena , Cardiovascular System/anatomy & histology , Cardiovascular System/diagnostic imaging , Diagnostic Imaging , Molecular Imaging , Research , Animals , Cardiovascular System/metabolism , Computational Biology/methods , Diagnostic Imaging/methods , Fluorescent Antibody Technique , Humans , Imaging, Three-Dimensional/methods , Mice , Molecular Imaging/methods , Organ Specificity
13.
Circulation ; 142(19): 1848-1862, 2020 11 10.
Article in English | MEDLINE | ID: mdl-32929989

ABSTRACT

BACKGROUND: Endothelial cells (ECs) display considerable functional heterogeneity depending on the vessel and tissue in which they are located. Whereas these functional differences are presumably imprinted in the transcriptome, the pathways and networks that sustain EC heterogeneity have not been fully delineated. METHODS: To investigate the transcriptomic basis of EC specificity, we analyzed single-cell RNA sequencing data from tissue-specific mouse ECs generated by the Tabula Muris consortium. We used a number of bioinformatics tools to uncover markers and sources of EC heterogeneity from single-cell RNA sequencing data. RESULTS: We found a strong correlation between tissue-specific EC transcriptomic measurements generated by either single-cell RNA sequencing or bulk RNA sequencing, thus validating the approach. Using a graph-based clustering algorithm, we found that certain tissue-specific ECs cluster strongly by tissue (eg, liver, brain), whereas others (ie, adipose, heart) have considerable transcriptomic overlap with ECs from other tissues. We identified novel markers of tissue-specific ECs and signaling pathways that may be involved in maintaining their identity. Sex was a considerable source of heterogeneity in the endothelial transcriptome and we discovered Lars2 to be a gene that is highly enriched in ECs from male mice. We found that markers of heart and lung ECs in mice were conserved in human fetal heart and lung ECs. We identified potential angiocrine interactions between tissue-specific ECs and other cell types by analyzing ligand and receptor expression patterns. CONCLUSIONS: We used single-cell RNA sequencing data generated by the Tabula Muris consortium to uncover transcriptional networks that maintain tissue-specific EC identity and to identify novel angiocrine and functional relationships between tissue-specific ECs.


Subject(s)
Databases, Nucleic Acid , Endothelial Cells/metabolism , RNA-Seq , Sex Characteristics , Single-Cell Analysis , Transcriptome , Animals , Female , Male , Mice , Organ Specificity
14.
Basic Res Cardiol ; 116(1): 19, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33742276

ABSTRACT

Endogenous capability of the post-mitotic human heart holds great promise to restore the injured myocardium. Recent evidence indicates that the extracellular vesicles (EVs) regulate cardiac homeostasis and regeneration. Here, we investigated the molecular mechanism of EVs for self-repair. We isolated EVs from human iPSC-derived cardiomyocytes (iCMs), which were exposed to hypoxic (hEVs) and normoxic conditions (nEVs), and examined their roles in in vitro and in vivo models of cardiac injury. hEV treatment significantly improved the viability of hypoxic iCMs in vitro and cardiac function of severely injured murine myocardium in vivo. Microarray analysis of the EVs revealed significantly enriched expression of the miR-106a-363 cluster (miR cluster) in hEVs vs. nEVs. This miR cluster preserved survival and contractility of hypoxia-injured iCMs and maintained murine left-ventricular (LV) chamber size, improved LV ejection fraction, and reduced myocardial fibrosis of the injured myocardium. RNA-Seq analysis identified Jag1-Notch3-Hes1 as a target intracellular pathway of the miR cluster. Moreover, the study found that the cell cycle activator and cytokinesis genes were significantly up-regulated in the iCMs treated with miR cluster and Notch3 siRNA. Together, these results suggested that the miR cluster in the EVs stimulated cardiomyocyte cell cycle re-entry by repressing Notch3 to induce cell proliferation and augment myocardial self-repair. The miR cluster may represent an effective therapeutic approach for ischemic cardiomyopathy.


Subject(s)
Cell Proliferation , Extracellular Vesicles/transplantation , Induced Pluripotent Stem Cells/transplantation , MicroRNAs/metabolism , Myocardial Infarction/surgery , Myocytes, Cardiac/metabolism , Receptor, Notch3/metabolism , Regeneration , Animals , Cell Hypoxia , Cell Line , Disease Models, Animal , Extracellular Vesicles/metabolism , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice, Inbred C57BL , Mice, SCID , MicroRNAs/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Receptor, Notch3/genetics , Recovery of Function , Signal Transduction , Ventricular Function, Left
16.
Bioessays ; 41(3): e1800198, 2019 03.
Article in English | MEDLINE | ID: mdl-30805984

ABSTRACT

A tree-like hierarchical branching structure is present in many biological systems, such as the kidney, lung, mammary gland, and blood vessels. Most of these organs form through branching morphogenesis, where outward growth results in smaller and smaller branches. However, the blood vasculature is unique in that it exists as two trees (arterial and venous) connected at their tips. Obtaining this organization might therefore require unique developmental mechanisms. As reviewed here, recent data indicate that arterial trees often form in reverse order. Accordingly, initial arterial endothelial cell differentiation occurs outside of arterial vessels. These pre-artery cells then build trees by following a migratory path from smaller into larger arteries, a process guided by the forces imparted by blood flow. Thus, in comparison to other branched organs, arteries can obtain their structure through inward growth and coalescence. Here, new information on the underlying mechanisms is discussed, and how defects can lead to pathologies, such as hypoplastic arteries and arteriovenous malformations.


Subject(s)
Arteries/embryology , Arteries/growth & development , Neovascularization, Physiologic , Veins/embryology , Veins/growth & development , Animals , Cell Differentiation/physiology , Cell Movement , Cell Plasticity , Epithelial Cells/physiology , Humans , Mice , Morphogenesis , Receptors, CXCR4/metabolism , Receptors, Notch/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish
17.
Curr Cardiol Rep ; 23(4): 30, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33655379

ABSTRACT

PURPOSE OF REVIEW: Collateral arteries create artery-artery anastomoses that could serve as natural bypasses that in the heart could relieve the various complications of ischemia heart disease. Recent work using animal models have begun to reveal details of how coronary collateral arteries form. RECENT FINDINGS: Mouse genetics has been used to study the cellular and molecular mechanisms of collateral artery development. Collateral arteries are not pre-existing in the mouse heart, and only form in response to injury. Myocardial infarction creates tissue hypoxia that triggers the expression of growth factors and chemokines that guide collaterogenesis. Collateral development is more robust in neonatal hearts when compared with adults, and contributes to neonatal heart regeneration. The identification of signaling pathways and cellular responses underlying coronary collateral artery development suggests potential translational strategies. Continued investigation into this subject could lead to the identification of targetable pathways that induce collateral arteries for cardiac revascularization.


Subject(s)
Collateral Circulation , Coronary Artery Disease , Animals , Heart , Humans , Mice , Neovascularization, Physiologic
18.
Annu Rev Physiol ; 79: 1-19, 2017 02 10.
Article in English | MEDLINE | ID: mdl-27959616

ABSTRACT

Coronary artery disease (CAD) is the number one cause of death worldwide and involves the accumulation of plaques within the artery wall that can occlude blood flow to the heart and cause myocardial infarction. The high mortality associated with CAD makes the development of medical interventions that repair and replace diseased arteries a high priority for the cardiovascular research community. Advancements in arterial regenerative medicine could benefit from a detailed understanding of coronary artery development during embryogenesis and of how these pathways might be reignited during disease. Recent research has advanced our knowledge on how the coronary vasculature is built and revealed unexpected features of progenitor cell deployment that may have implications for organogenesis in general. Here, we highlight these recent findings and discuss how they set the stage to interrogate developmental pathways during injury and disease.


Subject(s)
Cell Differentiation/physiology , Coronary Artery Disease/pathology , Coronary Vessels/growth & development , Coronary Vessels/physiology , Organogenesis/physiology , Stem Cells/physiology , Animals , Heart/physiology , Humans
19.
Development ; 144(18): 3241-3252, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28760815

ABSTRACT

How mechanotransduction intersects with chemical and transcriptional factors to shape organogenesis is an important question in developmental biology. This is particularly relevant to the cardiovascular system, which uses mechanical signals from flowing blood to stimulate cytoskeletal and transcriptional responses that form a highly efficient vascular network. Using this system, artery size and structure are tightly regulated, but the underlying mechanisms are poorly understood. Here, we demonstrate that deletion of Smad4 increased the diameter of coronary arteries during mouse embryonic development, a phenotype that followed the initiation of blood flow. At the same time, the BMP signal transducers SMAD1/5/8 were activated in developing coronary arteries. In a culture model of blood flow-induced shear stress, human coronary artery endothelial cells failed to align when either BMPs were inhibited or SMAD4 was depleted. In contrast to control cells, SMAD4-deficient cells did not migrate against the direction of shear stress and increased proliferation rates specifically under flow. Similar alterations were seen in coronary arteries in vivo Thus, endothelial cells perceive the direction of blood flow and respond through SMAD signaling to regulate artery size.


Subject(s)
Coronary Vessels/anatomy & histology , Coronary Vessels/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Mechanotransduction, Cellular , Signal Transduction , Smad Proteins/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Cell Movement , Cell Polarity , Cell Proliferation , Cell Size , Coronary Circulation , Dilatation, Pathologic , Female , Humans , Male , Mice, Inbred C57BL , Organ Size , Phosphorylation , RNA, Small Interfering/metabolism , Stress, Mechanical
20.
Circ Res ; 123(4): 443-450, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29986945

ABSTRACT

RATIONALE: Human-induced pluripotent stem cell-derived endothelial cells (iPSC-ECs) have risen as a useful tool in cardiovascular research, offering a wide gamut of translational and clinical applications. However, inefficiency of the currently available iPSC-EC differentiation protocol and underlying heterogeneity of derived iPSC-ECs remain as major limitations of iPSC-EC technology. OBJECTIVE: Here, we performed droplet-based single-cell RNA sequencing (scRNA-seq) of the human iPSCs after iPSC-EC differentiation. Droplet-based scRNA-seq enables analysis of thousands of cells in parallel, allowing comprehensive analysis of transcriptional heterogeneity. METHODS AND RESULTS: Bona fide iPSC-EC cluster was identified by scRNA-seq, which expressed high levels of endothelial-specific genes. iPSC-ECs, sorted by CD144 antibody-conjugated magnetic sorting, exhibited standard endothelial morphology and function including tube formation, response to inflammatory signals, and production of NO. Nonendothelial cell populations resulting from the differentiation protocol were identified, which included immature cardiomyocytes, hepatic-like cells, and vascular smooth muscle cells. Furthermore, scRNA-seq analysis of purified iPSC-ECs revealed transcriptional heterogeneity with 4 major subpopulations, marked by robust enrichment of CLDN5, APLNR, GJA5, and ESM1 genes, respectively. CONCLUSIONS: Massively parallel, droplet-based scRNA-seq allowed meticulous analysis of thousands of human iPSCs subjected to iPSC-EC differentiation. Results showed inefficiency of the differentiation technique, which can be improved with further studies based on identification of molecular signatures that inhibit expansion of nonendothelial cell types. Subtypes of bona fide human iPSC-ECs were also identified, allowing us to sort for iPSC-ECs with specific biological function and identity.


Subject(s)
Endothelial Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Transcriptome , Apelin Receptors/genetics , Apelin Receptors/metabolism , Cell Differentiation , Cells, Cultured , Claudin-5/genetics , Claudin-5/metabolism , Connexins/genetics , Connexins/metabolism , Endothelial Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Single-Cell Analysis , Gap Junction alpha-5 Protein
SELECTION OF CITATIONS
SEARCH DETAIL