Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Clin Ophthalmol ; 18: 2147-2154, 2024.
Article in English | MEDLINE | ID: mdl-39070106

ABSTRACT

Purpose: To evaluate the efficacy of subcutaneous injection of triamcinolone acetonide (SCTA) in treating upper eyelid retraction and swelling in patients with thyroid eye disease (TED). Patients and Methods: This case series included consecutive patients (aged 16-69 years, monitored from June 2012 to December 2015) with TED-related eyelid symptom and without an enlarged extraocular muscle on magnetic resonance imaging (MRI). SCTA (0.5 mL, 40 mg/mL) was administered to target the orbital fat around the levator palpebrae superioris (LPS) muscle. Patients who did not exhibit improvement after the first trial received an additional injection. Follow-up was conducted for 12 months with 3-month intervals. Eyelid retraction, eyelid swelling, and eyelid lag were evaluated at each follow-up visit. Results: In total, 116 eyelids of 102 patients were analyzed. SCTA led to significant improvement in 93% of eyes (108/116), disappearance of eyelid symptoms (74%, 87%, and 73% in retraction, swelling, and lag, respectively), and improvement of scores (from 1.64 to 0.12, 1.32 to 0.26, and 1.72 to 0.30, respectively). Improvement in eyelid symptoms was observed in eight eyes; however, additional steroid therapy was required in these cases due to the emergence of other extraocular muscle inflammation. Additional injection was required in 39.8% of patients. The clinical activity score was lower in the single SCTA group than in the multiple SCTA group (1.5 vs 0.9; p < 0.01). However, the levels of thyroid-stimulating hormone receptor antibody and MRI findings were not significantly different between the two groups. No elevation in intraocular pressure was observed. Eight female patients experienced menstrual disorder. Conclusion: SCTA effectively reduced LPS muscle enlargement and fat tissue swelling in patients with TED. A single SCTA was sufficient in almost 60% of the patients; nevertheless, follow-up is necessary to detect early signs of orbital inflammation even in eyelid-symptom-improved patients.

2.
Nat Commun ; 15(1): 1622, 2024 Mar 04.
Article in English | MEDLINE | ID: mdl-38438343

ABSTRACT

Alveologenesis is a spatially coordinated morphogenetic event, during which alveolar myofibroblasts surround the terminal sacs constructed by epithelial cells and endothelial cells (ECs), then contract to form secondary septa to generate alveoli in the lungs. Recent studies have demonstrated the important role of alveolar ECs in this morphogenetic event. However, the mechanisms underlying EC-mediated alveologenesis remain unknown. Herein, we show that ECs regulate alveologenesis by constructing basement membranes (BMs) acting as a scaffold for myofibroblasts to induce septa formation through activating mechanical signaling. Rap1, a small GTPase of the Ras superfamily, is known to stimulate integrin-mediated cell adhesions. EC-specific Rap1-deficient (Rap1iECKO) mice exhibit impaired septa formation and hypo-alveolarization due to the decreased mechanical signaling in myofibroblasts. In Rap1iECKO mice, ECs fail to stimulate integrin ß1 to recruit Collagen type IV (Col-4) into BMs required for myofibroblast-mediated septa formation. Consistently, EC-specific integrin ß1-deficient mice show hypo-alveolarization, defective mechanical signaling in myofibroblasts, and disorganized BMs. These data demonstrate that alveolar ECs promote integrin ß1-mediated Col-4 recruitment in a Rap1-dependent manner, thereby constructing BMs acting as a scaffold for myofibroblasts to induce mechanical signal-mediated alveologenesis. Thus, this study unveils a mechanism of organ morphogenesis mediated by ECs through intrinsic functions.


Subject(s)
Endothelial Cells , Myofibroblasts , Animals , Mice , Basement Membrane , Integrin beta1/genetics , Morphogenesis
3.
FASEB J ; 37(12): e23310, 2023 12.
Article in English | MEDLINE | ID: mdl-38010922

ABSTRACT

Vascular permeability is dynamically but tightly controlled by vascular endothelial (VE)-cadherin-mediated endothelial cell-cell junctions to maintain homeostasis. Thus, impairments of VE-cadherin-mediated cell adhesions lead to hyperpermeability, promoting the development and progression of various disease processes. Notably, the lungs are a highly vulnerable organ wherein pulmonary inflammation and infection result in vascular leakage. Herein, we showed that Rap1, a small GTPase, plays an essential role for maintaining pulmonary endothelial barrier function in mice. Endothelial cell-specific Rap1a/Rap1b double knockout mice exhibited severe pulmonary edema. They also showed vascular leakage in the hearts, but not in the brains. En face analyses of the pulmonary arteries and 3D-immunofluorescence analyses of the lungs revealed that Rap1 potentiates VE-cadherin-mediated endothelial cell-cell junctions through dynamic actin cytoskeleton reorganization. Rap1 inhibits formation of cytoplasmic actin bundles perpendicularly binding VE-cadherin adhesions through inhibition of a Rho-ROCK pathway-induced activation of cytoplasmic nonmuscle myosin II (NM-II). Simultaneously, Rap1 induces junctional NM-II activation to create circumferential actin bundles, which anchor and stabilize VE-cadherin at cell-cell junctions. We also showed that the mice carrying only one allele of either Rap1a or Rap1b out of the two Rap1 genes are more vulnerable to lipopolysaccharide (LPS)-induced pulmonary vascular leakage than wild-type mice, while activation of Rap1 by administration of 007, an activator for Epac, attenuates LPS-induced increase in pulmonary endothelial permeability in wild-type mice. Thus, we demonstrate that Rap1 plays an essential role for maintaining pulmonary endothelial barrier functions under physiological conditions and provides protection against inflammation-induced pulmonary vascular leakage.


Subject(s)
Actins , rap1 GTP-Binding Proteins , Animals , Mice , Actins/metabolism , Cadherins/metabolism , Capillary Permeability , Cell Adhesion/physiology , Endothelium, Vascular/metabolism , Lipopolysaccharides/metabolism , Lung/metabolism , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
4.
Lab Chip ; 23(2): 306-317, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36537555

ABSTRACT

Blood vessel morphology is dictated by mechanical and biochemical cues. Flow-induced shear stress and pericytes both play important roles, and they have previously been studied using on-chip vascular networks to uncover their connection to angiogenic sprouting and network stabilization. However, it is unknown which shear stress values promote angiogenesis, how pericytes are directed to sprouts, and how shear stress and pericytes affect the overall vessel morphology. Here, we employed a microfluidic device to study these phenomena in three-dimensional (3D) self-assembled vasculature. Computational fluid dynamics solver (COMSOL) simulations indicated that sprouts form most frequently at locations of relatively low shear stresses (0.5-1.5 dyn cm-2). Experimental results show that pericytes limit vascular diameter. Interestingly, when treated with imatinib or crenolanib, which are chemotherapeutic drugs and inhibitors of platelet-derived growth factor receptor ß (PDGFRß), the pericyte coverage of vessels decreased significantly but vessel diameter remained unchanged. This furthers our understanding of the mechanisms underlying vascular development and demonstrates the value of this microfluidic device in future studies on drug development and vascular biology.


Subject(s)
Lab-On-A-Chip Devices , Pericytes , Stress, Mechanical , Imatinib Mesylate/metabolism , Pericytes/metabolism
5.
Cancer Sci ; 113(10): 3579-3592, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35848891

ABSTRACT

The arachidonic acid cascade is a major inflammatory pathway that produces prostaglandin E2 (PGE2). Although inhibition of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) is reported to lead to PGE2 accumulation, the role of 15-PGDH expression in the tumor microenvironment remains unclear. We utilized Panc02 murine pancreatic cancer cells for orthotopic transplantation into wild-type and 15-pgdh+/- mice and found that 15-pgdh depletion in the tumor microenvironment leads to enhanced tumorigenesis accompanied by an increase in cancer-associated fibroblasts (CAFs) and the promotion of fibrosis. The fibrotic tumor microenvironment is widely considered to be hypovascular; however, we found that the angiogenesis level is maintained in 15-pgdh+/- mice, and these changes were also observed in a genetically engineered PDAC mouse model. Further confirmation revealed that fibroblast growth factor 1 (FGF1) is secreted by pancreatic cancer cells after PGE2 stimulation, consequently promoting CAF proliferation and vascular endothelial growth factor A (VEGFA) expression in the tumor microenvironment. Finally, in 15-pgdh+/- Acta2-TK mice, depletion of fibroblasts inhibited angiogenesis and cancer cell viability in orthotopically transplanted tumors. These findings highlighted the role of 15-pgdh downregulation in enhancing PGE2 accumulation in the pancreatic tumor microenvironment and in subsequently maintaining the angiogenesis level in fibrotic tumors along with CAF expansion.


Subject(s)
Pancreatic Neoplasms , Vascular Endothelial Growth Factor A , Animals , Arachidonic Acid , Cell Line, Tumor , Dinoprostone/metabolism , Dinoprostone/pharmacology , Fibroblast Growth Factor 1 , Fibrosis , Hydroxyprostaglandin Dehydrogenases/genetics , Hydroxyprostaglandin Dehydrogenases/metabolism , Mice , Pancreatic Neoplasms/genetics , Tumor Microenvironment , Vascular Endothelial Growth Factor A/genetics , Pancreatic Neoplasms
6.
Nat Commun ; 13(1): 2594, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35551172

ABSTRACT

Angiogenesis is regulated in coordinated fashion by chemical and mechanical cues acting on endothelial cells (ECs). However, the mechanobiological mechanisms of angiogenesis remain unknown. Herein, we demonstrate a crucial role of blood flow-driven intraluminal pressure (IP) in regulating wound angiogenesis. During wound angiogenesis, blood flow-driven IP loading inhibits elongation of injured blood vessels located at sites upstream from blood flow, while downstream injured vessels actively elongate. In downstream injured vessels, F-BAR proteins, TOCA1 and CIP4, localize at leading edge of ECs to promote N-WASP-dependent Arp2/3 complex-mediated actin polymerization and front-rear polarization for vessel elongation. In contrast, IP loading expands upstream injured vessels and stretches ECs, preventing leading edge localization of TOCA1 and CIP4 to inhibit directed EC migration and vessel elongation. These data indicate that the TOCA family of F-BAR proteins are key actin regulatory proteins required for directed EC migration and sense mechanical cell stretching to regulate wound angiogenesis.


Subject(s)
Actins , Carrier Proteins , Actin-Related Protein 2-3 Complex/metabolism , Actins/metabolism , Carrier Proteins/metabolism , Endothelial Cells/metabolism , Morphogenesis
7.
Biochem Biophys Res Commun ; 571: 201-209, 2021 09 24.
Article in English | MEDLINE | ID: mdl-34332425

ABSTRACT

Cell signaling and the following gene regulation are tightly regulated to keep homeostasis. NF-κB is a famous key transcription factor for inflammatory cell regulations that obtain a closed feedback loop with IκB. Similarly, we show here, NFAT is also tightly regulated via its downstream target, down syndrome critical region (DSCR)-1. In primary cultured endothelium, either shear stress or VEGF treatment revealed quick NFAT1 nuclear localization following the DSCR-1 transactivation, which in turn induced NFAT1 cytoplasm sequestration. Interestingly, both NFAT and DSCR-1 can be competitive substrates for calcineurin phosphatase and DSCR-1 is known to unstable protein, which caused NFAT1-nucleocytoplasmic damped oscillation via sustained shear stress or VEGF stimulation in endothelial cell (EC)s. To understand the molecular mechanism underlying the NFAT1 oscillation, we built a mathematical model of spatiotemporal regulation of NFAT1 combined with calcineurin and DSCR-1. Theoretically, manipulation of DSCR-1 expression in simulation predicted that DSCR-1 reduction would cause nuclear retention of dephosphorylated NFAT1 and disappearance of NFAT1 oscillation. To confirm this in ECs, DSCR-1 knockdown analysis was performed. DSCR-1 reduction indeed increased dephosphorylated NFAT1 in both the nucleus and cytoplasm, which eventually led to nuclear retention of NFAT1. Taken together, these studies suggest that DSCR-1 is a responsible critical factor for NFAT1 nucleocytoplasmic oscillation in shear stress or VEGF treated ECs. Our mathematical model successfully reproduced the experimental observations of NFAT1 dynamics. Combined mathematical and experimental approaches would provide a quantitative understanding way for the spatiotemporal NFAT1 feedback system.


Subject(s)
Calcineurin/metabolism , DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Muscle Proteins/metabolism , NFATC Transcription Factors/metabolism , Active Transport, Cell Nucleus , Cell Line , Humans , Signal Transduction
8.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Article in English | MEDLINE | ID: mdl-33941693

ABSTRACT

Along with blood vessels, lymphatic vessels play an important role in the circulation of body fluid and recruitment of immune cells. Postnatal lymphangiogenesis commonly occurs from preexisting lymphatic vessels by sprouting, which is induced by lymphangiogenic factors such as vascular endothelial growth factor C (VEGF-C). However, the key signals and cell types that stimulate pathological lymphangiogenesis, such as human cystic lymphangioma, are less well known. Here, we found that mouse dermal fibroblasts that infiltrate to sponges subcutaneously implanted express VEGF-D and sushi, Von Willebrand factor type A, EGF, and pentraxin domain containing 1 (SVEP1) in response to PDGFRß signal. In vitro, Pdgfrb knockout (ß-KO) fibroblasts had reduced expression of VEGF-D and SVEP1 and overproduced Amphiregulin. Dysregulation of these three factors was involved in the cyst-like and uneven distribution of lymphatic vessels observed in the ß-KO mice. Similarly, in human cystic lymphangioma, which is one of the intractable diseases and mostly occurs in childhood, fibroblasts surrounding cystic lymphatics highly expressed Amphiregulin. Moreover, fibroblast-derived Amphiregulin could induce the expression of Amphiregulin in lymphatic endothelial cells. The dual source of Amphiregulin activated EGFR expressed on the lymphatic endothelial cells. This exacerbation cascade induced proliferation of lymphatic endothelial cells to form cystic lymphangioma. Ultimately, excessive Amphiregulin produced by fibroblasts surrounding lymphatics and by lymphatic endothelial cells per se results in pathogenesis of cystic lymphangioma and will be a fascinating therapeutic target of cystic lymphangioma.


Subject(s)
Amphiregulin/metabolism , Amphiregulin/pharmacology , Lymphangiogenesis/drug effects , Lymphangiogenesis/physiology , Lymphangioma, Cystic/metabolism , Amphiregulin/genetics , Animals , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lymphangioma, Cystic/genetics , Lymphangioma, Cystic/pathology , Lymphatic Vessels/metabolism , Male , Mice , Mice, Knockout , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D
9.
Nat Metab ; 3(2): 196-210, 2021 02.
Article in English | MEDLINE | ID: mdl-33619377

ABSTRACT

Ketone bodies are generated in the liver and allow for the maintenance of systemic caloric and energy homeostasis during fasting and caloric restriction. It has previously been demonstrated that neonatal ketogenesis is activated independently of starvation. However, the role of ketogenesis during the perinatal period remains unclear. Here, we show that neonatal ketogenesis plays a protective role in mitochondrial function. We generated a mouse model of insufficient ketogenesis by disrupting the rate-limiting hydroxymethylglutaryl-CoA synthase 2 enzyme gene (Hmgcs2). Hmgcs2 knockout (KO) neonates develop microvesicular steatosis within a few days of birth. Electron microscopic analysis and metabolite profiling indicate a restricted energy production capacity and accumulation of acetyl-CoA in Hmgcs2 KO mice. Furthermore, acetylome analysis of Hmgcs2 KO cells revealed enhanced acetylation of mitochondrial proteins. These findings suggest that neonatal ketogenesis protects the energy-producing capacity of mitochondria by preventing the hyperacetylation of mitochondrial proteins.


Subject(s)
Energy Metabolism/physiology , Ketone Bodies/biosynthesis , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , 3-Hydroxybutyric Acid/metabolism , Acetylation , Animals , Animals, Newborn , Hydroxymethylglutaryl-CoA Synthase/genetics , Hydroxymethylglutaryl-CoA Synthase/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , Microvessels/physiology , Oxygen Consumption
10.
PLoS One ; 15(10): e0240552, 2020.
Article in English | MEDLINE | ID: mdl-33112918

ABSTRACT

A lack of perfusion has been one of the most significant obstacles for three-dimensional culture systems of organoids and embryonic tissues. Here, we developed a simple and reliable method to implement a perfusable capillary network in vitro. The method employed the self-organization of endothelial cells to generate a capillary network and a static pressure difference for culture medium circulation, which can be easily introduced to standard biological laboratories and enables long-term cultivation of vascular structures. Using this culture system, we perfused the lumen of the self-organized capillary network and observed a flow-induced vascular remodeling process, cell shape changes, and collective cell migration. We also observed an increase in cell proliferation around the self-organized vasculature induced by flow, indicating functional perfusion of the culture medium. We also reconstructed extravasation of tumor and inflammatory cells, and circulation inside spheroids including endothelial cells and human lung fibroblasts. In conclusion, this system is a promising tool to elucidate the mechanisms of various biological processes related to vascular flow.


Subject(s)
Cell Culture Techniques/methods , Perfusion , Tissue Engineering/methods , Animals , Cells, Cultured , Fibroblasts , Human Umbilical Vein Endothelial Cells , Humans , Mice
11.
EMBO J ; 39(12): e102930, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32347571

ABSTRACT

During angiogenesis, VEGF acts as an attractive cue for endothelial cells (ECs), while Sema3E mediates repulsive cues. Here, we show that the small GTPase RhoJ integrates these opposing signals in directional EC migration. In the GTP-bound state, RhoJ interacts with the cytoplasmic domain of PlexinD1. Upon Sema3E stimulation, RhoJ released from PlexinD1 induces cell contraction. PlexinD1-bound RhoJ further facilitates Sema3E-induced PlexinD1-VEGFR2 association, VEGFR2 transphosphorylation at Y1214, and p38 MAPK activation, leading to reverse EC migration. Upon VEGF stimulation, RhoJ is required for the formation of the holoreceptor complex comprising VEGFR2, PlexinD1, and neuropilin-1, thereby preventing degradation of internalized VEGFR2, prolonging downstream signal transductions via PLCγ, Erk, and Akt, and promoting forward EC migration. After conversion to the GDP-bound state, RhoJ shifts from PlexinD1 to VEGFR2, which then terminates the VEGFR2 signals. RhoJ deficiency in ECs efficiently suppressed aberrant angiogenesis in ischemic retina. These findings suggest that distinct Rho GTPases may act as context-dependent integrators of chemotactic cues in directional cell migration and may serve as candidate therapeutic targets to manipulate cell motility in disease or tissue regeneration.


Subject(s)
Cell Movement , Endothelial Cells/metabolism , Signal Transduction , rho GTP-Binding Proteins/metabolism , Animals , Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , rho GTP-Binding Proteins/genetics
12.
Biomaterials ; 229: 119547, 2020 01.
Article in English | MEDLINE | ID: mdl-31710953

ABSTRACT

Tumor vasculature creates a hostile tumor microenvironment (TME) in vivo and nourishes cancers, resulting in cancer progression and drug resistance. To mimic the biochemical and biomechanical environments of tumors in vitro, several models integrated with a vascular network have been reported. However, the tumor responses to biochemical and biomechanical stimuli were evaluated under static conditions and failed to incorporate the effects of blood flow to tumors. In this study, we present a tumor-on-a-chip platform that enables the evaluation of tumor activities with intraluminal flow in an engineered tumor vascular network. The fibroblasts in the tumor spheroid induced angiogenic sprouts, which constructed a perfusable vascular network in a tumor spheroid. The perfusability of the engineered vascular network was preserved during the culture. Moreover, perfusion for over 24 h significantly increased the proliferation activities of tumor cells and decreased cell death in the spheroid. Drug administration under perfusion condition did not show the dose-dependent effects of anticancer drugs on tumor activities in contrast to the results under static conditions. Our results demonstrate the importance of flow in a vascular network for the evaluation of tumor activities in a drug screening platform.


Subject(s)
Neoplasms , Pharmaceutical Preparations , Humans , Lab-On-A-Chip Devices , Neoplasms/drug therapy , Perfusion , Tumor Microenvironment
13.
Adv Exp Med Biol ; 1012: 41-48, 2018.
Article in English | MEDLINE | ID: mdl-29956193

ABSTRACT

Hypertension is a common noncommunicable disease. According to the World Health Organization, 1.13 billion people were suffering from hypertension in the year 2015. High blood pressure, hypertension, has a multifactorial etiology. Arterial atherosclerotic changes, systolic or diastolic dysfunction of the heart, and other noncardiac factors are involved. Epidemiological evidence has revealed that perinatal growth disturbance elevates the prevalence of hypertension. However, the specific effects of developmental disturbances on the pathological process of hypertension are poorly understood. Recently, it has become apparent that the perinatal period plays many essential roles in cardiovascular development. In this chapter, we focus on the perinatal development of the cardiovascular system, especially in murine models. Individual organs, blood, blood vessels, and the heart show unique growth characteristics during this period. We also introduce evidence from related clinical studies regarding the developmental origins of hypertension. Finally, evidence from several animal models is presented to reveal the effects of developmental disturbance or stress on arterial pathology. Improving our understanding of both developmental events and the results of clinical studies will give fresh insight into the fetal origins of hypertension.


Subject(s)
Fetus/physiopathology , Hypertension/etiology , Prenatal Exposure Delayed Effects/physiopathology , Animals , Blood Pressure/physiology , Cardiovascular System/embryology , Cardiovascular System/growth & development , Cardiovascular System/physiopathology , Female , Fetus/pathology , Humans , Hypertension/epidemiology , Hypertension/physiopathology , Pregnancy
14.
J Vis Exp ; (134)2018 04 04.
Article in English | MEDLINE | ID: mdl-29683439

ABSTRACT

A spheroid (a multicellular aggregate) is regarded as a good model of living tissues in the human body. Despite the significant advancement in the spheroid cultures, a perfusable vascular network in the spheroids remains a critical challenge for long-term culture required to maintain and develop their functions, such as protein expressions and morphogenesis. The protocol presents a novel method to integrate a perfusable vascular network within the spheroid in a microfluidic device. To induce a perfusable vascular network in the spheroid, angiogenic sprouts connected to microchannels were guided to the spheroid by utilizing angiogenic factors from human lung fibroblasts cultured in the spheroid. The angiogenic sprouts reached the spheroid, merged with the endothelial cells co-cultured in the spheroid, and formed a continuous vascular network. The vascular network could perfuse the interior of the spheroid without any leakage. The constructed vascular network may be further used as a route for supply of nutrients and removal of waste products, mimicking blood circulation in vivo. The method provides a new platform in spheroid culture toward better recapitulation of living tissues.


Subject(s)
Lab-On-A-Chip Devices , Neovascularization, Physiologic/physiology , Tissue Culture Techniques/methods , Tissue Engineering/methods , Humans
15.
J Am Heart Assoc ; 7(6)2018 03 23.
Article in English | MEDLINE | ID: mdl-29572323

ABSTRACT

BACKGROUND: Collateral arteries provide an alternative blood supply and protect tissues from ischemic damage in patients with peripheral artery disease. However, the mechanism of collateral artery development is difficult to validate. METHODS AND RESULTS: Collateral arteries were visualized using micro-x-ray computed tomography. Developmental characteristics were assessed using confocal microscopy. We conducted a single-center, retrospective, observational study and assessed the dilatation of collateral arteries on ischemic sides. We quantified the vascular volume in both ischemic and nonischemic legs. A prominent increase in vascular volume was observed in the ischemic leg using a murine hind-limb ischemia model. We also performed qualitative assessment and confirmed that the inferior gluteal artery functioned as a major collateral source. Serial analysis of murine hind-limb vessel development revealed that the inferior gluteal artery was a remnant of the ischial artery, which emerged as a representative vessel on the dorsal side during hind-limb organogenesis. We retrospectively analyzed consecutive patients who were admitted for the diagnosis or treatment of peripheral artery disease. The diameter of the inferior gluteal artery on the ischemic side showed significant dilatation compared with that on the nonischemic side. CONCLUSIONS: Our findings indicate that an embryonic remnant artery can become a collateral source under ischemic conditions. Flow enhancement in the inferior gluteal artery might become a novel therapeutic approach for patients with peripheral artery disease.


Subject(s)
Arteries/diagnostic imaging , Collateral Circulation , Computed Tomography Angiography/methods , Ischemia/diagnostic imaging , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Peripheral Arterial Disease/diagnostic imaging , X-Ray Microtomography , Aged , Animals , Arteries/physiopathology , Blood Flow Velocity , Disease Models, Animal , Female , Hindlimb , Humans , Ischemia/physiopathology , Laser-Doppler Flowmetry , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Middle Aged , Peripheral Arterial Disease/physiopathology , Regional Blood Flow , Retrospective Studies
16.
Angiogenesis ; 21(2): 415-423, 2018 05.
Article in English | MEDLINE | ID: mdl-29478089

ABSTRACT

Bone morphogenetic protein 9 (BMP9)/BMP10-ALK1 receptor signaling is essential for endothelial differentiation and vascular morphogenesis. Mutations in ALK1/ACVRL1 and other signal-related genes are implicated in human vascular diseases, and the Alk1/Acvrl1 deletion in mice causes severe impairment of vascular formation and embryonic lethality. In the microarray screen to search for novel downstream genes of ALK1 signaling, we found that the mRNA and protein expression of serum/glucocorticoid-regulated kinase 1 (SGK1) was rapidly up-regulated by the BMP9 stimulation of cultured human endothelial cells. The increase in SGK1 mRNA was completely blocked by the transcriptional inhibitor actinomycin D and significantly suppressed by the siRNA treatment against the co-SMAD transcription factor SMAD4. Upon the BMP9 treatment of endothelial cells, phosphorylated SMAD1/5/9 bound to a consensus site upstream of the SGK1 gene, which was necessary for BMP9-dependent increment of the luciferase reporter activity driven by the SGK1 proximal enhancer. The Sgk1 mRNA expression in mouse embryos was enriched in vascular endothelial cells at embryonic day 9.0-9.5, at which Sgk1 null mice showed embryonic lethality due to abnormal vascular formation, and its mRNA as well as protein expression was clearly reduced in Alk1/Acvrl1 null embryos. These results indicate that SGK1 is a novel target gene of BMP9/BMP10-ALK1 signaling in endothelial cells and further suggest a possibility that down-regulation of the Sgk1 expression may be involved in the mechanisms of vascular defects by the ALK1 signaling deficiency.


Subject(s)
Activin Receptors, Type I/metabolism , Growth Differentiation Factor 2/metabolism , Growth Differentiation Factors/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Immediate-Early Proteins/metabolism , Neovascularization, Physiologic , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription, Genetic , Activin Receptors, Type I/genetics , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Cell Line , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Growth Differentiation Factor 2/genetics , Growth Differentiation Factors/genetics , Human Umbilical Vein Endothelial Cells/cytology , Humans , Immediate-Early Proteins/genetics , Mice , Mutation , Protein Serine-Threonine Kinases/genetics
17.
Cancer Sci ; 109(1): 15-23, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28892582

ABSTRACT

We studied angiogenesis using mathematical models describing the dynamics of tip cells. We reviewed the basic ideas of angiogenesis models and its numerical simulation technique to produce realistic computer graphics images of sprouting angiogenesis. We examined the classical model of Anderson-Chaplain using fundamental concepts of mass transport and chemical reaction with ECM degradation included. We then constructed two types of numerical schemes, model-faithful and model-driven ones, where new techniques of numerical simulation are introduced, such as transient probability, particle velocity, and Boolean variables.


Subject(s)
Extracellular Matrix/pathology , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factors/metabolism , Computer Simulation , Extracellular Matrix/metabolism , Humans , Models, Biological , Models, Theoretical , Neovascularization, Pathologic/metabolism , Tumor Microenvironment
18.
Integr Biol (Camb) ; 9(6): 506-518, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28561127

ABSTRACT

Creating vascular networks in tissues is crucial for tissue engineering. Although recent studies have demonstrated the formation of vessel-like structures in a tissue model, long-term culture is still challenging due to the lack of active perfusion in vascular networks. Here, we present a method to create a three-dimensional cellular spheroid with a perfusable vascular network in a microfluidic device. By the definition of the cellular interaction between human lung fibroblasts (hLFs) in a spheroid and human umbilical vein endothelial cells (HUVECs) in microchannels, angiogenic sprouts were induced from microchannels toward the spheroid; the sprouts reached the vessel-like structures in a spheroid to form a continuous lumen. We demonstrated that the vascular network could administer biological substances to the interior of the spheroid. As cell density in the spheroid is similar to that of a tissue, the perfusable vasculature model opens up new possibilities for a long-term tissue culture in vitro.


Subject(s)
Blood Vessels/growth & development , Lab-On-A-Chip Devices , Neovascularization, Physiologic , Tissue Engineering/instrumentation , Blood Vessels/cytology , Coculture Techniques , Equipment Design , Fibroblasts/cytology , Fluorescent Dyes , Human Umbilical Vein Endothelial Cells , Humans , Lung/cytology , Perfusion , Spheroids, Cellular/cytology , Tissue Culture Techniques , Tissue Engineering/methods
19.
Cell Rep ; 13(9): 1814-27, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655898

ABSTRACT

Angiogenesis is a multicellular phenomenon driven by morphogenetic cell movements. We recently reported morphogenetic vascular endothelial cell (EC) behaviors to be dynamic and complex. However, the principal mechanisms orchestrating individual EC movements in angiogenic morphogenesis remain largely unknown. Here we present an experiment-driven mathematical model that enables us to systematically dissect cellular mechanisms in branch elongation. We found that cell-autonomous and coordinated actions governed these multicellular behaviors, and a cell-autonomous process sufficiently illustrated essential features of the morphogenetic EC dynamics at both the single-cell and cell-population levels. Through refining our model and experimental verification, we further identified a coordinated mode of tip EC behaviors regulated via a spatial relationship between tip and follower ECs, which facilitates the forward motility of tip ECs. These findings provide insights that enhance our mechanistic understanding of not only angiogenic morphogenesis, but also other types of multicellular phenomenon.


Subject(s)
Models, Biological , Animals , Aorta/cytology , Aorta/metabolism , Cell Movement/drug effects , Embryo, Nonmammalian/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Retina/drug effects , Retina/metabolism , Time-Lapse Imaging , Vascular Endothelial Growth Factor A/pharmacology , Zebrafish/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL