Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Nature ; 629(8013): 901-909, 2024 May.
Article in English | MEDLINE | ID: mdl-38658756

ABSTRACT

The liver is the main gateway from the gut, and the unidirectional sinusoidal flow from portal to central veins constitutes heterogenous zones, including the periportal vein (PV) and the pericentral vein zones1-5. However, functional differences in the immune system in each zone remain poorly understood. Here intravital imaging revealed that inflammatory responses are suppressed in PV zones. Zone-specific single-cell transcriptomics detected a subset of immunosuppressive macrophages enriched in PV zones that express high levels of interleukin-10 and Marco, a scavenger receptor that sequesters pro-inflammatory pathogen-associated molecular patterns and damage-associated molecular patterns, and consequently suppress immune responses. Induction of Marco+ immunosuppressive macrophages depended on gut microbiota. In particular, a specific bacterial family, Odoribacteraceae, was identified to induce this macrophage subset through its postbiotic isoallolithocholic acid. Intestinal barrier leakage resulted in inflammation in PV zones, which was markedly augmented in Marco-deficient conditions. Chronic liver inflammatory diseases such as primary sclerosing cholangitis (PSC) and non-alcoholic steatohepatitis (NASH) showed decreased numbers of Marco+ macrophages. Functional ablation of Marco+ macrophages led to PSC-like inflammatory phenotypes related to colitis and exacerbated steatosis in NASH in animal experimental models. Collectively, commensal bacteria induce Marco+ immunosuppressive macrophages, which consequently limit excessive inflammation at the gateway of the liver. Failure of this self-limiting system promotes hepatic inflammatory disorders such as PSC and NASH.


Subject(s)
Cholangitis, Sclerosing , Gastrointestinal Microbiome , Inflammation , Liver , Macrophages , Non-alcoholic Fatty Liver Disease , Symbiosis , Animals , Female , Humans , Male , Mice , Bacteroidetes/metabolism , Cholangitis, Sclerosing/immunology , Cholangitis, Sclerosing/microbiology , Cholangitis, Sclerosing/pathology , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Gene Expression Profiling , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interleukin-10/immunology , Interleukin-10/metabolism , Liver/immunology , Liver/pathology , Liver/microbiology , Macrophages/cytology , Macrophages/immunology , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/pathology , Portal Vein , Receptors, Immunologic/deficiency , Receptors, Immunologic/metabolism , Single-Cell Analysis , Symbiosis/immunology
2.
Biol Pharm Bull ; 47(1): 232-239, 2024.
Article in English | MEDLINE | ID: mdl-38246610

ABSTRACT

Biologics are essential for treating inflammatory bowel disease (IBD); however, only a few studies have validated cost-effective treatment options and patient factors for biologic use using real-world data from Japanese patients with IBD. Here, we aimed to provide pharmacoeconomic evidence to support clinical decisions for IBD treatment using biologics. We assessed 183 cases (127 patients) of IBD treated with biologics between November 2004 and September 2021. Data on patient background, treatment other than biologics, treatment-related medical costs, and effectiveness index (ratio of the C-reactive protein-negative period to drug survival time) were analyzed using univariate and multivariate logistic regression analyses. Drug survival was determined using Kaplan-Meier survival curve analysis. The outcomes were to validate a novel assessment index and elucidate the following aspects using this index: the effectiveness-cost relationship of long-term biologic use in IBD and cost-effectiveness-associated patient factors. Body mass index ≥25 kg/m2 and duration of hypoalbuminemia during drug survival correlated significantly with the therapeutic effectiveness of biologics. There were no significant differences in surgical, granulocyte apheresis, or adverse-event costs per drug survival time. Biologic costs were significantly higher in the group showing lower effectiveness than in the group showing higher effectiveness. These findings hold major pharmacoeconomic implications for not only improving therapeutic outcomes through the amelioration of low albumin levels and obesity but also potentially reducing healthcare expenditure related to the use of biotherapeutics. To our knowledge, this is the first pharmacoeconomic study based on real-world data from Japanese patients with IBD receiving long-term biologic therapy.


Subject(s)
Biological Products , Inflammatory Bowel Diseases , Humans , Japan , Economics, Pharmaceutical , Retrospective Studies , Inflammatory Bowel Diseases/drug therapy , Biological Products/therapeutic use
3.
Nat Commun ; 14(1): 4417, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37537159

ABSTRACT

Cholesteatoma, which potentially results from tympanic membrane retraction, is characterized by intractable local bone erosion and subsequent hearing loss and brain abscess formation. However, the pathophysiological mechanisms underlying bone destruction remain elusive. Here, we performed a single-cell RNA sequencing analysis on human cholesteatoma samples and identify a pathogenic fibroblast subset characterized by abundant expression of inhibin ßA. We demonstrate that activin A, a homodimer of inhibin ßA, promotes osteoclast differentiation. Furthermore, the deletion of inhibin ßA /activin A in these fibroblasts results in decreased osteoclast differentiation in a murine model of cholesteatoma. Moreover, follistatin, an antagonist of activin A, reduces osteoclastogenesis and resultant bone erosion in cholesteatoma. Collectively, these findings indicate that unique activin A-producing fibroblasts present in human cholesteatoma tissues are accountable for bone destruction via the induction of local osteoclastogenesis, suggesting a potential therapeutic target.


Subject(s)
Cholesteatoma , Osteogenesis , Humans , Mice , Animals , Osteogenesis/genetics , Transcriptome , Activins/genetics , Activins/metabolism , Follistatin/genetics , Follistatin/metabolism , Cholesteatoma/pathology , Fibroblasts/metabolism
4.
Nat Commun ; 14(1): 143, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36650150

ABSTRACT

Alveolar macrophages (AMs) are crucial for maintaining normal lung function. They are abundant in lung cancer tissues, but their pathophysiological significance remains unknown. Here we show, using an orthotopic murine lung cancer model and human carcinoma samples, that AMs support cancer cell proliferation and thus contribute to unfavourable outcome. Inhibin beta A (INHBA) expression is upregulated in AMs under tumor-bearing conditions, leading to the secretion of activin A, a homodimer of INHBA. Accordingly, follistatin, an antagonist of activin A is able to inhibit lung cancer cell proliferation. Single-cell RNA sequence analysis identifies a characteristic subset of AMs specifically induced in the tumor environment that are abundant in INHBA, and distinct from INHBA-expressing AMs in normal lungs. Moreover, postnatal deletion of INHBA/activin A could limit tumor growth in experimental models. Collectively, our findings demonstrate the critical pathological role of activin A-producing AMs in tumorigenesis, and provides means to clearly distinguish them from their healthy counterparts.


Subject(s)
Carcinoma , Lung Neoplasms , Humans , Animals , Mice , Macrophages, Alveolar/metabolism , Activins/metabolism , Follistatin/genetics , Follistatin/metabolism , Lung/pathology , Lung Neoplasms/pathology , Carcinoma/metabolism
5.
Cancer Sci ; 113(8): 2916-2925, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35579268

ABSTRACT

Histopathological diagnosis is the ultimate method of attaining the final diagnosis; however, the observation range is limited to the two-dimensional plane, and it requires thin slicing of the tissue, which limits diagnostic information. To seek solutions for these problems, we proposed a novel imaging-based histopathological examination. We used the multiphoton excitation microscopy (MPM) technique to establish a method for visualizing unfixed/unstained human breast tissues. Under near-infrared ray excitation, fresh human breast tissues emitted fluorescent signals with three major peaks, which enabled visualizing the breast tissue morphology without any fixation or dye staining. Our study using human breast tissue samples from 32 patients indicated that experienced pathologists can estimate normal or cancerous lesions using only these MPM images with a kappa coefficient of 1.0. Moreover, we developed an image classification algorithm with artificial intelligence that enabled us to automatically define cancer cells in small areas with a high sensitivity of ≥0.942. Taken together, label-free MPM imaging is a promising method for the real-time automatic diagnosis of breast cancer.


Subject(s)
Breast Neoplasms , Artificial Intelligence , Breast , Breast Neoplasms/diagnostic imaging , Female , Humans , Microscopy, Fluorescence, Multiphoton/methods
6.
Dermatol Ther ; 35(5): e15375, 2022 05.
Article in English | MEDLINE | ID: mdl-35150057

ABSTRACT

The efficacy of biologics in psoriasis treatment is clinically proven; however, biologics are expensive. In this study, we assessed the real-world cost-effectiveness of biologics for psoriasis treatment by evaluating the relationship between biologic drug survival (DS) and total medical-treatment costs from a pharmacoeconomic viewpoint. Furthermore, the effects of patient factors on cost-effectiveness were investigated. We retrospectively reviewed the medical records of 135 cases who received either a tumor necrosis factor-alpha (TNF-α) monoclonal antibody (TNF-mab), interleukin (IL)-17 mab, or IL23p19-mab for psoriasis from January 2010 to June 2020 at Yamaguchi University Hospital. We compared the monthly medical-treatment costs according to biologic classification and found that costs of medical services, tests, and external preparations required for the treatment process were significantly higher in the TNF-mab group than in the other groups, and the total medical costs associated with TNF-mab treatment were significantly higher than those of IL17-mab treatment. The total monthly cost of medical care was lower in the long-term DS group than in the short-term group. The number of prescriptions for external preparations, comprising Vitamin D3 and corticosteroid, was significantly higher in the long-term DS group than in the short-term group; in the TNF-mab group, the proportion of patients without smoking habits was significantly higher in the long-term group as well. Our study indicated that when costly biologics are used for psoriasis treatment, the maintenance of long-term DS and appropriate patient guidance might improve the quality of medical care, thus allowing cost-effective medical care.


Subject(s)
Biological Products , Psoriasis , Antibodies, Monoclonal/therapeutic use , Biological Products/therapeutic use , Economics, Pharmaceutical , Humans , Psoriasis/diagnosis , Psoriasis/drug therapy , Retrospective Studies
7.
Biol Open ; 10(3)2021 03 26.
Article in English | MEDLINE | ID: mdl-33664097

ABSTRACT

The cell's movement and morphological change are two interrelated cellular processes. An integrated analysis is needed to explore the relationship between them. However, it has been challenging to investigate them as a whole. The cell's trajectory can be described by its speed, curvature, and torsion. On the other hand, the three-dimensional (3D) cell shape can be studied by using a shape descriptor such as spherical harmonic (SH) descriptor, which is an extension of a Fourier transform in 3D space. We propose a novel method using parallel-transport (PT) to integrate these shape-movement data by using moving frames as the 3D-shape coordinate system. This moving frame is purely determined by the velocity vector. On this moving frame, the movement change will influence the coordinate system for shape analysis. By analyzing the change of the SH coefficients over time in the moving frame, we can observe the relationship between shape and movement. We illustrate the application of our approach using simulated and real datasets in this paper.


Subject(s)
Cell Movement , Cell Physiological Phenomena , Cell Shape , Models, Biological , Algorithms , Cell Movement/genetics , Cell Shape/genetics , Computer Simulation
8.
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
9.
Int Immunol ; 32(11): 727-736, 2020 10 20.
Article in English | MEDLINE | ID: mdl-32275057

ABSTRACT

The sympathetic nervous system plays critical roles in the differentiation, maturation and recruitment of immune cells under homeostatic conditions, and in responses to environmental stimuli, although its role in the migratory control of immune cells during acute inflammation remains unclear. In this study, using an advanced intravital bone imaging system established in our laboratory, we demonstrated that the sympathetic nervous system locally regulates neutrophil egress from the bone marrow for mobilization to inflammatory foci. We found that sympathetic neurons were located close to blood vessels in the bone marrow cavity; moreover, upon lipopolysaccharide (LPS) administration, local sympathectomy delayed neutrophil egress from the bone marrow and increased the proportion of neutrophils that remained in place. We also showed that vascular endothelial cells produced C-X-C motif chemokine ligand 1 (CXCL1), which is responsible for neutrophil egress out of the bone marrow. Its expression was up-regulated during acute inflammation, and was suppressed by ß-adrenergic receptor blockade, which was accompanied with inhibition of neutrophil egress into the systemic circulation. Furthermore, systemic ß-adrenergic signaling blockade decreased the recruitment of neutrophils in the lung under conditions of acute systemic inflammation. Taken together, the results of this study first suggested a new regulatory system, wherein local sympathetic nervous activation promoted neutrophil egress by enhancing Cxcl1 expression in bone marrow endothelial cells in a ß-adrenergic signaling-dependent manner, contributing to the recruitment of neutrophils at the onset of inflammation in vivo.


Subject(s)
Bone Marrow/immunology , Inflammation/immunology , Neurons/immunology , Neutrophils/immunology , Animals , Male , Mice , Mice, Inbred C57BL
10.
Elife ; 82019 10 01.
Article in English | MEDLINE | ID: mdl-31570118

ABSTRACT

While the sphingosine-1-phosphate (S1P)/sphingosine-1-phosphate receptor-1 (S1PR1) axis is critically important for lymphocyte egress from lymphoid organs, S1PR1-activation also occurs in vascular endothelial cells (ECs), including those of the high-endothelial venules (HEVs) that mediate lymphocyte immigration into lymph nodes (LNs). To understand the functional significance of the S1P/S1PR1-Gi axis in HEVs, we generated Lyve1;Spns2Δ/Δ conditional knockout mice for the S1P-transporter Spinster-homologue-2 (SPNS2), as HEVs express LYVE1 during development. In these mice HEVs appeared apoptotic and were severely impaired in function, morphology and size; leading to markedly hypotrophic peripheral LNs. Dendritic cells (DCs) were unable to interact with HEVs, which was also observed in Cdh5CRE-ERT2;S1pr1Δ/Δ mice and wildtype mice treated with S1PR1-antagonists. Wildtype HEVs treated with S1PR1-antagonists in vitro and Lyve1-deficient HEVs show severely reduced release of the DC-chemoattractant CCL21 in vivo. Together, our results reveal that EC-derived S1P warrants HEV-integrity through autocrine control of S1PR1-Gi signaling, and facilitates concomitant HEV-DC interactions.


Subject(s)
Cell Movement , Dendritic Cells/physiology , Endothelial Cells/physiology , Lymph Nodes/cytology , Lysophospholipids/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Animals , Mice, Knockout , Sphingosine/metabolism
11.
Development ; 144(15): 2730-2736, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28694255

ABSTRACT

The molecular mechanism by which NSC number is controlled in the neurogenic regions of the adult brain is not fully understood but it has been shown that vascular niche signals regulate neural stem cell (NSC) quiescence and growth. Here, we have uncovered a role for soluble amyloid precursor protein (sAPP) as a vascular niche signal in the subventricular zone (SVZ) of the lateral ventricle of the adult mouse brain. sAPP suppresses NSC growth in culture. Further in vivo studies on the role of APP in regulating NSC number in the SVZ clearly demonstrate that endothelial deletion of App causes a significant increase in the number of BrdU label-retaining NSCs in the SVZ, whereas NSC/astrocyte deletion of App has no detectable effect on the NSC number. Taken together, these results suggest that endothelial APP functions as a vascular niche signal that negatively regulates NSC growth to control the NSC number in the SVZ.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Neural Stem Cells/cytology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Brain/cytology , Brain/metabolism , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Flow Cytometry , Immunohistochemistry , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Niche/genetics , Stem Cell Niche/physiology
12.
Cell Rep ; 18(12): 2991-3004, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28329690

ABSTRACT

Mural cells (pericytes and vascular smooth muscle cells) are essential for the regulation of vascular networks and maintenance of vascular integrity, but their origins are diverse in different tissues and not known in the organs that arise from the ectoderm, such as skin. Here, we show that tissue-localized myeloid progenitors contribute to pericyte development in embryonic skin vasculature. A series of in vivo fate-mapping experiments indicates that tissue myeloid progenitors differentiate into pericytes. Furthermore, depletion of tissue myeloid cells and their progenitors in PU.1 (also known as Spi1) mutants results in defective pericyte development. Fluorescence-activated cell sorting (FACS)-isolated myeloid cells and their progenitors from embryonic skin differentiate into pericytes in culture. At the molecular level, transforming growth factor-ß (TGF-ß) induces pericyte differentiation in culture. Furthermore, type 2 TGF-ß receptor (Tgfbr2) mutants exhibit deficient pericyte development in skin vasculature. Combined, these data suggest that pericytes differentiate from tissue myeloid progenitors in the skin vasculature through TGF-ß signaling.


Subject(s)
Cell Differentiation , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Pericytes/cytology , Signal Transduction , Skin/blood supply , Skin/embryology , Transforming Growth Factor beta/metabolism , Animals , Cell Lineage , Cells, Cultured , Dermis/cytology , Embryo, Mammalian/cytology , Hematopoiesis , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic
13.
J Clin Invest ; 126(7): 2437-51, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27214551

ABSTRACT

The lymphatic vasculature is essential for maintaining interstitial fluid homeostasis, and dysfunctional lymphangiogenesis contributes to various pathological processes, including inflammatory disease and tumor metastasis. Mutations in FOXC2 are dominantly associated with late-onset lymphedema; however, the precise role of FOXC2 and a closely related factor, FOXC1, in the lymphatic system remains largely unknown. Here we identified a molecular cascade by which FOXC1 and FOXC2 regulate ERK signaling in lymphatic vessel growth. In mice, lymphatic endothelial cell-specific (LEC-specific) deletion of Foxc1, Foxc2, or both resulted in increased LEC proliferation, enlarged lymphatic vessels, and abnormal lymphatic vessel morphogenesis. Compared with LECs from control animals, LECs from mice lacking both Foxc1 and Foxc2 exhibited aberrant expression of Ras regulators, and embryos with LEC-specific deletion of Foxc1 and Foxc2, alone or in combination, exhibited ERK hyperactivation. Pharmacological ERK inhibition in utero abolished the abnormally enlarged lymphatic vessels in FOXC-deficient embryos. Together, these results identify FOXC1 and FOXC2 as essential regulators of lymphangiogenesis and indicate a new potential mechanistic basis for lymphatic-associated diseases.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Forkhead Transcription Factors/metabolism , Gene Deletion , Gene Expression Regulation , Lymphangiogenesis , Animals , Cell Proliferation , Female , Gene Expression Profiling , Inflammation , Male , Mice , Mice, Knockout , Skin/metabolism
14.
Dev Biol ; 411(2): 183-194, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26872874

ABSTRACT

Angiogenesis, the formation of new blood vessels by remodeling and growth of pre-existing vessels, is a highly orchestrated process that requires a tight balance between pro-angiogenic and anti-angiogenic factors and the integration of their corresponding signaling networks. The family of Rho GTPases, including RhoA, Rac1, and Cdc42, play a central role in many cell biological processes that involve cytoskeletal changes and cell movement. Specifically for Rac1, we have shown that excision of Rac1 using a Tie2-Cre animal line results in embryonic lethality in midgestation (embryonic day (E) 9.5), with multiple vascular defects. However, Tie2-Cre can be also expressed during vasculogenesis, prior to angiogenesis, and is active in some hematopoietic precursors that can affect vessel formation. To circumvent these limitations, we have now conditionally deleted Rac1 in a temporally controlled and endothelial-restricted fashion using Cdh5(PAC)-iCreERT2 transgenic mice. In this highly controlled experimental in vivo system, we now show that Rac1 is required for embryonic vascular integrity and angiogenesis, and for the formation of superficial and deep vascular networks in the post-natal developing retina, the latter involving a novel specific function for Rac1 in vertical blood vessel sprouting. Aligned with these findings, we show that RAC1 is spatially involved in endothelial cell migration, invasion, and radial sprouting activities in 3D collagen matrix in vitro models. Hence, Rac1 and its downstream molecules may represent potential anti-angiogeneic therapeutic targets for the treatment of many human diseases that involve aberrant neovascularization and blood vessel overgrowth.


Subject(s)
Endothelial Cells/cytology , Gene Expression Regulation, Developmental , Neovascularization, Physiologic , Neuropeptides/physiology , Retina/embryology , Retinal Vessels/physiology , rac1 GTP-Binding Protein/physiology , Alleles , Animals , Cell Movement , Endothelium, Vascular/metabolism , Female , Genes, Reporter , Genotype , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , RNA, Small Interfering/metabolism , Retinal Vessels/embryology , rac1 GTP-Binding Protein/genetics
15.
J Thorac Cardiovasc Surg ; 151(4): 1126-35.e2, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26611747

ABSTRACT

OBJECTIVES: Patients with single ventricle congenital heart disease often form aortopulmonary collateral vessels via an unclear mechanism. To gain insights into the pathogenesis of aortopulmonary collateral vessels, we correlated angiogenic factor levels with in vitro activity and angiographic aortopulmonary collateral assessment and examined whether patients with single ventricle physiology have increased angiogenic factors that can stimulate endothelial cell sprouting in vitro. METHODS: In patients with single ventricle physiology (n = 27) and biventricular acyanotic control patients (n = 21), hypoxia-inducible angiogenic factor levels were measured in femoral venous and arterial plasma at cardiac catheterization. To assess plasma angiogenic activity, we used a 3-dimensional in vitro cell sprouting assay that recapitulates angiogenic sprouting. Aortopulmonary collateral angiograms were graded using a 4-point scale. RESULTS: Compared with controls, patients with single ventricle physiology had increased vascular endothelial growth factor (artery: 58.7 ± 1.2 pg/mL vs 35.3 ± 1.1 pg/mL, P < .01; vein: 34.8 ± 1.1 pg/mL vs 21 ± 1.2 pg/mL, P < .03), stromal-derived factor 1-alpha (artery: 1901.6 ± 1.1 pg/mL vs 1542.6 ± 1.1 pg/mL, P < .03; vein: 2092.8 pg/mL ± 1.1 vs 1752.9 ± 1.1 pg/mL, P < .02), and increased arterial soluble fms-like tyrosine kinase-1, a regulatory vascular endothelial growth factor receptor (612.3 ± 1.2 pg/mL vs 243.1 ± 1.2 pg/mL, P < .003). Plasma factors and sprout formation correlated poorly with aortopulmonary collateral severity. CONCLUSIONS: We are the first to correlate plasma angiogenic factor levels with angiography and in vitro angiogenic activity in patients with single ventricle disease with aortopulmonary collaterals. Patients with single ventricle disease have increased stromal-derived factor 1-alpha and soluble fms-like tyrosine kinase-1, and their roles in aortopulmonary collateral formation require further investigation. Plasma factors and angiogenic activity correlate poorly with aortopulmonary collateral severity in patients with single ventricles, suggesting complex mechanisms of angiogenesis.


Subject(s)
Angiogenic Proteins/blood , Aorta/physiopathology , Collateral Circulation , Endothelial Cells/metabolism , Heart Defects, Congenital/blood , Neovascularization, Physiologic , Pulmonary Artery/physiopathology , Pulmonary Circulation , Adolescent , Aortography , Case-Control Studies , Cells, Cultured , Chemokine CXCL12/blood , Child , Child, Preschool , Female , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/physiopathology , Humans , Infant , Male , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor Receptor-2/blood
16.
Biol Open ; 4(9): 1194-205, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26319580

ABSTRACT

The development of a patterned lymphatic vascular network is essential for proper lymphatic functions during organ development and homeostasis. Here we report that class 3 semaphorins (SEMA3s), SEMA3F and SEMA3G negatively regulate lymphatic endothelial cell (LEC) growth and sprouting to control dermal lymphatic network formation. Neuropilin2 (NRP2) functions as a receptor for SEMA3F and SEMA3G, as well as vascular endothelial growth factor C (VEGFC). In culture, Both SEMA3F and SEMA3G inhibit VEGFC-mediated sprouting and proliferation of human dermal LECs. In the developing mouse skin, Sema3f is expressed in the epidermis and Sema3g expression is restricted to arteries, whereas their receptor Nrp2 is preferentially expressed by lymphatic vessels. Both Sema3f;Sema3g double mutants and Nrp2 mutants exhibit increased LEC growth in the skin. In contrast, Sema3f;Sema3g double mutants display increased lymphatic branching, while Nrp2 mutants exhibit reduced lymphatic branching. A targeted mutation in PlexinA1 or PlexinA2, signal transducers forming a receptor complex with NRP2 for SEMA3s, exhibits an increase in LEC growth and lymphatic branching as observed in Sema3f;Sema3g double mutants. Our results provide the first evidence that SEMA3F and SEMA3G function as a negative regulator for dermal lymphangiogenesis in vivo. The reciprocal phenotype in lymphatic branching between Sema3f;Sema3g double mutants and Nrp2 mutants suggest a complex NRP2 function that regulates LEC behavior both positively and negatively, through a binding with VEGFC or SEMA3s.

17.
Nat Commun ; 5: 5325, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25358863

ABSTRACT

Reorganization of the actin cytoskeleton is an early cellular response to various extracellular signals. Sema3A, a repulsive axon guidance molecule, induces the reorganization of actin cytoskeleton in the growth cones. Collapsin response mediator protein 1 (CRMP1) mediates the intracellular Sema3A signalling through its Ser522 phosphorylation. Here we show that UNC-33, CRMP1 C. elegans homologue, interacts with FLN-1, an actin-binding Filamin-A orthologue. In nematodes, this interaction participates in the projection of DD/VD motor neurons. CRMP1 binds both the actin-binding domain and the last immunoglobulin-like repeat of Filamin-A. The alanine mutants of Filamin-A or CRMP1 in their interacting residues suppress the Sema3A repulsion in neurons. Conversely, a phosphor-mimicking mutant CRMP1(Ser522Asp) enhances the Sema3A response. Atomic-force microscopy analysis reveals that the V-shaped Filamin-A changes to a condensed form with CRMP1(Ser522Asp). CRMP1(Ser522Asp) weakens the F-actin gelation crosslinked by Filamin-A. Thus, phosphorylated CRMP1 may remove Filamin-A from the actin cytoskeleton to facilitate its remodelling.


Subject(s)
Actin Cytoskeleton/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Filamins/metabolism , Growth Cones/metabolism , Nerve Growth Factors/metabolism , Actins/metabolism , Animals , Caenorhabditis elegans/genetics , HEK293 Cells , Humans , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Phosphoproteins/metabolism , Rats, Wistar , Semaphorin-3A/metabolism
18.
Development ; 140(7): 1475-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23462468

ABSTRACT

Anatomical congruence of peripheral nerves and blood vessels is well recognized in a variety of tissues. Their physical proximity and similar branching patterns suggest that the development of these networks might be a coordinated process. Here we show that large diameter coronary veins serve as an intermediate template for distal sympathetic axon extension in the subepicardial layer of the dorsal ventricular wall of the developing mouse heart. Vascular smooth muscle cells (VSMCs) associate with large diameter veins during angiogenesis. In vivo and in vitro experiments demonstrate that these cells mediate extension of sympathetic axons via nerve growth factor (NGF). This association enables topological targeting of axons to final targets such as large diameter coronary arteries in the deeper myocardial layer. As axons extend along veins, arterial VSMCs begin to secrete NGF, which allows axons to reach target cells. We propose a sequential mechanism in which initial axon extension in the subepicardium is governed by transient NGF expression by VSMCs as they are recruited to coronary veins; subsequently, VSMCs in the myocardium begin to express NGF as they are recruited by remodeling arteries, attracting axons toward their final targets. The proposed mechanism underlies a distinct, stereotypical pattern of autonomic innervation that is adapted to the complex tissue structure and physiology of the heart.


Subject(s)
Coronary Vessels/physiology , Heart/embryology , Heart/innervation , Sympathetic Nervous System/embryology , Animals , Axons/physiology , Cells, Cultured , Chick Embryo , Coronary Vessels/embryology , Coronary Vessels/innervation , Embryo Culture Techniques , Embryo, Mammalian , Mice , Mice, Transgenic , Models, Biological , Muscle, Smooth, Vascular/embryology , Muscle, Smooth, Vascular/innervation , Muscle, Smooth, Vascular/metabolism , Pericardium/embryology , Pericardium/innervation , Sympathetic Nervous System/physiology
19.
Dev Cell ; 24(4): 359-71, 2013 Feb 25.
Article in English | MEDLINE | ID: mdl-23395391

ABSTRACT

In developing limb skin, peripheral nerves provide a spatial template that controls the branching pattern and differentiation of arteries. Our previous studies indicate that nerve-derived VEGF-A is required for arterial differentiation but not for nerve-vessel alignment. In this study, we demonstrate that nerve-vessel alignment depends on the activity of Cxcl12-Cxcr4 chemokine signaling. Genetic inactivation of Cxcl12-Cxcr4 signaling perturbs nerve-vessel alignment and abolishes arteriogenesis. Further in vitro assays allow us to uncouple nerve-vessel alignment and arteriogenesis, revealing that nerve-derived Cxcl12 stimulates endothelial cell migration, whereas nerve-derived VEGF-A is responsible for arterial differentiation. These findings suggest a coordinated sequential action in which nerve Cxcl12 functions over a distance to recruit vessels to align with nerves, and subsequent arterial differentiation presumably requires a local action of nerve VEGF-A in the nerve-associated vessels.


Subject(s)
Arteries/cytology , Chemokine CXCL12/physiology , Extremities/embryology , Ganglia, Spinal/metabolism , Receptors, CXCR4/physiology , Skin/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Arteries/embryology , Arteries/metabolism , Blotting, Western , Cell Differentiation , Cell Movement , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Flow Cytometry , In Situ Hybridization , Integrases/metabolism , Mice , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin/embryology , Vascular Endothelial Growth Factor A/genetics
20.
Methods Mol Biol ; 843: 69-78, 2012.
Article in English | MEDLINE | ID: mdl-22222522

ABSTRACT

We introduce a whole-mount immunohistochemistry method for analyzing intricate vascular network formation in mouse embryonic tissues. Laser scanning confocal microscopy with multiple labeling allows for robust imaging of blood and lymphatic vessel branching morphogenesis with excellent resolution.


Subject(s)
Blood Vessels/cytology , Blood Vessels/embryology , Microscopy, Confocal/methods , Animals , Cricetinae , Lymphatic Vessels/cytology , Lymphatic Vessels/embryology , Mice , Myocardium/cytology , Rabbits , Rats , Skin/blood supply , Specimen Handling , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL
...