Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Exp Mol Med ; 55(2): 470-484, 2023 02.
Article in English | MEDLINE | ID: mdl-36828931

ABSTRACT

Tumor progression is intimately associated with the vasculature, as tumor proliferation induces angiogenesis and tumor cells metastasize to distant organs via blood vessels. However, whether tumor invasion is associated with blood vessels remains unknown. As glioblastoma (GBM) is featured by aggressive invasion and vascular abnormalities, we characterized the onset of vascular remodeling in the diffuse tumor infiltrating zone by establishing new spontaneous GBM models with robust invasion capacity. Normal brain vessels underwent a gradual transition to severely impaired tumor vessels at the GBM periphery over several days. Increasing vasodilation from the tumor periphery to the tumor core was also found in human GBM. The levels of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) showed a spatial correlation with the extent of vascular abnormalities spanning the tumor-invading zone. Blockade of VEGFR2 suppressed vascular remodeling at the tumor periphery, confirming the role of VEGF-VEGFR2 signaling in the invasion-associated vascular transition. As angiopoietin-2 (ANGPT2) was expressed in only a portion of the central tumor vessels, we developed a ligand-independent tunica interna endothelial cell kinase 2 (Tie2)-activating antibody that can result in Tie2 phosphorylation in vivo. This agonistic anti-Tie2 antibody effectively normalized the vasculature in both the tumor periphery and tumor center, similar to the effects of VEGFR2 blockade. Mechanistically, this antibody-based Tie2 activation induced VE-PTP-mediated VEGFR2 dephosphorylation in vivo. Thus, our study reveals that the normal-to-tumor vascular transition is spatiotemporally associated with GBM invasion and may be controlled by Tie2 activation via a novel mechanism of action.


Subject(s)
Glioblastoma , Humans , Glioblastoma/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Remodeling , Signal Transduction , Vascular Endothelial Growth Factors
2.
Circ Res ; 131(10): 792-806, 2022 10 28.
Article in English | MEDLINE | ID: mdl-36205124

ABSTRACT

BACKGROUND: In large-scale genomic studies, Sox17, an endothelial-specific transcription factor, has been suggested as a putative causal gene of pulmonary arterial hypertension (PAH); however, its role and molecular mechanisms remain to be elucidated. We investigated the functional impacts and acting mechanisms of impaired Sox17 (SRY-related HMG-box17) pathway in PAH and explored its potential as a therapeutic target. METHODS: In adult mice, Sox17 deletion in pulmonary endothelial cells (ECs) induced PAH under hypoxia with high penetrance and severity, but not under normoxia. RESULTS: Key features of PAH, such as hypermuscularization, EC hyperplasia, and inflammation in lung arterioles, right ventricular hypertrophy, and elevated pulmonary arterial pressure, persisted even after long rest in normoxia. Mechanistically, transcriptomic profiling predicted that the combination of Sox17 deficiency and hypoxia activated c-Met signaling in lung ECs. HGF (hepatocyte grow factor), a ligand of c-Met, was upregulated in Sox17-deficient lung ECs. Pharmacologic inhibition of HGF/c-Met signaling attenuated and reversed the features of PAH in both preventive and therapeutic settings. Similar to findings in animal models, Sox17 levels in lung ECs were repressed in 26.7% of PAH patients (4 of 15), while those were robust in all 14 non-PAH controls. HGF levels in pulmonary arterioles were increased in 86.7% of patients with PAH (13 of 15), but none of the controls showed that pattern. CONCLUSIONS: The downregulation of Sox17 levels in pulmonary arterioles increases the susceptibility to PAH, particularly when exposed to hypoxia. Our findings suggest the reactive upregulation of HGF/c-Met signaling as a novel druggable target for PAH treatment.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Animals , Mice , Endothelial Cells/metabolism , HMGB Proteins/metabolism , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypoxia/complications , Hypoxia/metabolism , Pulmonary Arterial Hypertension/genetics , Pulmonary Artery/metabolism , Signal Transduction , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Proto-Oncogene Proteins c-met/metabolism
3.
FASEB Bioadv ; 4(8): 547-559, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35949509

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) is commonly used to treat patients with various blood disorders, genetic and immunological diseases, and solid tumors. Several systemic complications following HSCT are critical limiting factors for achieving a successful outcome. These systemic complications are mainly due to the lack of initial engraftment after transplantation. However, the detailed underlying cellular dynamics of early engraftment have not been fully characterized yet. We performed in vivo longitudinal visualization of early engraftment characteristics of transplanted hematopoietic stem and progenitor cells (HSPCs) in the mouse calvarial bone marrow (BM). To achieve this, we utilized an in vivo laser-scanning confocal microscopy imaging system with a cranial BM imaging window and stereotaxic device. We observed two distinct cellular behaviors of HSPCs in vivo, cluster formation and cluster dissociation, early after transplantation. Furthermore, we successfully identified three cellular phases of engraftment with distinct cellular distances which are coordinated with cell proliferation and cell migration dynamics during initial engraftment.

4.
Adv Wound Care (New Rochelle) ; 9(11): 591-601, 2020 11.
Article in English | MEDLINE | ID: mdl-33095124

ABSTRACT

Objective: This study aimed to elucidate the role of the proangiogenic transcription factors Sox7 and Sox17 in the wound healing process and investigate the therapeutic potential of Dll4 blockade, which is an upstream regulator of Sox17, for the treatment of nonhealing wounds. Approach: After generating a full-thickness skin defect wound model of endothelial Sox7- and/or Sox17-deficient mice, we measured the wound healing rates and performed histological analysis. The effects of an anti-Dll4 antibody on wound angiogenesis in Sox7-deficient mice and db/db diabetic mice were assessed. Results: Sox7 and/or Sox17 deletion delayed wound healing. Moreover, the loss of Sox7 and Sox17 inhibited wound angiogenesis, without affecting the expression of the other. Of interest, after anti-Dll4 antibody treatment, Sox17 levels were increased and the suppression of angiogenesis was alleviated in Sox7-deficient mice and db/db diabetic mice. Consequently, Dll4 blockade effectively recovered the observed delay in wound healing. Innovation: The proangiogenic role of Sox7 and Sox17 in wound angiogenesis was addressed and effective treatment of nonhealing wounds by Dll4 blockade was suggested. Conclusion: This study revealed the proangiogenic role of the transcription factors Sox7 and Sox17 in wound angiogenesis. Furthermore, we suggest a novel method for treating nonhealing wounds by particularly targeting the Dll4-Sox17 axis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Neovascularization, Pathologic/drug therapy , Neovascularization, Physiologic/physiology , SOXF Transcription Factors/metabolism , Wound Healing/drug effects , Adaptor Proteins, Signal Transducing , Angiogenesis Inhibitors/pharmacology , Animals , Calcium-Binding Proteins , HMGB Proteins/deficiency , Male , Mice , Mice, Inbred NOD , Morphogenesis/genetics , SOXF Transcription Factors/deficiency , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism
5.
Nat Commun ; 11(1): 3866, 2020 07 31.
Article in English | MEDLINE | ID: mdl-32737287

ABSTRACT

Upon severe head injury (HI), blood vessels of the meninges and brain parenchyma are inevitably damaged. While limited vascular regeneration of the injured brain has been studied extensively, our understanding of meningeal vascular regeneration following head injury is quite limited. Here, we identify key pathways governing meningeal vascular regeneration following HI. Rapid and complete vascular regeneration in the meninges is predominantly driven by VEGFR2 signaling. Substantial increase of VEGFR2 is observed in both human patients and mouse models of HI, and endothelial cell-specific deletion of Vegfr2 in the latter inhibits meningeal vascular regeneration. We further identify the facilitating, stabilizing and arresting roles of Tie2, PDGFRß and Dll4 signaling, respectively, in meningeal vascular regeneration. Prolonged inhibition of this angiogenic process following HI compromises immunological and stromal integrity of the injured meninges. These findings establish a molecular framework for meningeal vascular regeneration after HI, and may guide development of wound healing therapeutics.


Subject(s)
Craniocerebral Trauma/genetics , Endothelial Cells/metabolism , Neovascularization, Physiologic/genetics , Regeneration/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cerebrovascular Circulation , Craniocerebral Trauma/metabolism , Craniocerebral Trauma/pathology , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation/genetics , Humans , Macrophages/metabolism , Macrophages/pathology , Meninges/injuries , Meninges/metabolism , Mice , Mice, Knockout , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing/genetics
6.
Cancers (Basel) ; 12(3)2020 Feb 27.
Article in English | MEDLINE | ID: mdl-32120820

ABSTRACT

Manipulating autophagy is a promising strategy for treating cancer as several autophagy inhibitors are shown to induce autophagic cell death. One of these, autophagonizer (APZ), induces apoptosis-independent cell death by binding an unknown target via an unknown mechanism. To identify APZ targets, we used a label-free drug affinity responsive target stability (DARTS) approach with a liquid chromatography/tandem mass spectrometry (LC-MS/MS) readout. Of 35 protein interactors, we identified Hsp70 as a key target protein of unmodified APZ in autophagy. Either APZ treatment or Hsp70 inhibition attenuates integrity of lysosomes, which leads to autophagic cell death exhibiting an excellent synergism with a clinical drug, temozolomide, in vitro, in vivo, and orthotropic glioma xenograft model. These findings demonstrate the potential of APZ to induce autophagic cell death and its development to combinational chemotherapeutic agent for glioma treatment. Collectively, our study demonstrated that APZ, a new autophagy inhibitor, can be used as a potent antitumor drug candidate to get over unassailable glioma and revealed a novel function of Hsp70 in lysosomal integrity regulation of autophagy.

7.
Circ Res ; 126(6): 767-783, 2020 03 13.
Article in English | MEDLINE | ID: mdl-32078435

ABSTRACT

RATIONALE: Central nervous system has low vascular permeability by organizing tight junction (TJ) and limiting endothelial transcytosis. While TJ has long been considered to be responsible for vascular barrier in central nervous system, suppressed transcytosis in endothelial cells is now emerging as a complementary mechanism. Whether transcytosis regulation is independent of TJ and its dysregulation dominantly causes diseases associated with edema remain elusive. Dll4 signaling is important for various vascular contexts, but its role in the maintenance of vascular barrier in central nervous system remains unknown. OBJECTIVE: To find a TJ-independent regulatory mechanism selective for transcytosis and identify its dysregulation as a cause of pathological leakage. METHODS AND RESULTS: We studied transcytosis in the adult mouse retina with low vascular permeability and employed a hypertension-induced retinal edema model for its pathological implication. Both antibody-based and genetic inactivation of Dll4 or Notch1 induce hyperpermeability by increasing transcytosis without junctional destabilization in arterial endothelial cells, leading to nonhemorrhagic leakage predominantly in the superficial retinal layer. Endothelial Sox17 deletion represses Dll4 in retinal arteries, phenocopying Dll4 blocking-driven vascular leakage. Ang II (angiotensin II)-induced hypertension represses arterial Sox17 and Dll4, followed by transcytosis-driven retinal edema, which is rescued by a gain of Notch activity. Transcriptomic profiling of retinal endothelial cells suggests that Dll4 blocking activates SREBP1 (sterol regulatory element-binding protein 1)-mediated lipogenic transcription and enriches gene sets favorable for caveolae formation. Profiling also predicts the activation of VEGF (vascular endothelial growth factor) signaling by Dll4 blockade. Inhibition of SREBP1 or VEGF-VEGFR2 (VEGF receptor 2) signaling attenuates both Dll4 blockade-driven and hypertension-induced retinal leakage. CONCLUSIONS: In the retina, Sox17-Dll4-SREBP1 signaling axis controls transcytosis independently of TJ in superficial arteries among heterogeneous regulations for the whole vessels. Uncontrolled transcytosis via dysregulated Dll4 underlies pathological leakage in hypertensive retina and could be a therapeutic target for treating hypertension-associated retinal edema.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Blood-Retinal Barrier/metabolism , Calcium-Binding Proteins/metabolism , Hypertensive Retinopathy/metabolism , Transcytosis , Adaptor Proteins, Signal Transducing/genetics , Animals , Arteries/metabolism , Calcium-Binding Proteins/genetics , Caveolae/metabolism , Endothelial Cells/metabolism , HMGB Proteins/metabolism , Homeostasis , Mice , Mice, Inbred C57BL , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , SOXF Transcription Factors/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , Tight Junctions/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
8.
J Allergy Clin Immunol ; 144(2): 561-573.e6, 2019 08.
Article in English | MEDLINE | ID: mdl-30928652

ABSTRACT

BACKGROUND: IL-33, levels of which are known to be increased in patients with eosinophilic asthma and which is suggested as a therapeutic target for it, activates endothelial cells in which Sry-related high-mobility-group box (Sox) 17, an endothelium-specific transcription factor, was upregulated. OBJECTIVE: We investigated the relationship between Sox17 and IL-33 and the possible role of Sox17 in the pathogenesis of asthma using a mouse model of airway inflammation. METHODS: We used ovalbumin (OVA) to induce airway inflammation in endothelium-specific Sox17 null mutant mice and used IL-33 neutralizing antibody to evaluate the interplay between IL-33 and Sox17. We evaluated airway inflammation and measured levels of various cytokines, chemokines, and adhesion molecules. We also carried out loss- or gain-of-function experiments for Sox17 in human endothelial cells. RESULTS: Levels of IL-33 and Sox17 were significantly increased in the lungs of OVA-challenged mice. Anti-IL-33 neutralizing antibody treatment attenuated not only OVA-induced airway inflammation but also Sox17 expression in pulmonary endothelial cells. Importantly, endothelium-specific deletion of Sox17 resulted in significant alleviation of various clinical features of asthma, including airway inflammation, immune cell infiltration, cytokine/chemokine production, and airway hyperresponsiveness. Sox17 deletion also resulted in decreased densities of Ly6chigh monocytes and inflammatory dendritic cells in the lungs. In IL-33-stimulated human endothelial cells, Sox17 showed positive correlation with CCL2 and intercellular adhesion molecule 1 levels. Lastly, Sox17 promoted monocyte adhesion to endothelial cells and upregulated the extracellular signal-regulated kinase-signal transducer and activator of transcription 3 pathway. CONCLUSION: Sox17 was regulated by IL-33, and its genetic ablation in endothelial cells resulted in alleviation of asthma-related pathophysiologic features. Sox17 might be a potential target for asthma management.


Subject(s)
Asthma/immunology , Endothelium, Vascular/immunology , HMGB Proteins/immunology , Lung/immunology , SOXF Transcription Factors/immunology , Animals , Asthma/genetics , Asthma/pathology , Chemokines/genetics , Chemokines/immunology , Endothelium, Vascular/pathology , HMGB Proteins/genetics , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-33/genetics , Interleukin-33/immunology , Lung/pathology , Mice , Mice, Mutant Strains , SOXF Transcription Factors/genetics
9.
J Exp Med ; 215(3): 963-983, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29444818

ABSTRACT

High-grade glioma (HGG) is highly angiogenic, but antiangiogenic therapy has transient clinical benefit in only a fraction of patients. Vascular regulators of these heterogeneous responses remain undetermined. We found up-regulation of Sox7 and down-regulation of Sox17 in tumor endothelial cells (tECs) in mouse HGG. Sox7 deletion suppressed VEGFR2 expression, vascular abnormality, hypoxia-driven invasion, regulatory T cell infiltration, and tumor growth. Conversely, Sox17 deletion exacerbated these phenotypes by up-regulating Sox7 in tECs. Anti-VEGFR2 antibody treatment delayed tumor growth by normalizing Sox17-deficient abnormal vessels with high Sox7 levels but promoted it by regressing Sox7-deficient vessels, recapitulating variable therapeutic responses to antiangiogenic therapy in HGG patients. Our findings establish that Sox7 promotes tumor growth via vessel abnormalization, and its level determines the therapeutic outcome of VEGFR2 inhibition in HGG. In 189 HGG patients, Sox7 expression was heterogeneous in tumor vessels, and high Sox7 levels correlated with poor survival, early recurrence, and impaired vascular function, emphasizing the clinical relevance of Sox7 in HGG.


Subject(s)
Blood Vessels/abnormalities , Glioma/metabolism , Glioma/pathology , SOXF Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Calcium-Binding Proteins , Down-Regulation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Deletion , Glioma/blood supply , Glioma/immunology , Humans , Immunity , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Neoplasm Grading , Prognosis , Up-Regulation , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
10.
PLoS One ; 12(11): e0187660, 2017.
Article in English | MEDLINE | ID: mdl-29099870

ABSTRACT

Bone marrow is a vital tissue that produces the majority of erythrocytes, thrombocytes, and immune cells. Bone marrow transplantation (BMT) has been widely performed in patients with blood disorders and cancers. However, the cellular-level behaviors of the transplanted bone marrow cells over wide-areas of the host bone marrow after the BMT are not fully understood yet. In this work, we performed a longitudinal wide-area cellular-level observation of the calvarial bone marrow after the BMT in vivo. Using a H2B-GFP/ß-actin-DsRed double-transgenic mouse model as a donor, a subcellular-level nuclear-cytoplasmic visualization of the transplanted bone marrow cells was achieved, which enabled a direct in vivo dynamic monitoring of the distribution and proliferation of the transplanted bone marrow cells. The same spots in the wide-area of the calvarial bone marrow were repeatedly identified using fluorescently labeled vasculature as a distinct landmark. It revealed various dynamic cellular-level behaviors of the transplanted BM cells in early stage such as cluster formation, migration, and active proliferation in vivo.


Subject(s)
Bone Marrow Transplantation , Bone Marrow/pathology , Cell Differentiation , Cell Nucleus/metabolism , Cytoplasm/metabolism , Animals , Bone Marrow/diagnostic imaging , Cell Lineage , Flow Cytometry , Humans , Longitudinal Studies , Mice , Mice, Inbred C57BL , Microscopy, Confocal
11.
Elife ; 62017 01 31.
Article in English | MEDLINE | ID: mdl-28137360

ABSTRACT

A small molecule called Sm4 can disrupt interactions involving a transcription factor called Sox18, while having little impact on other members of the SoxF family.


Subject(s)
Breast Neoplasms , SOXF Transcription Factors/genetics , Animals , Gene Expression Regulation , Humans , Mice
12.
Sci Rep ; 7: 41840, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165490

ABSTRACT

Isolating actively proliferating cardioblasts is the first crucial step for cardiac regeneration through cell implantation. However, the origin and identity of putative cardioblasts are still unclear. Here, we uncover a novel class of cardiac lineage cells, PDGFRα+Flk1- cardioblasts (PCBs), from mouse and human pluripotent stem cells induced using CsAYTE, a combination of the small molecules Cyclosporin A, the rho-associated coiled-coil kinase inhibitor Y27632, the antioxidant Trolox, and the ALK5 inhibitor EW7197. This novel population of actively proliferating cells is cardiac lineage-committed but in a morphologically and functionally immature state compared to mature cardiomyocytes. Most important, most of CsAYTE-induced PCBs spontaneously differentiated into functional αMHC+ cardiomyocytes (M+CMs) and could be a potential cellular resource for cardiac regeneration.


Subject(s)
Cell Differentiation , Myoblasts/cytology , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Amides/pharmacology , Aniline Compounds/pharmacology , Animals , Antioxidants/pharmacology , Cell Line , Cells, Cultured , Chromans/pharmacology , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Mice , Myoblasts/metabolism , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Pyridines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Triazoles/pharmacology
14.
Cancer Cell ; 30(6): 953-967, 2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27960088

ABSTRACT

A destabilized tumor vasculature leads to limited drug delivery, hypoxia, detrimental tumor microenvironment, and even metastasis. We performed a side-by-side comparison of ABTAA (Ang2-Binding and Tie2-Activating Antibody) and ABA (Ang2-Blocking Antibody) in mice with orthotopically implanted glioma, with subcutaneously implanted Lewis lung carcinoma, and with spontaneous mammary cancer. We found that Tie2 activation induced tumor vascular normalization, leading to enhanced blood perfusion and chemotherapeutic drug delivery, markedly lessened lactate acidosis, and reduced tumor growth and metastasis. Moreover, ABTAA favorably altered the immune cell profile within tumors. Together, our findings establish that simultaneous Tie2 activation and Ang2 inhibition form a powerful therapeutic strategy to elicit a favorable tumor microenvironment and enhanced delivery of a chemotherapeutic agent into tumors.


Subject(s)
Antibodies/administration & dosage , Breast Neoplasms/drug therapy , Carcinoma, Lewis Lung/drug therapy , Glioma/drug therapy , Receptor, TIE-2/metabolism , Ribonuclease, Pancreatic/antagonists & inhibitors , Animals , Antibodies/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Breast Neoplasms/blood supply , Carcinoma, Lewis Lung/blood supply , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Drug Synergism , Female , Glioma/blood supply , Humans , Mice , Neoplasm Transplantation , Protein Binding/drug effects , Temozolomide , Treatment Outcome , Tumor Microenvironment/drug effects
15.
Circ Res ; 119(7): 839-52, 2016 Sep 16.
Article in English | MEDLINE | ID: mdl-27528602

ABSTRACT

RATIONALE: Vascular endothelial growth factor (VEGF) signaling is a key pathway for angiogenesis and requires highly coordinated regulation. Although the Notch pathway-mediated suppression of excessive VEGF activity via negative feedback is well known, the positive feedback control for augmenting VEGF signaling remains poorly understood. Transcription factor Sox17 is indispensable for angiogenesis, but its association with VEGF signaling is largely unknown. The contribution of other Sox members to angiogenesis also remains to be determined. OBJECTIVE: To reveal the genetic interaction of Sox7, another Sox member, with Sox17 in developmental angiogenesis and their functional relationship with VEGF signaling. METHODS AND RESULTS: Sox7 is expressed specifically in endothelial cells and its global and endothelial-specific deletion resulted in embryonic lethality with severely impaired angiogenesis in mice, substantially overlapping with Sox17 in both expression and function. Interestingly, compound heterozygosity for Sox7 and Sox17 phenocopied vascular defects of Sox7 or Sox17 homozygous knockout, indicating that the genetic cooperation of Sox7 and Sox17 is sensitive to their combined gene dosage. VEGF signaling upregulated both Sox7 and Sox17 expression in angiogenesis via mTOR pathway. Furthermore, Sox7 and Sox17 promoted VEGFR2 (VEGF receptor 2) expression in angiogenic vessels, suggesting a positive feedback loop between VEGF signaling and SoxF. CONCLUSIONS: Our findings demonstrate that SoxF transcription factors are indispensable players in developmental angiogenesis by acting as positive feedback regulators of VEGF signaling.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , SOXF Transcription Factors/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Culture Techniques , Female , Humans , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy
16.
Sci Transl Med ; 8(335): 335ra55, 2016 04 20.
Article in English | MEDLINE | ID: mdl-27099174

ABSTRACT

Protection of endothelial integrity has been recognized as a frontline approach to alleviating sepsis progression, yet no effective agent for preserving endothelial integrity is available. Using an unusual anti-angiopoietin 2 (ANG2) antibody, ABTAA (ANG2-binding and TIE2-activating antibody), we show that activation of the endothelial receptor TIE2 protects the vasculature from septic damage and provides survival benefit in three sepsis mouse models. Upon binding to ANG2, ABTAA triggers clustering of ANG2, assembling an ABTAA/ANG2 complex that can subsequently bind and activate TIE2. Compared with a conventional ANG2-blocking antibody, ABTAA was highly effective in augmenting survival from sepsis by strengthening the endothelial glycocalyx, reducing cytokine storms, vascular leakage, and rarefaction, and mitigating organ damage. Together, our data advance the role of TIE2 activation in ameliorating sepsis progression and open a potential therapeutic avenue for sepsis to address the lack of sepsis-specific treatment.


Subject(s)
Antibodies/therapeutic use , Receptor, TIE-2/metabolism , Sepsis/drug therapy , Sepsis/metabolism , Angiopoietin-2/metabolism , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Neutrophil Infiltration/drug effects , Ribonuclease, Pancreatic/metabolism , Vesicular Transport Proteins/metabolism
17.
Circulation ; 131(11): 995-1005, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25596186

ABSTRACT

BACKGROUND: Intracranial aneurysm (IA) is a common vascular disorder that frequently leads to fatal vascular rupture. Although various acquired risk factors associated with IA have been identified, the hereditary basis of IA remains poorly understood. As a result, genetically modified animals accurately modeling IA and related pathogenesis have been lacking, and subsequent drug development has been delayed. METHODS AND RESULTS: The transcription factor Sox17 is robustly expressed in endothelial cells of normal intracerebral arteries. The combination of Sox17 deficiency and angiotensin II infusion in mice induces vascular abnormalities closely resembling the cardinal features of IA such as luminal dilation, wall thinning, tortuosity, and subarachnoid hemorrhages. This combination impairs junctional assembly, cell-matrix adhesion, regeneration capacity, and paracrine secretion in endothelial cells of intracerebral arteries, highlighting key endothelial dysfunctions that lead to IA pathogenesis. Moreover, human IA samples showed reduced Sox17 expression and impaired endothelial integrity, further strengthening the applicability of this animal model to clinical settings. CONCLUSIONS: Our findings demonstrate that Sox17 deficiency in mouse can induce IA under hypertensive conditions, suggesting Sox17 deficiency as a potential genetic factor for IA formation. The Sox17-deficient mouse model provides a novel platform to develop therapeutics for incurable IA.


Subject(s)
Endothelium, Vascular/pathology , HMGB Proteins/deficiency , Intracranial Aneurysm/genetics , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/physiology , Adult , Aged , Angiotensin II/toxicity , Animals , Aorta/pathology , Cells, Cultured , Cerebral Arteries/chemistry , Cerebral Arteries/pathology , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dilatation, Pathologic/genetics , Dilatation, Pathologic/pathology , Disease Models, Animal , Endothelium, Vascular/metabolism , Female , HMGB Proteins/genetics , HMGB Proteins/physiology , Humans , Hypertension/complications , Intracranial Aneurysm/etiology , Intracranial Aneurysm/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/chemistry , Paracrine Communication , RNA Interference , SOXF Transcription Factors/analysis , SOXF Transcription Factors/genetics , Specific Pathogen-Free Organisms , Subarachnoid Hemorrhage/etiology , Transcription, Genetic , Up-Regulation , Veins/chemistry
18.
Circ Res ; 115(2): 215-26, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24755984

ABSTRACT

RATIONALE: The Notch pathway stabilizes sprouting angiogenesis by favoring stalk cells over tip cells at the vascular front. Because tip and stalk cells have different properties in morphology and function, their transcriptional regulation remains to be distinguished. Transcription factor Sox17 is specifically expressed in endothelial cells, but its expression and role at the vascular front remain largely unknown. OBJECTIVE: To specify the role of Sox17 and its relationship with the Notch pathway in sprouting angiogenesis. METHODS AND RESULTS: Endothelial-specific Sox17 deletion reduces sprouting angiogenesis in mouse embryonic and postnatal vascular development, whereas Sox17 overexpression increases it. Sox17 promotes endothelial migration by destabilizing endothelial junctions and rearranging cytoskeletal structure and upregulates expression of several genes preferentially expressed in tip cells. Interestingly, Sox17 expression is suppressed in stalk cells in which Notch signaling is relatively high. Notch activation by overexpressing Notch intracellular domain reduces Sox17 expression both in primary endothelial cells and in retinal angiogenesis, whereas Notch inhibition by delta-like ligand 4 (Dll4) blockade increases it. The Notch pathway regulates Sox17 expression mainly at the post-transcriptional level. Furthermore, endothelial Sox17 ablation rescues vascular network from excessive tip cell formation and hyperbranching under Notch inhibition in developmental and tumor angiogenesis. CONCLUSIONS: Our findings demonstrate that the Notch pathway restricts sprouting angiogenesis by reducing the expression of proangiogenic regulator Sox17.


Subject(s)
Endothelial Cells/metabolism , HMGB Proteins/physiology , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Receptors, Notch/physiology , SOXF Transcription Factors/physiology , Signal Transduction/physiology , Animals , Carcinoma, Lewis Lung/blood supply , Cell Differentiation , Cell Movement , Cytoskeleton/ultrastructure , Embryo, Mammalian/blood supply , Embryonic Stem Cells , Gene Expression Regulation , HMGB Proteins/biosynthesis , HMGB Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Morphogenesis/genetics , Protein Structure, Tertiary , RNA, Small Interfering/pharmacology , Receptor, Notch1/genetics , Receptor, Notch1/physiology , Recombinant Fusion Proteins , Retinal Vessels/growth & development , SOXF Transcription Factors/biosynthesis , SOXF Transcription Factors/genetics , Specific Pathogen-Free Organisms , Transcription, Genetic
19.
J Am Heart Assoc ; 3(2): e000693, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24627421

ABSTRACT

BACKGROUND: Cardiomyocytes that differentiate from pluripotent stem cells (PSCs) provide a crucial cellular resource for cardiac regeneration. The mechanisms of mitochondrial metabolic and redox regulation for efficient cardiomyocyte differentiation are, however, still poorly understood. Here, we show that inhibition of the mitochondrial permeability transition pore (mPTP) by Cyclosporin A (CsA) promotes cardiomyocyte differentiation from PSCs. METHODS AND RESULTS: We induced cardiomyocyte differentiation from mouse and human PSCs and examined the effect of CsA on the differentiation process. The cardiomyogenic effect of CsA mainly resulted from mPTP inhibition rather than from calcineurin inhibition. The mPTP inhibitor NIM811, which does not have an inhibitory effect on calcineurin, promoted cardiomyocyte differentiation as much as CsA did, but calcineurin inhibitor FK506 only slightly increased cardiomyocyte differentiation. CsA-treated cells showed an increase in mitochondrial calcium, mitochondrial membrane potential, oxygen consumption rate, ATP level, and expression of genes related to mitochondrial function. Furthermore, inhibition of mitochondrial oxidative metabolism reduced the cardiomyogenic effect of CsA while antioxidant treatment augmented the cardiomyogenic effect of CsA. CONCLUSIONS: Our data show that mPTP inhibition by CsA alters mitochondrial oxidative metabolism and redox signaling, which leads to differentiation of functional cardiomyocytes from PSCs.


Subject(s)
Cell Differentiation/drug effects , Cyclosporine/pharmacology , Energy Metabolism/drug effects , Mitochondria, Heart/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Signal Transduction/drug effects , Animals , Cell Line , Cell Proliferation/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Feeder Cells , Humans , Mice , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Pluripotent Stem Cells/metabolism , Time Factors
20.
Cancer Cell ; 25(1): 102-17, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24434213

ABSTRACT

Current antiangiogenic therapy is limited by its cytostatic nature and systemic side effects. To address these limitations, we have unveiled the role of RhoJ, an endothelial-enriched Rho GTPase, during tumor progression. RhoJ blockade provides a double assault on tumor vessels by both inhibiting tumor angiogenesis and disrupting the preformed tumor vessels through the activation of the RhoA-ROCK (Rho kinase) signaling pathway in tumor endothelial cells, consequently resulting in a functional failure of tumor vasculatures. Moreover, enhanced anticancer effects were observed when RhoJ blockade was employed in concert with a cytotoxic chemotherapeutic agent, angiogenesis-inhibiting agent, or vascular-disrupting agent. These results identify RhoJ blockade as a selective and effective therapeutic strategy for targeting tumor vasculature with minimal side effects.


Subject(s)
GTP Phosphohydrolases/metabolism , Neoplasms, Experimental/enzymology , Neovascularization, Pathologic/enzymology , rho GTP-Binding Proteins/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , RNA, Small Interfering , Signal Transduction/drug effects , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...