Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Sci Rep ; 14(1): 13603, 2024 06 13.
Article in English | MEDLINE | ID: mdl-38866944

ABSTRACT

Notch signaling guides vascular development and function by regulating diverse endothelial cell behaviors, including migration, proliferation, vascular density, endothelial junctions, and polarization in response to flow. Notch proteins form transcriptional activation complexes that regulate endothelial gene expression, but few of the downstream effectors that enable these phenotypic changes have been characterized in endothelial cells, limiting our understanding of vascular Notch activities. Using an unbiased screen of translated mRNA rapidly regulated by Notch signaling, we identified novel in vivo targets of Notch signaling in neonatal mouse brain endothelium, including UNC5B, a member of the netrin family of angiogenic-regulatory receptors. Endothelial Notch signaling rapidly upregulates UNC5B in multiple endothelial cell types. Loss or gain of UNC5B recapitulated specific Notch-regulated phenotypes. UNC5B expression inhibited endothelial migration and proliferation and was required for stabilization of endothelial junctions in response to shear stress. Loss of UNC5B partially or wholly blocked the ability of Notch activation to regulate these endothelial cell behaviors. In the developing mouse retina, endothelial-specific loss of UNC5B led to excessive vascularization, including increased vascular outgrowth, density, and branchpoint count. These data indicate that Notch signaling upregulates UNC5B as an effector protein to control specific endothelial cell behaviors and inhibit angiogenic growth.


Subject(s)
Cell Movement , Cell Proliferation , Endothelial Cells , Netrin Receptors , Receptors, Notch , Retina , Signal Transduction , Animals , Netrin Receptors/metabolism , Receptors, Notch/metabolism , Mice , Endothelial Cells/metabolism , Retina/metabolism , Humans , Retinal Vessels/metabolism , Neovascularization, Physiologic
2.
Angiogenesis ; 27(2): 273-283, 2024 May.
Article in English | MEDLINE | ID: mdl-37796367

ABSTRACT

Notch and its ligands play a critical role in rheumatoid arthritis (RA) pathogenesis. Hence, studies were conducted to delineate the functional significance of the Notch pathway in RA synovial tissue (ST) cells and the influence of RA therapies on their expression. Morphological studies reveal that JAG1, DLL4, and Notch1 are highly enriched in RA ST lining and sublining CD68+CD14+ MΦs. JAG1 and DLL4 transcription is jointly upregulated in RA MΦs reprogrammed by TLR4/5 ligation and TNF, whereas Syntenin-1 exposure expands JAG1, DLL4, and Notch1 expression levels in these cells. Single-cell RNA-seq data exhibit that JAG1 and Notch3 are overexpressed on all fibroblast-like synoviocyte (FLS) subpopulations, in parallel, JAG2, DLL1, and Notch1 expression levels are modest on RA FLS and are predominately potentiated by TLR4 ligation. Intriguingly, JAG1, DLL1/4, and Notch1/3 are presented on RA endothelial cells, and their expression is mutually reconfigured by TLR4/5 ligation in the endothelium. Synovial JAG1/JAG2/DLL1 or Notch1/3 transcriptomes were unchanged in patients who received disease-modifying anti-rheumatic drugs (DMARDs) or IL-6R Ab therapy regardless of disease activity score. Uniquely, RA MΦs and endothelial cells rewired by IL-6 displayed DLL4 transcriptional upregulation, and IL-6R antibody treatment disrupted RA ST DLL4 transcription in good responders compared to non-responders or moderate responders. Nevertheless, the JAG1/JAG2/DLL1/DLL4 transcriptome was diminished in anti-TNF good responders with myeloid pathotype and was unaltered in the fibroid pathotype except for DLL4. Taken together, our findings suggest that RA myeloid Notch ligands can serve as markers for anti-TNF responsiveness and trans-activate Notch receptors expressed on RA FLS and/or endothelial cells.


Subject(s)
Arthritis, Rheumatoid , Tumor Necrosis Factor Inhibitors , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Calcium-Binding Proteins/metabolism , Membrane Proteins/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Endothelial Cells/metabolism , Toll-Like Receptor 4/metabolism , Receptors, Notch/metabolism , Biomarkers , Arthritis, Rheumatoid/drug therapy , Ligands , Receptor, Notch1/metabolism
4.
Microcirculation ; 30(7): e12826, 2023 10.
Article in English | MEDLINE | ID: mdl-37605603

ABSTRACT

OBJECTIVE: Three-dimensional (3D) microscopy and image data analysis are necessary for studying the morphology of cardiac lymphatic vessels (LyVs) and their association with other cell types. We aimed to develop a methodology for 3D multiplexed lightsheet microscopy and highly sensitive and quantitative image analysis to identify pathological remodeling in the 3D morphology of LyVs in young adult mouse hearts with familial hypertrophic cardiomyopathy (HCM). METHODS: We developed a 3D lightsheet microscopy workflow providing a quick turn-around (as few as 5-6 days), multiplex fluorescence detection, and preservation of LyV structure and epitope markers. Hearts from non-transgenic and transgenic (TG) HCM mice were arrested in diastole, retrograde perfused, immunolabeled, optically cleared, and imaged. We built an image-processing pipeline to quantify LyV morphological parameters at the chamber and branch levels. RESULTS: Chamber-specific pathological alterations of LyVs were identified, and significant changes were seen in the right atrium (RA). TG hearts had a higher volume percent of ER-TR7+ fibroblasts and reticular fibers. In the RA, we found associations between ER-TR7+ volume percent and both LyV segment density and median diameter. CONCLUSIONS: This workflow and study enabled multi-scale analysis of pathological changes in cardiac LyVs of young adult mice, inviting ideas for research on LyVs in cardiac disease.


Subject(s)
Heart , Lymphatic Vessels , Mice , Animals , Mice, Transgenic , Coronary Vessels , Image Processing, Computer-Assisted , Imaging, Three-Dimensional
5.
Arterioscler Thromb Vasc Biol ; 43(8): 1441-1454, 2023 08.
Article in English | MEDLINE | ID: mdl-37317855

ABSTRACT

BACKGROUND: Endothelial CLICs (chloride intracellular channel proteins) CLIC1 and CLIC4 are required for the GPCRs (G-protein-coupled receptors) S1PR1 (sphingosine-1-phosphate receptor 1) and S1PR3 to activate the small GTPases Rac1 (Ras-related C3 botulinum toxin substrate 1) and RhoA (Ras homolog family member A). To determine whether CLIC1 and CLIC4 function in additional endothelial GPCR pathways, we evaluated CLIC function in thrombin signaling via the thrombin-regulated PAR1 (protease-activated receptor 1) and downstream effector RhoA. METHODS: We assessed the ability of CLIC1 and CLIC4 to relocalize to cell membranes in response to thrombin in human umbilical vein endothelial cells (HUVEC). We examined CLIC1 and CLIC4 function in HUVEC by knocking down expression of each CLIC protein and compared thrombin-mediated RhoA or Rac1 activation, ERM (ezrin/radixin/moesin) phosphorylation, and endothelial barrier modulation in control and CLIC knockdown HUVEC. We generated a conditional murine allele of Clic4 and examined PAR1-mediated lung microvascular permeability and retinal angiogenesis in mice with endothelial-specific loss of Clic4. RESULTS: Thrombin promoted relocalization of CLIC4, but not CLIC1, to HUVEC membranes. Knockdown of CLIC4 in HUVEC reduced thrombin-mediated RhoA activation, ERM phosphorylation, and endothelial barrier disruption. Knockdown of CLIC1 did not reduce thrombin-mediated RhoA activity but prolonged the RhoA and endothelial barrier response to thrombin. Endothelial-specific deletion of Clic4 in mice reduced lung edema and microvascular permeability induced by PAR1 activating peptide. CONCLUSIONS: CLIC4 is a critical effector of endothelial PAR1 signaling and is required to regulate RhoA-mediated endothelial barrier disruption in cultured endothelial cells and murine lung endothelium. CLIC1 was not critical for thrombin-mediated barrier disruption but contributed to the barrier recovery phase after thrombin treatment.


Subject(s)
Receptor, PAR-1 , rhoA GTP-Binding Protein , Humans , Mice , Animals , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , rhoA GTP-Binding Protein/metabolism , Thrombin/pharmacology , Thrombin/metabolism , Endothelium/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Cells, Cultured , Chloride Channels/genetics , Chloride Channels/metabolism , Mitochondrial Proteins/metabolism
6.
bioRxiv ; 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36865302

ABSTRACT

Glioma cells hijack developmental transcriptional programs to control cell state. During neural development, lineage trajectories rely on specialized metabolic pathways. However, the link between tumor cell state and metabolic programs is poorly understood in glioma. Here we uncover a glioma cell state-specific metabolic liability that can be leveraged therapeutically. To model cell state diversity, we generated genetically engineered murine gliomas, induced by deletion of p53 alone (p53) or with constitutively active Notch signaling (N1IC), a pathway critical in controlling cellular fate. N1IC tumors harbored quiescent astrocyte-like transformed cell states while p53 tumors were predominantly comprised of proliferating progenitor-like cell states. N1IC cells exhibit distinct metabolic alterations, with mitochondrial uncoupling and increased ROS production rendering them more sensitive to inhibition of the lipid hydroperoxidase GPX4 and induction of ferroptosis. Importantly, treating patient-derived organotypic slices with a GPX4 inhibitor induced selective depletion of quiescent astrocyte-like glioma cell populations with similar metabolic profiles.

7.
bioRxiv ; 2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36778334

ABSTRACT

Objective: 3D microscopy and image data analysis are necessary for studying the morphology of cardiac lymphatic vessels (LyVs) and association with other cell types. We aimed to develop a methodology for 3D multiplexed lightsheet microscopy and highly sensitive and quantitative image analysis to identify pathological remodeling in the 3D morphology of LyVs in young adult mouse hearts with familial hypertrophic cardiomyopathy (HCM). Methods: We developed a 3D lightsheet microscopy workflow providing a quick turn-around (as few as 5-6 days), multiplex fluorescence detection, and preservation of LyV structure and epitope markers. Hearts from non-transgenic (NTG) and transgenic (TG) HCM mice were arrested in diastole, retrograde perfused, immunolabeled, optically cleared, and imaged. We built an image processing pipeline to quantify LyV morphological parameters at the chamber and branch levels. Results: Chamber-specific pathological alterations of LyVs were identified, but most significantly in the right atrium (RA). TG hearts had a higher volume fraction of ER-TR7 + fibroblasts and reticular fibers. In the RA, we found associations between ER-TR7 + volume fraction and both LyV segment density and median diameter. Conclusions: This workflow and study enabled multi-scale analysis of pathological changes in cardiac LyVs of young adult mice, inviting ideas for research on LyVs in cardiac disease.

8.
Angiogenesis ; 26(2): 249-263, 2023 05.
Article in English | MEDLINE | ID: mdl-36376768

ABSTRACT

The Notch signaling pathway is an important therapeutic target for the treatment of inflammatory diseases and cancer. We previously created ligand-specific inhibitors of Notch signaling comprised of Fc fusions to specific EGF-like repeats of the Notch1 extracellular domain, called Notch decoys, which bound ligands, blocked Notch signaling, and showed anti-tumor activity with low toxicity. However, the study of their function depended on virally mediated expression, which precluded dosage control and limited clinical applicability. We have refined the decoy design to create peptibody-based Notch inhibitors comprising the core binding domains, EGF-like repeats 10-14, of either Notch1 or Notch4. These Notch peptibodies showed high secretion properties and production yields that were improved by nearly 100-fold compared to previous Notch decoys. Using surface plasmon resonance spectroscopy coupled with co-immunoprecipitation assays, we observed that Notch1 and Notch4 peptibodies demonstrate strong but distinct binding properties to Notch ligands DLL4 and JAG1. Both Notch1 and Notch4 peptibodies interfere with Notch signaling in endothelial cells and reduce expression of canonical Notch targets after treatment. While prior DLL4 inhibitors cause hyper-sprouting, the Notch1 peptibody reduced angiogenesis in a 3-dimensional in vitro sprouting assay. Administration of Notch1 peptibodies to neonate mice resulted in reduced radial outgrowth of retinal vasculature, confirming anti-angiogenic properties. We conclude that purified Notch peptibodies comprising EGF-like repeats 10-14 bind to both DLL4 and JAG1 ligands and exhibit anti-angiogenic properties. Based on their secretion profile, unique Notch inhibitory activities, and anti-angiogenic properties, Notch peptibodies present new opportunities for therapeutic Notch inhibition.


Subject(s)
Angiogenesis Inhibitors , Endothelial Cells , Receptor, Notch1 , Receptor, Notch4 , Animals , Mice , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epidermal Growth Factor/metabolism , Immunoprecipitation , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Ligands , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch4/genetics , Receptor, Notch4/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Vessels/drug effects , Surface Plasmon Resonance
9.
Article in English | MEDLINE | ID: mdl-35288401

ABSTRACT

As vascular networks form, endothelial cells (ECs) undergo cell fate decisions that determine whether they become tip or stalk cells of the developing vascular plexus or mature into arterial, venous, or lymphatic endothelium. EC fate decisions are coordinated with neighboring cells to initiate sprouting, maintain endothelial barrier, or ensure appropriate specialization of vessels. We describe mechanisms that control EC fate at specific steps in these processes, with an emphasis on the role of the Notch signaling pathway. Specific EC fate determination steps that are highlighted are tip/stalk selection during sprouting angiogenesis, venous-arterial specification, arteriogenesis, lymphatic vessel specification, and lymphatic valve formation.


Subject(s)
Endothelial Cells , Receptors, Notch , Humans , Signal Transduction , Cell Differentiation , Morphogenesis , Neovascularization, Physiologic
10.
Sci Rep ; 12(1): 1655, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102202

ABSTRACT

To control sprouting angiogenesis, endothelial Notch signaling suppresses tip cell formation, migration, and proliferation while promoting barrier formation. Each of these responses may be regulated by distinct Notch-regulated effectors. Notch activity is highly dynamic in sprouting endothelial cells, while constitutive Notch signaling drives homeostatic endothelial polarization, indicating the need for both rapid and constitutive Notch targets. In contrast to previous screens that focus on genes regulated by constitutively active Notch, we characterized the dynamic response to Notch. We examined transcriptional changes from 1.5 to 6 h after Notch signal activation via ligand-specific or EGTA induction in cultured primary human endothelial cells and neonatal mouse brain. In each combination of endothelial type and Notch manipulation, transcriptomic analysis identified distinct but overlapping sets of rapidly regulated genes and revealed many novel Notch target genes. Among the novel Notch-regulated signaling pathways identified were effectors in GPCR signaling, notably, the constitutively active GTPase RND1. In endothelial cells, RND1 was shown to be a novel direct Notch transcriptional target and required for Notch control of sprouting angiogenesis, endothelial migration, and Ras activity. We conclude that RND1 is directly regulated by endothelial Notch signaling in a rapid fashion in order to suppress endothelial migration.


Subject(s)
Brain/blood supply , Cell Movement , Endothelial Cells/enzymology , Neovascularization, Physiologic , Receptors, Notch/metabolism , rho GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Proliferation , Gene Expression Regulation, Enzymologic , HEK293 Cells , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Notch/genetics , Signal Transduction , Time Factors , Transcription, Genetic , ras Proteins/genetics , ras Proteins/metabolism , rho GTP-Binding Proteins/genetics
11.
Cancer Res Commun ; 2(12): 1601-1616, 2022 12.
Article in English | MEDLINE | ID: mdl-36970723

ABSTRACT

Expression of the Notch family of receptors is often upregulated in pancreatic ductal adenocarcinoma (PDAC). In this study, we focused on Notch4, which had not been investigated in PDAC. We generated KC (LSL-KrasG12D;p48-Cre), N4 - / - KC (Notch4- / -;LSL-KrasG12D;p48-Cre), PKC (p16fl/fl;LSL-KrasG12D;p48-Cre), and N4 - / - PKC (Notch4-/ -; p16fl/f l;LSL-KrasG12D;p48-Cre) genetically engineered mouse models (GEMM). We performed caerulein treatment in both KC and N4 - / - KC mice, and the development of acinar-to-ductal metaplasia (ADM) and pancreatic intraepithelial neoplasia (PanIN) lesions were significantly diminished in the N4 - / - KC than in the KC GEMM (P = 0.01). This in vivo result was validated by in vitro ADM induction of the explant cultures of pancreatic acinar cells from the N4 - / - KC and KC mice (P < 0.001), confirming that Notch4 is an important contributor to early pancreatic tumorigenesis. To evaluate the role of Notch4 in the later stage of pancreatic tumorigenesis, we compared the PKC and N4 - / - PKC mice. The N4 - / - PKC mice had better overall survival (P = 0.012) and significantly reduced tumor burden (PanIN: P = 0.018 at 2 months, PDAC: P = 0.039 at 5 months) compared with the PKC GEMM. RNA-sequencing analysis of pancreatic tumor cell lines derived from the PKC and N4 - / - PKC GEMMs revealed that 408 genes were differentially expressed (FDR < 0.05) and Pcsk5 is a potential downstream effector of the Notch4 signaling pathway (P < 0.001). Low expression of Pcsk5 positively correlates with good survival in patients with PDAC (P = 0.028). We have identified a novel role for Notch4 signaling with tumor-promoting function in pancreatic tumorigenesis. Our study also uncovered a novel association between Pcsk5 and Notch4 signaling in PDAC. Significance: We demonstrated that global inactivation of Notch4 significantly improved the survival of an aggressive mouse model for PDAC and provided preclinical evidence that Notch4 and Pcsk5 are novel targets for PDAC therapies.


Subject(s)
Carcinoma in Situ , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Mice , Animals , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma in Situ/genetics , Pancreatic Neoplasms
12.
Angiogenesis ; 25(2): 205-224, 2022 05.
Article in English | MEDLINE | ID: mdl-34665379

ABSTRACT

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


Subject(s)
Lymphangiogenesis , Lymphatic Vessels , Animals , Endothelial Cells/metabolism , Lymphangiogenesis/physiology , Lymphatic System/metabolism , Lymphatic Vessels/metabolism , Mice , Receptors, Notch/metabolism , Signal Transduction
13.
Dev Cell ; 56(19): 2752-2764.e6, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34610330

ABSTRACT

Neuroblastoma (NB), the most common cancer in the first year of life, presents almost exclusively in the trunk. To understand why an early-onset cancer would have such a specific localization, we xenotransplanted human NB cells into discrete neural crest (NC) streams in zebrafish embryos. Here, we demonstrate that human NB cells remain in an undifferentiated, tumorigenic state when comigrating posteriorly with NC cells but, upon comigration into the head, differentiate into neurons and exhibit decreased survival. Furthermore, we demonstrate that this in vivo differentiation requires retinoic acid and brain-derived neurotrophic factor signaling from the microenvironment, as well as cell-autonomous intersectin-1-dependent phosphoinositide 3-kinase-mediated signaling, likely via Akt kinase activation. Our findings suggest a microenvironment-driven explanation for NB's trunk-biased localization and highlight the potential for induced differentiation to promote NB resolution in vivo.


Subject(s)
Cell Differentiation/physiology , Neuroblastoma/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cell Line, Tumor , Cell Movement/physiology , Female , Humans , Male , Mice , Neural Crest/metabolism , Neurons/cytology , Neurons/physiology , Signal Transduction , Transplantation, Heterologous/methods , Tretinoin/metabolism , Tretinoin/pharmacology , Tumor Microenvironment , Zebrafish/metabolism
15.
Science ; 372(6547): 1205-1209, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34112692

ABSTRACT

Quiescent neural stem cells (NSCs) in the adult mouse ventricular-subventricular zone (V-SVZ) undergo activation to generate neurons and some glia. Here we show that platelet-derived growth factor receptor beta (PDGFRß) is expressed by adult V-SVZ NSCs that generate olfactory bulb interneurons and glia. Selective deletion of PDGFRß in adult V-SVZ NSCs leads to their release from quiescence, uncovering gliogenic domains for different glial cell types. These domains are also recruited upon injury. We identify an intraventricular oligodendrocyte progenitor derived from NSCs inside the brain ventricles that contacts supraependymal axons. Together, our findings reveal that the adult V-SVZ contains spatial domains for gliogenesis, in addition to those for neurogenesis. These gliogenic NSC domains tend to be quiescent under homeostasis and may contribute to brain plasticity.


Subject(s)
Adult Stem Cells/physiology , Cerebral Ventricles/physiology , Lateral Ventricles/physiology , Neural Stem Cells/physiology , Neuroglia/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Animals , Astrocytes/cytology , Astrocytes/physiology , Axons/physiology , Cell Differentiation , Cell Division , Cerebral Ventricles/cytology , Ependyma/cytology , Ependyma/physiology , Female , Gene Expression Profiling , Homeostasis , Lateral Ventricles/cytology , Male , Mice , Neurogenesis , Olfactory Bulb/cytology , Olfactory Bulb/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics
16.
Angiogenesis ; 24(4): 789-805, 2021 11.
Article in English | MEDLINE | ID: mdl-33956260

ABSTRACT

Collagen type IV (Col IV) is a basement membrane protein associated with early blood vessel morphogenesis and is essential for blood vessel stability. Defects in vascular Col IV deposition are the basis of heritable disorders, such as small vessel disease, marked by cerebral hemorrhage and drastically shorten lifespan. To date, little is known about how endothelial cells regulate the intracellular transport and selective secretion of Col IV in response to angiogenic cues, leaving a void in our understanding of this critical process. Our aim was to identify trafficking pathways that regulate Col IV deposition during angiogenic blood vessel development. We have identified the GTPase Rab10 as a major regulator of Col IV vesicular trafficking during vascular development using both in vitro imaging and biochemistry as well as in vivo models. Knockdown of Rab10 reduced de novo Col IV secretion in vivo and in vitro. Mechanistically, we determined that Rab10 is an indirect mediator of Col IV secretion, partnering with atypical Rab25 to deliver the enzyme lysyl hydroxylase 3 (LH3) to Col IV-containing vesicles staged for secretion. Loss of Rab10 or Rab25 results in depletion of LH3 from Col IV-containing vesicles and rapid lysosomal degradation of Col IV. Furthermore, we demonstrate that Rab10 is Notch responsive, indicating a novel connection between permissive Notch-based vessel maturation programs and vesicle trafficking. Our results illustrate both a new trafficking-based component in the regulated secretion of Col IV and how this vesicle trafficking program interfaces with Notch signaling to fine-tune basement membrane secretion during blood vessel development.


Subject(s)
Collagen Type IV , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase , Basement Membrane , Collagen Type IV/genetics , Endothelial Cells , Morphogenesis
17.
Sci Signal ; 14(679)2021 04 20.
Article in English | MEDLINE | ID: mdl-33879602

ABSTRACT

Chloride intracellular channels 1 (CLIC1) and 4 (CLIC4) are expressed in endothelial cells and regulate angiogenic behaviors in vitro, and the expression of Clic4 is important for vascular development and function in mice. Here, we found that CLIC1 and CLIC4 in endothelial cells regulate critical G protein-coupled receptor (GPCR) pathways associated with vascular development and disease. In cultured endothelial cells, we found that CLIC1 and CLIC4 transiently translocated to the plasma membrane in response to sphingosine 1-phosphate (S1P). Both CLIC1 and CLIC4 were essential for mediating S1P-induced activation of the small guanosine triphosphatase (GTPase) Rac1 downstream of S1P receptor 1 (S1PR1). In contrast, only CLIC1 was essential for S1P-induced activation of the small GTPase RhoA downstream of S1PR2 and S1PR3. Neither were required for other S1P-S1PR signaling outputs. Rescue experiments revealed that CLIC1 and CLIC4 were not functionally interchangeable, suggesting distinct and specific functions for CLICs in transducing GPCR signaling. These CLIC-mediated mechanisms were critical for S1P-induced stimulation of the barrier function in endothelial cell monolayers. Our results define CLICs as previously unknown players in the pathways linking GPCRs to small GTPases and vascular endothelial function.


Subject(s)
Chloride Channels/metabolism , Mitochondrial Proteins/metabolism , Neuropeptides , Sphingosine-1-Phosphate Receptors , rac1 GTP-Binding Protein , rhoA GTP-Binding Protein , Animals , Cell Line , Cells, Cultured , Endothelial Cells , Lysophospholipids , Mice , Neuropeptides/metabolism , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine , Sphingosine-1-Phosphate Receptors/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
19.
Reproduction ; 161(6): 681-696, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33784241

ABSTRACT

In the first trimester of human pregnancy, low oxygen tension or hypoxia is essential for proper placentation and placenta function. Low oxygen levels and activation of signaling pathways have been implicated as critical mediators in the promotion of trophoblast differentiation, migration, and invasion with inappropriate changes in oxygen tension and aberrant Notch signaling both individually reported as causative to abnormal placentation. Despite crosstalk between hypoxia and Notch signaling in multiple cell types, the relationship between hypoxia and Notch in first trimester trophoblast function is not understood. To determine how a low oxygen environment impacts Notch signaling and cellular motility, we utilized the human first trimester trophoblast cell line, HTR-8/SVneo. Gene set enrichment and ontology analyses identified pathways involved in angiogenesis, Notch and cellular migration as upregulated in HTR-8/SVneo cells exposed to hypoxic conditions. DAPT, a γ-secretase inhibitor that inhibits Notch activation, was used to interrogate the crosstalk between Notch and hypoxia pathways in HTR-8/SVneo cells. We found that hypoxia requires Notch activation to mediate HTR-8/SVneo cell migration, but not invasion. To determine if our in vitro findings were associated with preeclampsia, we analyzed the second trimester chorionic villous sampling (CVS) samples and third trimester placentas. We found a significant decrease in expression of migration and invasion genes in CVS from preeclamptic pregnancies and significantly lower levels of JAG1 in placentas from pregnancies with early-onset preeclampsia with severe features. Our data support a role for Notch in mediating hypoxia-induced trophoblast migration, which may contribute to preeclampsia development.


Subject(s)
Cell Movement , Hypoxia/physiopathology , Jagged-1 Protein/metabolism , Placenta/pathology , Pre-Eclampsia/pathology , Receptors, Notch/metabolism , Trophoblasts/pathology , Adult , Female , Humans , Jagged-1 Protein/genetics , Placenta/metabolism , Pre-Eclampsia/etiology , Pre-Eclampsia/metabolism , Pregnancy , Pregnancy Trimester, Second , Pregnancy Trimester, Third , Receptors, Notch/genetics , Signal Transduction , Trophoblasts/metabolism
20.
J Cardiovasc Pharmacol ; 77(3): 317-322, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33298734

ABSTRACT

ABSTRACT: A dominant mechanism of sudden cardiac death in the young is the progression of maladaptive responses to genes encoding proteins linked to hypertrophic cardiomyopathy. Most are mutant sarcomere proteins that trigger the progression by imposing a biophysical defect on the dynamics and levels of myofilament tension generation. We discuss approaches for personalized treatments that are indicated by recent advanced understanding of the progression.


Subject(s)
Cardiomyopathy, Hypertrophic/therapy , Death, Sudden, Cardiac/prevention & control , Precision Medicine , COVID-19/complications , Cardiomyopathy, Hypertrophic/complications , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Clinical Decision-Making , Death, Sudden, Cardiac/etiology , Genetic Predisposition to Disease , Humans , Mutation , Phenotype , Prognosis , Risk Assessment , Risk Factors , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL
...