Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
J Med Chem ; 66(23): 16342-16363, 2023 12 14.
Article in English | MEDLINE | ID: mdl-38031930

ABSTRACT

Janus kinase 1 (JAK1) plays a pivotal role in regulating inflammation and fibrosis via the JAK/STAT signaling pathway, making it a promising target for associated diseases. In this study, we explored the modification of an N-methyl 1H-pyrrolo[2,3-b]pyridine-5-carboxylate core, leading to the identification of 4-(((2S,4S)-1-(4-trifluoromethyl)-2-methylpiperidin-4-yl)amino)-N-methyl-1H-pyrrolo[2,3-b]pyridine-5-carboxamide (36b) as a highly potent and selective JAK1 inhibitor. Compound 36b exhibited an impressive IC50 value of 0.044 nM for JAK1 and demonstrated remarkable selectivity of 382-fold, 210-fold, and 1325-fold specificity over JAK2, JAK3, and TYK2, respectively. The kinase panel assays further confirmed its specificity, and cell-based experiments established its efficacy in inhibiting JAK1-STAT phosphorylation in human L-132 or SK-MES-1 cells. Pharmacokinetic studies revealed that compound 36b boasts an oral bioavailability exceeding 36%. In a bleomycin-induced fibrosis mouse model, compound 36b significantly reduced STAT3 phosphorylation, resulting in improvement in body weight and reduced collagen deposition, all achieved without significant side effects.


Subject(s)
Janus Kinase Inhibitors , Pulmonary Fibrosis , Mice , Animals , Humans , Structure-Activity Relationship , Protein Kinase Inhibitors/pharmacology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/drug therapy , Janus Kinase Inhibitors/pharmacology , Janus Kinase 1 , Pyridines
2.
Circ Res ; 133(6): 463-480, 2023 09.
Article in English | MEDLINE | ID: mdl-37555328

ABSTRACT

BACKGROUND: Cardiac valve disease is observed in 2.5% of the general population and 10% of the elderly people. Effective pharmacological treatments are currently not available, and patients with severe cardiac valve disease require surgery. PROX1 (prospero-related homeobox transcription factor 1) and FOXC2 (Forkhead box C2 transcription factor) are transcription factors that are required for the development of lymphatic and venous valves. We found that PROX1 and FOXC2 are expressed in a subset of valvular endothelial cells (VECs) that are located on the downstream (fibrosa) side of cardiac valves. Whether PROX1 and FOXC2 regulate cardiac valve development and disease is not known. METHODS: We used histology, electron microscopy, and echocardiography to investigate the structure and functioning of heart valves from Prox1ΔVEC mice in which Prox1 was conditionally deleted from VECs. Isolated valve endothelial cells and valve interstitial cells were used to identify the molecular mechanisms in vitro, which were tested in vivo by RNAScope, additional mouse models, and pharmacological approaches. The significance of our findings was tested by evaluation of human samples of mitral valve prolapse and aortic valve insufficiency. RESULTS: Histological analysis revealed that the aortic and mitral valves of Prox1ΔVEC mice become progressively thick and myxomatous. Echocardiography revealed that the aortic valves of Prox1ΔVEC mice are stenotic. FOXC2 was downregulated and PDGF-B (platelet-derived growth factor-B) was upregulated in the VECs of Prox1ΔVEC mice. Conditional knockdown of FOXC2 and conditional overexpression of PDGF-B in VECs recapitulated the phenotype of Prox1ΔVEC mice. PDGF-B was also increased in mice lacking FOXC2 and in human mitral valve prolapse and insufficient aortic valve samples. Pharmacological inhibition of PDGF-B signaling with imatinib partially ameliorated the valve defects of Prox1ΔVEC mice. CONCLUSIONS: PROX1 antagonizes PDGF-B signaling partially via FOXC2 to maintain the extracellular matrix composition and prevent myxomatous degeneration of cardiac valves.


Subject(s)
Heart Valve Diseases , Mitral Valve Prolapse , Animals , Humans , Mice , Endothelial Cells/metabolism , Heart Valve Diseases/genetics , Heart Valve Diseases/prevention & control , Heart Valve Diseases/metabolism , Mitral Valve/metabolism , Mitral Valve Prolapse/metabolism , Transcription Factors/metabolism , Proto-Oncogene Proteins c-sis/metabolism
3.
Front Pharmacol ; 14: 1163970, 2023.
Article in English | MEDLINE | ID: mdl-37274097

ABSTRACT

Anoctamin 1 (ANO1), a drug target for various cancers, including prostate and oral cancers, is an intracellular calcium-activated chloride ion channel that plays various physiopathological roles, especially in the induction of cancer growth and metastasis. In this study, we tested a novel compound isolated from Schisandra sphenanthera, known as schisandrathera D, for its inhibitory effect on ANO1. Schisandrathera D dose-dependently suppressed the ANO1 activation-mediated decrease in fluorescence of yellow fluorescent protein; however, it did not affect the adenosine triphosphate-induced increase in the intracellular calcium concentration or forskolin-induced cystic fibrosis transmembrane conductance regulator activity. Specifically, schisandrathera D gradually decreased the levels of ANO1 protein and significantly reduced the cell viability in ANO1-expressing cells when compared to those in ANO1-knockout cells. These effects could be attributed to the fact that schisandrathera D displayed better binding capacity to ANO1 protein than the previously known ANO1 inhibitor, Ani9. Finally, schisandrathera D increased the levels of caspase-3 and cleaved poly (ADP-ribose) polymerase 1, thereby indicating that its anticancer effect is mediated through apoptosis. Thus, this study highlights that schisandrathera D, which reduces ANO1 protein levels, has apoptosis-mediated anticancer effects in prostate and oral cancers, and thus, can be further developed into an anticancer agent.

4.
Microbiol Immunol ; 67(3): 160-165, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36504397

ABSTRACT

To investigate viruses in measles-negative cases, 221 measles-suspected samples collected in Gyeonggi Province, South Korea were tested using a real-time PCR assay. Rubella virus was not detected. However, 11 cases of parvovirus B19 (5.0%), 47 cases of human herpesvirus 6 (21.3%), 25 cases of human herpesvirus 7 (11.3%), and one case of co-infection with parvovirus B19 and human herpesvirus 7 were confirmed, as were eight cases of co-infection with human herpesvirus 6 and human herpesvirus 7. This study showed that parvovirus B19, human herpesvirus 6, and human herpesvirus 7 should be considered by physicians for the diagnosis of measles-suspected patients.


Subject(s)
Coinfection , Herpesvirus 6, Human , Measles , Parvovirus B19, Human , Humans , Antibodies, Viral , Immunoglobulin M , Measles/diagnosis , Measles/epidemiology , Republic of Korea/epidemiology
5.
BMB Rep ; 54(6): 285-294, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33691907

ABSTRACT

The lymphatic vasculature plays important role in regulating fluid homeostasis, intestinal lipid absorption, and immune surveillance in humans. Malfunction of lymphatic vasculature leads to several human diseases. Understanding the fundamental mechanism in lymphatic vascular development not only expand our knowledge, but also provide a new therapeutic insight. Recently, Hippo-YAP/TAZ signaling pathway, a key mechanism of organ size and tissue homeostasis, has emerged as a critical player that regulate lymphatic specification, sprouting, and maturation. In this review, we discuss the mechanistic regulation and pathophysiological significant of Hippo pathway in lymphatic vascular development. [BMB Reports 2021; 54(6): 285-294].


Subject(s)
Hippo Signaling Pathway , Lymphangiogenesis , Lymphatic System/cytology , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , YAP-Signaling Proteins/metabolism , Humans , Lymphatic System/metabolism
6.
Development ; 147(23)2020 12 13.
Article in English | MEDLINE | ID: mdl-33060128

ABSTRACT

Lymphatic vasculature is an integral part of digestive, immune and circulatory systems. The homeobox transcription factor PROX1 is necessary for the development of lymphatic vessels, lymphatic valves (LVs) and lymphovenous valves (LVVs). We and others previously reported a feedback loop between PROX1 and vascular endothelial growth factor-C (VEGF-C) signaling. PROX1 promotes the expression of the VEGF-C receptor VEGFR3 in lymphatic endothelial cells (LECs). In turn, VEGF-C signaling maintains PROX1 expression in LECs. However, the mechanisms of PROX1/VEGF-C feedback loop remain poorly understood. Whether VEGF-C signaling is necessary for LV and LVV development is also unknown. Here, we report for the first time that VEGF-C signaling is necessary for valve morphogenesis. We have also discovered that the transcriptional co-activators YAP and TAZ are required to maintain PROX1 expression in LVs and LVVs in response to VEGF-C signaling. Deletion of Yap and Taz in the lymphatic vasculature of mouse embryos did not affect the formation of LVs or LVVs, but resulted in the degeneration of these structures. Our results have identified VEGF-C, YAP and TAZ as a crucial molecular pathway in valve development.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Homeodomain Proteins/genetics , Lymphangiogenesis/genetics , Trans-Activators/genetics , Tumor Suppressor Proteins/genetics , Vascular Endothelial Growth Factor C/genetics , Animals , Embryo, Mammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Mice , Morphogenesis/genetics , Signal Transduction/genetics , Venous Valves/growth & development , Venous Valves/metabolism , YAP-Signaling Proteins
7.
JCI Insight ; 5(14)2020 07 23.
Article in English | MEDLINE | ID: mdl-32544090

ABSTRACT

During the growth of lymphatic vessels (lymphangiogenesis), lymphatic endothelial cells (LECs) at the growing front sprout by forming filopodia. Those tip cells are not exposed to circulating lymph, as they are not lumenized. In contrast, LECs that trail the growing front are exposed to shear stress, become quiescent, and remodel into stable vessels. The mechanisms that coordinate the opposed activities of lymphatic sprouting and maturation remain poorly understood. Here, we show that the canonical tip cell marker Delta-like 4 (DLL4) promotes sprouting lymphangiogenesis by enhancing VEGF-C/VEGF receptor 3 (VEGFR3) signaling. However, in lumenized lymphatic vessels, laminar shear stress (LSS) inhibits the expression of DLL4, as well as additional tip cell markers. Paradoxically, LSS also upregulates VEGF-C/VEGFR3 signaling in LECs, but sphingosine 1-phosphate receptor 1 (S1PR1) activity antagonizes LSS-mediated VEGF-C signaling to promote lymphatic vascular quiescence. Correspondingly, S1pr1 loss in LECs induced lymphatic vascular hypersprouting and hyperbranching, which could be rescued by reducing Vegfr3 gene dosage in vivo. In addition, S1PR1 regulates lymphatic vessel maturation by inhibiting RhoA activity to promote membrane localization of the tight junction molecule claudin-5. Our findings suggest a potentially new paradigm in which LSS induces quiescence and promotes the survival of LECs by downregulating DLL4 and enhancing VEGF-C signaling, respectively. S1PR1 dampens LSS/VEGF-C signaling, thereby preventing sprouting from quiescent lymphatic vessels. These results also highlight the distinct roles that S1PR1 and DLL4 play in LECs when compared with their known roles in the blood vasculature.


Subject(s)
Lymphangiogenesis/genetics , Sphingosine-1-Phosphate Receptors/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Animals , Cell Line , Cell Proliferation , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Membrane Proteins/genetics , Mice , Pseudopodia/genetics , Pseudopodia/metabolism , Signal Transduction , Stress, Mechanical
8.
Development ; 146(21)2019 11 05.
Article in English | MEDLINE | ID: mdl-31582413

ABSTRACT

Mutations in the transcription factor GATA2 cause lymphedema. GATA2 is necessary for the development of lymphatic valves and lymphovenous valves, and for the patterning of lymphatic vessels. Here, we report that GATA2 is not necessary for valvular endothelial cell (VEC) differentiation. Instead, GATA2 is required for VEC maintenance and morphogenesis. GATA2 is also necessary for the expression of the cell junction molecules VE-cadherin and claudin 5 in lymphatic vessels. We identified miR-126 as a target of GATA2, and miR-126-/- embryos recapitulate the phenotypes of mice lacking GATA2. Primary human lymphatic endothelial cells (HLECs) lacking GATA2 (HLECΔGATA2) have altered expression of claudin 5 and VE-cadherin, and blocking miR-126 activity in HLECs phenocopies these changes in expression. Importantly, overexpression of miR-126 in HLECΔGATA2 significantly rescues the cell junction defects. Thus, our work defines a new mechanism of GATA2 activity and uncovers miR-126 as a novel regulator of mammalian lymphatic vascular development.


Subject(s)
Endothelial Cells/metabolism , GATA2 Transcription Factor/metabolism , MicroRNAs/metabolism , Mutation , Angiopoietin-2/metabolism , Animals , CRISPR-Cas Systems , Calcium-Binding Proteins/metabolism , Cell Differentiation , Cell Line , Claudin-5/metabolism , EGF Family of Proteins/metabolism , Endothelium, Vascular/metabolism , Female , Gene Deletion , Humans , Lymphatic Vessels/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA-Seq
9.
Cell Rep ; 28(9): 2397-2412.e4, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31461654

ABSTRACT

The lymphatic vasculature requires intraluminal valves to maintain forward lymph flow. Lymphatic valves form and are constantly maintained by oscillatory fluid flow throughout life, yet the earliest steps of how lymphatic endothelial cells are able to respond to fluid shear stress remain unknown. Here, we show that the adherens junction protein VE-cadherin is required for the upregulation of valve-specific transcription factors. Conditional deletion of VE-cadherin in vivo prevented valve formation in the embryo and caused postnatal regression of nearly all lymphatic valves in multiple tissues. Since VE-cadherin is known to signal through ß-catenin and the VEGFR/AKT pathway, each pathway was probed. Expression of a constitutively active ß-catenin mutant or direct pharmacologic activation of AKT in vivo significantly rescued valve regression in the VE-cadherin-deficient lymphatic vessels. In conclusion, VE-cadherin-dependent signaling is required for lymphatic valve formation and maintenance and therapies to augment downstream pathways hold potential to treat lymphedema in patients.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Lymphatic Vessels/metabolism , Animals , Antigens, CD/genetics , Cadherins/genetics , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Lymphatic Vessels/embryology , Lymphatic Vessels/physiology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , beta Catenin/metabolism
10.
Proc Natl Acad Sci U S A ; 116(19): 9423-9432, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31000600

ABSTRACT

The Hippo-YAP/TAZ signaling pathway plays a pivotal role in growth control during development and regeneration and its dysregulation is widely implicated in various cancers. To further understand the cellular and molecular mechanisms underlying Hippo signaling regulation, we have found that activities of core Hippo signaling components, large tumor suppressor (LATS) kinases and YAP/TAZ transcription factors, oscillate during mitotic cell cycle. We further identified that the anaphase-promoting complex/cyclosome (APC/C)Cdh1 E3 ubiquitin ligase complex, which plays a key role governing eukaryotic cell cycle progression, intrinsically regulates Hippo signaling activities. CDH1 recognizes LATS kinases to promote their degradation and, hence, YAP/TAZ regulation by LATS phosphorylation is under cell cycle control. As a result, YAP/TAZ activities peak in G1 phase. Furthermore, we show in Drosophila eye and wing development that Cdh1 is required in vivo to regulate the LATS homolog Warts with a conserved mechanism. Cdh1 reduction increased Warts levels, which resulted in reduction of the eye and wing sizes in a Yorkie dependent manner. Therefore, LATS degradation by APC/CCdh1 represents a previously unappreciated and evolutionarily conserved layer of Hippo signaling regulation.


Subject(s)
Anaphase-Promoting Complex-Cyclosome/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cdh1 Proteins/metabolism , Drosophila Proteins/metabolism , G1 Phase/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Anaphase-Promoting Complex-Cyclosome/genetics , Animals , Antigens, CD/genetics , Cadherins/genetics , Cdh1 Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics
11.
JCI Insight ; 4(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30676326

ABSTRACT

The lymphatic system plays crucial roles in tissue homeostasis, lipid absorption, and immune cell trafficking. Although lymphatic valves ensure unidirectional lymph flows, the flow itself controls lymphatic valve formation. Here, we demonstrate that a mechanically activated ion channel Piezo1 senses oscillating shear stress (OSS) and incorporates the signal into the genetic program controlling lymphatic valve development and maintenance. Time-controlled deletion of Piezo1 using a pan-endothelial Cre driver (Cdh5[PAC]-CreERT2) or lymphatic-specific Cre driver (Prox1-CreERT2) equally inhibited lymphatic valve formation in newborn mice. Furthermore, Piezo1 deletion in adult lymphatics caused substantial lymphatic valve degeneration. Piezo1 knockdown in cultured lymphatic endothelial cells (LECs) largely abrogated the OSS-induced upregulation of the lymphatic valve signature genes. Conversely, ectopic Piezo1 overexpression upregulated the lymphatic valve genes in the absence of OSS. Remarkably, activation of Piezo1 using chemical agonist Yoda1 not only accelerated lymphatic valve formation in animals, but also triggered upregulation of some lymphatic valve genes in cultured LECs without exposure to OSS. In summary, our studies together demonstrate that Piezo1 is the force sensor in the mechanotransduction pathway controlling lymphatic valve development and maintenance, and Piezo1 activation is a potentially novel therapeutic strategy for congenital and surgery-associated lymphedema.


Subject(s)
Ion Channels/metabolism , Lymphangiogenesis/genetics , Lymphangiogenesis/physiology , Lymphatic Vessels/metabolism , Transcriptome , Animals , Antigens, CD , Cadherins , Endothelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Ion Channels/genetics , Lymphatic Vessels/pathology , Mechanotransduction, Cellular/physiology , Mice , Mice, Knockout , Models, Animal , Stress, Mechanical , Up-Regulation
12.
Cell Rep ; 25(3): 571-584.e5, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332639

ABSTRACT

Wnt/ß-catenin signaling is necessary for lymphatic vascular development. Oscillatory shear stress (OSS) enhances Wnt/ß-catenin signaling in cultured lymphatic endothelial cells (LECs) to induce expression of the lymphedema-associated transcription factors GATA2 and FOXC2. However, the mechanisms by which OSS regulates Wnt/ß-catenin signaling and GATA2 and FOXC2 expression are unknown. We show that OSS activates autocrine Wnt/ß-catenin signaling in LECs in vitro. Tissue-specific deletion of Wntless, which is required for the secretion of Wnt ligands, reveals that LECs and vascular smooth muscle cells are complementary sources of Wnt ligands that regulate lymphatic vascular development in vivo. Further, the LEC master transcription factor PROX1 forms a complex with ß-catenin and the TCF/LEF transcription factor TCF7L1 to enhance Wnt/ß-catenin signaling and promote FOXC2 and GATA2 expression in LECs. Thus, our work defines Wnt sources, reveals that PROX1 directs cell fate by acting as a Wnt signaling component, and dissects the mechanisms of PROX1 and Wnt synergy.


Subject(s)
Endothelial Cells/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Muscle, Smooth, Vascular/cytology , Tumor Suppressor Proteins/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Female , Forkhead Transcription Factors/metabolism , GATA2 Transcription Factor/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Transcription Factor 7-Like 1 Protein/metabolism , Wnt Signaling Pathway
13.
Gut ; 67(9): 1692-1703, 2018 09.
Article in English | MEDLINE | ID: mdl-28866620

ABSTRACT

OBJECTIVE: Hippo signalling is a recently identified major oncosuppressive pathway that plays critical roles in inhibiting hepatocyte proliferation, survival and hepatocellular carcinoma (HCC) formation. Hippo kinase (Mst1 and Mst2) inhibits HCC proliferation by suppressing Yap/Taz transcription activities. As human HCC is mainly driven by chronic liver inflammation, it is not clear whether Hippo signalling inhibits HCC by shaping its inflammatory microenvironment. DESIGN: We have established a genetic HCC model by deleting Mst1 and Mst2 in hepatocytes. Functions of inflammatory responses in this model were characterised by molecular, cellular and FACS analysis, immunohistochemistry and genetic deletion of monocyte chemoattractant protein-1 (Mcp1) or Yap. Human HCC databases and human HCC samples were analysed by immunohistochemistry. RESULTS: Genetic deletion of Mst1 and Mst2 in hepatocytes (DKO) led to HCC development, highly upregulated Mcp1 expression and massive infiltration of macrophages with mixed M1 and M2 phenotypes. Macrophage ablation or deletion of Mcp1 in DKO mice markedly reduced hepatic inflammation and HCC development. Moreover, Yap removal abolished induction of Mcp1 expression and restored normal liver growth in the Mst1/Mst2 DKO mice. Finally, we showed that MCP1 is a direct transcription target of YAP in hepatocytes and identified a strong gene expression correlation between YAP targets and MCP-1 in human HCCs. CONCLUSIONS: Hippo signalling in hepatocytes maintains normal liver growth by suppressing macrophage infiltration during protumoural microenvironment formation through the inhibition of Yap-dependent Mcp1 expression, providing new targets and strategies to treat HCCs.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms, Experimental/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Animals , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/therapy , Cell Proliferation/genetics , Cell Transformation, Neoplastic , Hepatocyte Growth Factor/genetics , Hepatocytes/metabolism , Hippo Signaling Pathway , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/therapy , Mice , Mice, Mutant Strains , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Serine-Threonine Kinase 3 , Signal Transduction/genetics , Transcription Factors/genetics
14.
Dis Model Mech ; 10(11): 1273-1287, 2017 11 01.
Article in English | MEDLINE | ID: mdl-29125824

ABSTRACT

The circulatory system consists of the heart, blood vessels and lymphatic vessels, which function in parallel to provide nutrients and remove waste from the body. Vascular function depends on valves, which regulate unidirectional fluid flow against gravitational and pressure gradients. Severe valve disorders can cause mortality and some are associated with severe morbidity. Although cardiac valve defects can be treated by valve replacement surgery, no treatment is currently available for valve disorders of the veins and lymphatics. Thus, a better understanding of valves, their development and the progression of valve disease is warranted. In the past decade, molecules that are important for vascular function in humans have been identified, with mouse studies also providing new insights into valve formation and function. Intriguing similarities have recently emerged between the different types of valves concerning their molecular identity, architecture and development. Shear stress generated by fluid flow has also been shown to regulate endothelial cell identity in valves. Here, we review our current understanding of valve development with an emphasis on its mechanobiology and significance to human health, and highlight unanswered questions and translational opportunities.


Subject(s)
Heart Valves/physiology , Animals , Genetic Predisposition to Disease , Heart Valve Diseases/genetics , Heart Valve Diseases/pathology , Humans , Models, Biological , Stress, Mechanical
15.
Biochim Biophys Acta Mol Basis Dis ; 1863(4): 1013-1022, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28185954

ABSTRACT

Mice null for wild-type p53-induced phosphatase 1 (WIP1) display defects in testis development and spermatogenesis, resulting in reduced fertility. However, the molecular mechanism underlying these abnormalities in the testis remains uncharacterized. We report that the phosphatase activity of WIP1 increases Wnt activity through Nemo-like kinase (NLK). WIP1 directly interacted with NLK, which is highly homologous to p38 MAPK, a WIP1 substrate, and dephosphorylated its activation site. The WIP1-mediated inhibition of NLK activity markedly decreased the phosphorylation of lymphoid enhancer-binding factor 1 (LEF1), enhancing its interaction with ß-catenin. Additionally, WIP1 depletion impaired germ cell development, as evidenced by the expression of Oct4 and the germ cell-specific markers Ddx4, Nanos3 and Dnd1 during the development of germ cells from Oct4-GFP transgenic (OG2) mouse embryonic stem cells (mESCs). The expression of WIP1, whose level was significantly lower after the differentiation of germ cells from mESCs, occurred in parallel with the expression of germ cell development markers and SRY-box 17 (Sox17), a downstream target of Wnt. These results indicate that WIP1 is essential for germ cell development, which is known to require Wnt activity.


Subject(s)
Germ Cells/cytology , Mitogen-Activated Protein Kinases/metabolism , Mouse Embryonic Stem Cells/cytology , Protein Phosphatase 2C/metabolism , Wnt Proteins/metabolism , Animals , Cell Differentiation , Cell Line , Gene Deletion , Gene Expression Regulation, Developmental , Germ Cells/metabolism , HEK293 Cells , Humans , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , Mice, Transgenic , Mouse Embryonic Stem Cells/metabolism , Phosphorylation , Protein Phosphatase 2C/genetics , Protein Serine-Threonine Kinases , Testis/cytology , Testis/metabolism , beta Catenin/metabolism
16.
EMBO Rep ; 18(1): 61-71, 2017 01.
Article in English | MEDLINE | ID: mdl-27979972

ABSTRACT

Hippo signaling controls organ size by regulating cell proliferation and apoptosis. Yes-associated protein (YAP) is a key downstream effector of Hippo signaling, and LATS-mediated phosphorylation of YAP at Ser127 inhibits its nuclear localization and transcriptional activity. Here, we report that Nemo-like kinase (NLK) phosphorylates YAP at Ser128 both in vitro and in vivo, which blocks interaction with 14-3-3 and enhances its nuclear localization. Depletion of NLK increases YAP phosphorylation at Ser127 and reduces YAP-mediated reporter activity. These results suggest that YAP phosphorylation at Ser128 and at Ser127 may be mutually exclusive. We also find that with the increase in cell density, nuclear localization and the level of NLK are reduced, resulting in reduction in YAP phosphorylation at Ser128. Furthermore, knockdown of Nemo (the Drosophila NLK) in fruit fly wing imaginal discs results in reduced expression of the Yorkie (the Drosophila YAP) target genes expanded and DIAP1, while Nemo overexpression reciprocally increased the expression. Overall, our data suggest that NLK/Nemo acts as an endogenous regulator of Hippo signaling by controlling nuclear localization and activity of YAP/Yorkie.


Subject(s)
14-3-3 Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cell Count , Cell Cycle Proteins , Cell Line , Cell Movement , Cell Nucleus/metabolism , Drosophila , Humans , Mice , Nuclear Proteins/chemistry , Phosphorylation , Protein Binding , Protein Transport , Serine/chemistry , Serine/metabolism , Transcription Factors/chemistry , Transcription, Genetic
17.
BMB Rep ; 49(8): 403-4, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27418286

ABSTRACT

The Wnt/ß-catenin signaling is an evolutionarily conserved pathway that plays a pivotal role in embryonic development and adult homeostasis. However, we have limited information about the involvement of Wnt/ß-catenin signaling in the lymphatic vascular system that regulates fluid homeostasis by absorbing interstitial fluid and returning it to blood circulation. In this recent publication we report that canonical Wnt/ß-catenin signaling is highly active and critical for the formation of lymphovenus valves (LVVs) and lymphatic valves (LVs). ß-catenin directly associates with the regulatory elements of the lymphedema-associated transcription factor, FOXC2 and activates its expression in an oscillatory shear stress (OSS)-dependent manner. The phenotype of ß-catenin null embryos was rescued by FOXC2 overexpression. These results suggest that Wnt/ß-catenin signaling is a mechanotransducer that links fluid force with lymphatic vascular development. [BMB Reports 2016; 49(8): 403-404].


Subject(s)
Lymphangiogenesis , Mechanotransduction, Cellular , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Endothelial Cells/metabolism , Humans , Lymphatic System/metabolism , Models, Biological
18.
Genes Dev ; 30(12): 1454-69, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27313318

ABSTRACT

Lymphatic vasculature regulates fluid homeostasis by returning interstitial fluid to blood circulation. Lymphatic endothelial cells (LECs) are the building blocks of the entire lymphatic vasculature. LECs originate as a homogeneous population of cells predominantly from the embryonic veins and undergo stepwise morphogenesis to become the lymphatic capillaries, collecting vessels or valves. The molecular mechanisms underlying the morphogenesis of the lymphatic vasculature remain to be fully understood. Here we show that canonical Wnt/ß-catenin signaling is necessary for lymphatic vascular morphogenesis. Lymphatic vascular-specific ablation of ß-catenin in mice prevents the formation of lymphatic and lymphovenous valves. Additionally, lymphatic vessel patterning is defective in these mice, with abnormal recruitment of mural cells. We found that oscillatory shear stress (OSS), which promotes lymphatic vessel maturation, triggers Wnt/ß-catenin signaling in LECs. In turn, Wnt/ß-catenin signaling controls the expression of several molecules, including the lymphedema-associated transcription factor FOXC2. Importantly, FOXC2 completely rescues the lymphatic vessel patterning defects in mice lacking ß-catenin. Thus, our work reveals that mechanical stimulation is a critical regulator of lymphatic vascular development via activation of Wnt/ß-catenin signaling and, in turn, FOXC2.


Subject(s)
Lymphangiogenesis/physiology , Mechanotransduction, Cellular/physiology , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Cells, Cultured , Endothelial Cells/cytology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Gene Silencing , Humans , Lymphatic Vessels/embryology , Mice , beta Catenin/genetics
19.
Dev Biol ; 409(1): 218-233, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26542011

ABSTRACT

Lymph is returned to the blood circulation exclusively via four lymphovenous valves (LVVs). Despite their vital importance, the architecture and development of LVVs is poorly understood. We analyzed the formation of LVVs at the molecular and ultrastructural levels during mouse embryogenesis and identified three critical steps. First, LVV-forming endothelial cells (LVV-ECs) differentiate from PROX1(+) progenitors and delaminate from the luminal side of the veins. Second, LVV-ECs aggregate, align perpendicular to the direction of lymph flow and establish lympho-venous connections. Finally, LVVs mature with the recruitment of mural cells. LVV morphogenesis is disrupted in four different mouse models of primary lymphedema and the severity of LVV defects correlate with that of lymphedema. In summary, we have provided the first and the most comprehensive analysis of LVV development. Furthermore, our work suggests that aberrant LVVs contribute to lymphedema.


Subject(s)
Lymphatic Vessels/embryology , Lymphedema/embryology , Lymphedema/pathology , Venous Valves/embryology , Animals , Animals, Newborn , Cell Differentiation , Disease Models, Animal , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Lymphatic Vessels/ultrastructure , Mice, Inbred C57BL , Morphogenesis , Penetrance , Phenotype , Venous Valves/ultrastructure
20.
Mol Cells ; 38(8): 723-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26126536

ABSTRACT

Smurf2, a member of the HECT domain E3 ligase family, is well known for its role as a negative regulator of TGF-ß signaling by targeting Smads and TGF-ß receptor. However, the regulatory mechanism of Smurf2 has not been elucidated. Arginine methylation is a type of post-translational modification that produces monomethylated or dimethylated arginine residues. In this report, we demonstrated methylation of Smurf2 by PRMT1. In vitro methylation assay showed that Smurf2, not Smurf1, was methylated by PRMT1. Among the type I PRMT family, only PRMT1 showed activity for Smurf2. Transiently expressed Smurf2 was methylated by PRMT1, indicating Smurf2 is a novel substrate of PRMT1. Using deletion constructs, methylation sites were shown to be located within amino acid region 224-298 of Smurf2. In vitro methylation assay following point mutation of putative methylation sites confirmed the presence of Arg232, Arg234, Arg237, and Arg239. Knockdown of PRMT1 resulted in increased Smurf2 expression as well as inhibition of TGF-ß-mediated reporter activity. Although it is unclear whether or not increased Smurf2 expression can be directly attributed to lack of methylation of arginine residues, our results suggest that methylation by PRMT1 may regulate Smurf2 stability and control TGF-ß signaling.


Subject(s)
Protein-Arginine N-Methyltransferases/metabolism , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Arginine/metabolism , HEK293 Cells , Humans , Methylation , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL