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 Biomed Sci ; 30(1): 33, 2023 May 26.
Article in English | MEDLINE | ID: mdl-37237303

ABSTRACT

The SCUBE [Signal peptide-Complement C1r/C1s, Uegf, Bmp1 (CUB)-Epithelial growth factor domain-containing protein] family consists of three proteins in vertebrates, SCUBE1, 2 and 3, which are highly conserved in zebrafish, mice and humans. Each SCUBE gene encodes a polypeptide of approximately 1000 amino acids that is organized into five modular domains: (1) an N-terminal signal peptide sequence, (2) nine tandem epidermal growth factor (EGF)-like repeats, (3) a large spacer region, (4) three cysteine-rich (CR) motifs, and (5) a CUB domain at the C-terminus. Murine Scube genes are expressed individually or in combination during the development of various tissues, including those in the central nervous system and the axial skeleton. The cDNAs of human SCUBE orthologs were originally cloned from vascular endothelial cells, but SCUBE expression has also been found in platelets, mammary ductal epithelium and osteoblasts. Both soluble and membrane-associated SCUBEs have been shown to play important roles in physiology and pathology. For instance, upregulation of SCUBEs has been reported in acute myeloid leukemia, breast cancer and lung cancer. In addition, soluble SCUBE1 is released from activated platelets and can be used as a clinical biomarker for acute coronary syndrome and ischemic stroke. Soluble SCUBE2 enhances distal signaling by facilitating the secretion of dual-lipidated hedgehog from nearby ligand-producing cells in a paracrine manner. Interestingly, the spacer regions and CR motifs can increase or enable SCUBE binding to cell surfaces via electrostatic or glycan-lectin interactions. As such, membrane-associated SCUBEs can function as coreceptors that enhance the signaling activity of various serine/threonine kinase or tyrosine kinase receptors. For example, membrane-associated SCUBE3 functions as a coreceptor that promotes signaling in bone morphogenesis. In humans, SCUBE3 mutations are linked to abnormalities in growth and differentiation of both bones and teeth. In addition to studies on human SCUBE function, experimental results from genetically modified mouse models have yielded important insights in the field of systems biology. In this review, we highlight novel molecular discoveries and critical directions for future research on SCUBE proteins in the context of cancer, skeletal disease and cardiovascular disease.


Subject(s)
Endothelial Cells , Zebrafish , Humans , Animals , Mice , Zebrafish/metabolism , Endothelial Cells/metabolism , Cell Membrane/metabolism , Protein Sorting Signals , Biology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism
2.
Haematologica ; 108(5): 1284-1299, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36005562

ABSTRACT

A hallmark of mixed lineage leukemia gene-rearranged (MLL-r) acute myeloid leukemia that offers an opportunity for targeted therapy is addiction to protein tyrosine kinase signaling. One such signal is the receptor tyrosine kinase Fms-like receptor tyrosine kinase 3 (FLT3) upregulated by cooperation of the transcription factors homeobox A9 (HOXA9) and Meis homeobox 1 (MEIS1). Signal peptide-CUB-EGF-like repeat-containing protein (SCUBE) family proteins have previously been shown to act as a co-receptor for augmenting signaling activity of a receptor tyrosine kinase (e.g., vascular endothelial growth factor receptor). However, whether SCUBE1 is involved in the pathological activation of FLT3 during MLL-r leukemogenesis remains unknown. Here we first show that SCUBE1 is a direct target of HOXA9/MEIS1 that is highly expressed on the MLL-r cell surface and predicts poor prognosis in de novo acute myeloid leukemia. We further demonstrate, by using a conditional knockout mouse model, that Scube1 is required for both the initiation and maintenance of MLL-AF9-induced leukemogenesis in vivo. Further proteomic, molecular and biochemical analyses revealed that the membrane-tethered SCUBE1 binds to the FLT3 ligand and the extracellular ligand-binding domains of FLT3, thus facilitating activation of the signal axis FLT3-LYN (a non-receptor tyrosine kinase) to initiate leukemic growth and survival signals. Importantly, targeting surface SCUBE1 by an anti-SCUBE1 monomethyl auristatin E antibody-drug conjugate led to significantly decreased cell viability specifically in MLL-r leukemia. Our study indicates a novel function of SCUBE1 in leukemia and unravels the molecular mechanism of SCUBE1 in MLL-r acute myeloid leukemia. Thus, SCUBE1 is a potential therapeutic target for treating leukemia caused by MLL rearrangements.


Subject(s)
Epidermal Growth Factor , Leukemia, Myeloid, Acute , Animals , Mice , fms-Like Tyrosine Kinase 3 , Leukemia, Myeloid, Acute/pathology , Mice, Knockout , Myeloid Ecotropic Viral Integration Site 1 Protein , Myeloid-Lymphoid Leukemia Protein/metabolism , Proteomics , Receptor Protein-Tyrosine Kinases , Vascular Endothelial Growth Factor A
3.
Dev Cell ; 57(14): 1758-1775.e7, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35777353

ABSTRACT

Hair follicle stem cells are regulated by dermal papilla fibroblasts, their principal signaling niche. Overactivation of Hedgehog signaling in the niche dramatically accelerates hair growth and induces follicle multiplication in mice. On single-cell RNA sequencing, dermal papilla fibroblasts increase heterogeneity to include new Wnt5ahigh states. Transcriptionally, mutant fibroblasts activate regulatory networks for Gli1, Alx3, Ebf1, Hoxc8, Sox18, and Zfp239. These networks jointly upregulate secreted factors for multiple hair morphogenesis and hair-growth-related pathways. Among these is non-conventional TGF-ß ligand Scube3. We show that in normal mouse skin, Scube3 is expressed only in dermal papillae of growing, but not in resting follicles. SCUBE3 protein microinjection is sufficient to induce new hair growth, and pharmacological TGF-ß inhibition rescues mutant hair hyper-activation phenotype. Moreover, dermal-papilla-enriched expression of SCUBE3 and its growth-activating effect are partially conserved in human scalp hair follicles. Thus, Hedgehog regulates mesenchymal niche function in the hair follicle via SCUBE3/TGF-ß mechanism.


Subject(s)
Hair Follicle , Hedgehog Proteins , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Fibroblasts/metabolism , Hair , Hair Follicle/metabolism , Hedgehog Proteins/metabolism , Humans , Mice , SOXF Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
4.
Circ Res ; 131(1): 6-20, 2022 06 24.
Article in English | MEDLINE | ID: mdl-35611699

ABSTRACT

BACKGROUND: The sino atrial node (SAN) is characterized by the microenvironment of pacemaker cardiomyocytes (PCs) encased with fibroblasts. An altered microenvironment leads to rhythm failure. Operable cell or tissue models are either generally lacking or difficult to handle. The biological process behind the milieu of SANs to evoke pacemaker rhythm is unknown. We explored how fibroblasts interact with PCs and regulate metabolic reprogramming and rhythmic activity in the SAN. METHODS: Tbx18 (T-box transcription factor 18)-induced PCs and fibroblasts were used for cocultures and engineered tissues, which were used as the in vitro models to explore how fibroblasts regulate the functional integrity of SANs. RNA-sequencing, metabolomics, and cellular and molecular techniques were applied to characterize the molecular signals underlying metabolic reprogramming and identify its critical regulators. These pathways were further validated in vivo in rodents and induced human pluripotent stem cell-derived cardiomyocytes. RESULTS: We observed that rhythmicity in Tbx18-induced PCs was regulated by aerobic glycolysis. Fibroblasts critically activated metabolic reprogramming and aerobic glycolysis within PCs, and, therefore, regulated pacemaker activity in PCs. The metabolic reprogramming was attributed to the exclusive induction of Aldoc (aldolase c) within PCs after fibroblast-PC integration. Fibroblasts activated the integrin-dependent mitogen-activated protein kinase-E2F1 signal through cell-cell contact and turned on Aldoc expression in PCs. Interruption of fibroblast-PC interaction or Aldoc knockdown nullified electrical activity. Engineered Tbx18-PC tissue sheets were generated to recapitulate the microenvironment within SANs. Aldoc-driven rhythmic machinery could be replicated within tissue sheets. Similar machinery was faithfully validated in de novo PCs of adult mice and rats, and in human PCs derived from induced pluripotent stem cells. CONCLUSIONS: Fibroblasts drive Aldoc-mediated metabolic reprogramming and rhythmic regulation in SANs. This work details the cellular machinery behind the complex milieu of vertebrate SANs and opens a new direction for future therapy.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Animals , Cellular Reprogramming , Coculture Techniques , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Myocytes, Cardiac/metabolism , Rats , Sinoatrial Node/metabolism
5.
Cardiovasc Res ; 118(4): 1074-1087, 2022 03 16.
Article in English | MEDLINE | ID: mdl-33788916

ABSTRACT

AIMS: The secreted and membrane-anchored signal peptide-CUB-EGF domain-containing proteins (SCUBE) gene family composed of three members was originally identified from endothelial cells (ECs). We recently showed that membrane SCUBE2 binds vascular endothelial growth factor (VEGF) and acts as a co-receptor for VEGF receptor 2 to modulate EC migration, proliferation, and tube formation during postnatal and tumour angiogenesis. However, whether these SCUBE genes cooperate in modulating VEGF signalling during embryonic vascular development remains unknown. METHODS AND RESULTS: To further dissect the genetic interactions of these scube genes, transcription activator-like effector nuclease-mediated genome editing was used to generate knockout (KO) alleles of each scube gene. No overt vascular phenotypes were seen in any single scube KO mutants because of compensation by other scube genes during zebrafish development. However, scube1 and scube2 double KO (DKO) severely impaired EC filopodia extensions, migration, and proliferation, thus disrupting proper vascular lumen formation during vasculogenesis and angiogenesis as well as development of the organ-specific intestinal vasculature. Further genetic, biochemical, and molecular analyses revealed that Scube1 and Scube2 might act cooperatively at the cell-surface receptor level to facilitate Vegfa signalling during zebrafish embryonic vascularization. CONCLUSIONS: We showed for the first time that cooperation between scube1 and scube2 is critical for proper regulation of angiogenic cell behaviours and formation of functional vessels during zebrafish embryonic development.


Subject(s)
Vascular Endothelial Growth Factor A , Zebrafish , Adaptor Proteins, Signal Transducing/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Endothelial Cells/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/genetics , Zebrafish/metabolism
6.
Nat Biomed Eng ; 6(4): 421-434, 2022 04.
Article in English | MEDLINE | ID: mdl-34811487

ABSTRACT

Pacemaker cells can be differentiated from stem cells or transdifferentiated from quiescent mature cardiac cells via genetic manipulation. Here we show that the exposure of rat quiescent ventricular cardiomyocytes to a silk-fibroin hydrogel activates the direct conversion of the quiescent cardiomyocytes to pacemaker cardiomyocytes by inducing the ectopic expression of the vascular endothelial cell-adhesion glycoprotein cadherin. The silk-fibroin-induced pacemaker cells exhibited functional and morphological features of genuine sinoatrial-node cardiomyocytes in vitro, and pacemaker cells generated via the injection of silk fibroin in the left ventricles of rats functioned as a surrogate in situ sinoatrial node. Biomaterials with suitable surface structure, mechanics and biochemistry could facilitate the scalable production of biological pacemakers for human use.


Subject(s)
Fibroins , Myocytes, Cardiac , Animals , Biocompatible Materials , Cell Differentiation , Fibroins/metabolism , Fibroins/pharmacology , Rats , Sinoatrial Node/metabolism
7.
Am J Hum Genet ; 108(1): 115-133, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33308444

ABSTRACT

Signal peptide-CUB-EGF domain-containing protein 3 (SCUBE3) is a member of a small family of multifunctional cell surface-anchored glycoproteins functioning as co-receptors for a variety of growth factors. Here we report that bi-allelic inactivating variants in SCUBE3 have pleiotropic consequences on development and cause a previously unrecognized syndromic disorder. Eighteen affected individuals from nine unrelated families showed a consistent phenotype characterized by reduced growth, skeletal features, distinctive craniofacial appearance, and dental anomalies. In vitro functional validation studies demonstrated a variable impact of disease-causing variants on transcript processing, protein secretion and function, and their dysregulating effect on bone morphogenetic protein (BMP) signaling. We show that SCUBE3 acts as a BMP2/BMP4 co-receptor, recruits the BMP receptor complexes into raft microdomains, and positively modulates signaling possibly by augmenting the specific interactions between BMPs and BMP type I receptors. Scube3-/- mice showed craniofacial and dental defects, reduced body size, and defective endochondral bone growth due to impaired BMP-mediated chondrogenesis and osteogenesis, recapitulating the human disorder. Our findings identify a human disease caused by defective function of a member of the SCUBE family, and link SCUBE3 to processes controlling growth, morphogenesis, and bone and teeth development through modulation of BMP signaling.


Subject(s)
Bone and Bones/metabolism , Calcium-Binding Proteins/metabolism , Developmental Disabilities/metabolism , Osteogenesis/physiology , Signal Transduction/physiology , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Line , Cell Line, Tumor , Female , Gene Expression Regulation, Developmental/physiology , HEK293 Cells , Hep G2 Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , MCF-7 Cells , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
8.
Exp Cell Res ; 370(2): 444-453, 2018 09 15.
Article in English | MEDLINE | ID: mdl-29981340

ABSTRACT

BACKGROUND: Metastatic and/or recurrent breast carcinomas are leading causes of cancer-related death worldwide. Breast cancer stem cells (BCSCs) have been implicated in cancer metastases and progression, thus, the need for the discovery and development of effective BCSCs-specific therapies against metastatic and triple negative breast cancer (TNBC). The expression of SCUBE2, originally identified in vascular endothelia, then in several non-endothelial cell types, is downregulated in invasive breast carcinomas. However, the role of SCUBE2 in BCSCs remains unknown. This present study investigated the probable involvements of SCUBE2 in BCSCs and TNBC metastasis. METHODS: The mRNA expression of SCUBE2, stemness and EMT markers in MDA-MB-231 and Hs578T tumorspheres or adherent cells were evaluated by qRT-PCR and microarray analyses. Using gene overexpression, in vitro migration and invasion assays, as well as in vivo bioluminescence imaging, we evaluated the role of SCUBE2 in MDA-MB-231 or Hs578T BCSCs. Western blot and cytotoxicity assays helped identify and validate SCUBE2 molecular target(s) and inhibitor(s). RESULTS: Concurrently increased SCUBE2 expression and cell motility were observed in TNBC tumorspheres compared to the parental adherent cells. SCUBE2 overexpression augmented BCSCs motility in vitro, and enhanced TNBC metastasis in vivo. While SCUBE2 overexpression activated Notch signaling its downregulation suppressed Notch signal effectors NICD, Jagged 1, HEY1, and HES1. CONCLUSIONS: We demonstrate that SCUBE2 expression is upregulated in BCSCs, promote EMT and enhance TNBC metastasis by activating Notch signaling. This reveals a potential druggable molecular target and an effective therapeutic strategy against metastatic and aggressive TNBC.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Membrane Proteins/metabolism , Neoplastic Stem Cells/metabolism , Triple Negative Breast Neoplasms/metabolism , Adaptor Proteins, Signal Transducing , Aggression/physiology , Breast/pathology , Calcium-Binding Proteins , Cell Line, Tumor , Cell Proliferation/physiology , Epithelial-Mesenchymal Transition/genetics , Humans , Membrane Proteins/genetics , Transcriptional Activation/physiology , Triple Negative Breast Neoplasms/genetics
9.
Nat Cell Biol ; 20(4): 479-491, 2018 04.
Article in English | MEDLINE | ID: mdl-29593326

ABSTRACT

Activation of metastatic reprogramming is critical for tumour metastasis. However, more detailed knowledge of the underlying mechanism is needed to enable targeted intervention. Here, we show that paraspeckle component 1 (PSPC1), identified in an aberrant 13q12.11 locus, is upregulated and associated with poor survival in patients with cancer. PSPC1 promotes tumorigenesis, epithelial-to-mesenchymal transition (EMT), stemness and metastasis in multiple cell types and in spontaneous mouse cancer models. PSPC1 is the master activator for transcription factors of EMT and stemness and accompanies c-Myc activation to facilitate tumour growth. PSPC1 increases transforming growth factor-ß1 (TGF-ß1) secretion through an interaction with phosphorylated and nuclear Smad2/3 to potentiate TGF-ß1 autocrine signalling. Moreover, PSPC1 acts as a contextual determinant of the TGF-ß1 pro-metastatic switch to alter Smad2/3 binding preference from tumour-suppressor to pro-metastatic genes. Having validated the PSPC1-Smads-TGF-ß1 axis in various cancers, we conclude that PSPC1 is a master activator of pro-metastatic switches and a potential target for anti-metastasis drugs.


Subject(s)
Autocrine Communication , Cell Movement , Epithelial-Mesenchymal Transition , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , A549 Cells , Animals , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Nuclear Proteins/genetics , PC-3 Cells , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA-Binding Proteins/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Time Factors , Transforming Growth Factor beta1/genetics
10.
Arterioscler Thromb Vasc Biol ; 38(5): 1202-1215, 2018 05.
Article in English | MEDLINE | ID: mdl-29545238

ABSTRACT

OBJECTIVE: SCUBE2 (signal peptide-CUB-EGF domain-containing protein 2), expressed on the endothelial cell surface, functions as a novel coreceptor for VEGFR2 (vascular endothelial growth factor receptor 2) and enhances VEGF-induced signaling in adult angiogenesis. However, whether SCUBE2 plays a role in pathological angiogenesis and whether anti-SCUBE2 antibody is an effective strategy for blocking tumor angiogenesis remain unknown. The aim of this study was to investigate the pathological role and targeting therapy of SCUBE2 in tumor vasculature. APPROACH AND RESULTS: Immunohistochemistry revealed that SCUBE2 is highly expressed in endothelial cells of numerous carcinomas. Genetic endothelial cell knockout of SCUBE2 and pharmacological inhibition with the anti-SCUBE2 monoclonal antibody SP.B1 significantly reduced xenograft tumor growth, decreased tumor vascular density, increased apoptosis, and decreased the proliferation of tumor cells. Mechanistic studies revealed that SP.B1 binds to SCUBE2 and induces its internalization for lysosomal degradation, thereby reducing its cell surface level and inhibiting the binding of and downstream signaling of VEGF, including VEGFR2 phosphorylation and AKT/MAPK (mitogen-activated protein kinase) activation. Importantly, dual combination therapy with anti-SCUBE2 monoclonal antibody and anti-VEGF antibody or chemotherapy was more effective than single-agent therapy. CONCLUSIONS: Endothelial cell surface SCUBE2 is a VEGFR2 coreceptor essential for pathological tumor angiogenesis, and anti-SCUBE2 monoclonal antibody acting as an internalization inducer may provide a potent combination therapy for tumor angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Endothelial Cells/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Neovascularization, Pathologic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adaptor Proteins, Signal Transducing , Animals , Apoptosis/drug effects , Calcium-Binding Proteins , Cell Proliferation/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Time Factors , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Breast Cancer Res ; 19(1): 111, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28982386

ABSTRACT

BACKGROUND: Core fucosylation (addition of fucose in α-1,6-linkage to core N-acetylglucosamine of N-glycans) catalyzed by fucosyltransferase 8 (FUT8) is critical for signaling receptors involved in many physiological and pathological processes such as cell growth, adhesion, and tumor metastasis. Transforming growth factor-ß (TGF-ß)-induced epithelial-mesenchymal transition (EMT) regulates the invasion and metastasis of breast tumors. However, whether receptor core fucosylation affects TGF-ß signaling during breast cancer progression remains largely unknown. METHOD: In this study, gene expression profiling and western blot were used to validate the EMT-associated expression of FUT8. Lentivirus-mediated gain-of-function study, short hairpin RNA (shRNA) or CRISPR/Cas9-mediated loss-of-function studies and pharmacological inhibition of FUT8 were used to elucidate the molecular function of FUT8 during TGF-ß-induced EMT in breast carcinoma cells. In addition, lectin blot, luciferase assay, and in vitro ligand binding assay were employed to demonstrate the involvement of FUT8 in the TGF-ß1 signaling pathway. The role of FUT8 in breast cancer migration, invasion, and metastasis was confirmed using an in vitro transwell assay and mammary fat pad xenograft in vivo tumor model. RESULTS: Gene expression profiling analysis revealed that FUT8 is upregulated in TGF-ß-induced EMT; the process was associated with the migratory and invasive abilities of several breast carcinoma cell lines. Gain-of-function and loss-of-function studies demonstrated that FUT8 overexpression stimulated the EMT process, whereas FUT8 knockdown suppressed the invasiveness of highly aggressive breast carcinoma cells. Furthermore, TGF-ß receptor complexes might be core fucosylated by FUT8 to facilitate TGF-ß binding and enhance downstream signaling. Importantly, FUT8 inhibition suppressed the invasive ability of highly metastatic breast cancer cells and impaired their lung metastasis. CONCLUSIONS: Our results reveal a positive feedback mechanism of FUT8-mediated receptor core fucosylation that promotes TGF-ß signaling and EMT, thus stimulating breast cancer cell invasion and metastasis.


Subject(s)
Breast Neoplasms/genetics , Fucosyltransferases/genetics , Neoplasm Invasiveness/genetics , Transforming Growth Factor beta1/genetics , Breast Neoplasms/pathology , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Fucose/genetics , Fucose/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Lentivirus/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Phosphorylation , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/genetics
12.
Arterioscler Thromb Vasc Biol ; 37(1): 144-155, 2017 01.
Article in English | MEDLINE | ID: mdl-27834687

ABSTRACT

OBJECTIVE: Vascular endothelial growth factor (VEGF), a major mediator of angiogenesis, exerts its proangiogenic action by binding to VEGFR2 (VEGF receptor 2), the activity of which is further modulated by VEGFR2 coreceptors such as neuropilins. However, whether VEGFR2 is regulated by additional coreceptors is not clear. To investigate whether SCUBE2 (signal peptide-CUB-EGF domain-containing protein 2), a peripheral membrane protein expressed in vascular endothelial cells (ECs) known to bind other signaling receptors, functions as a VEGFR2 coreceptor and to verify the role of SCUBE2 in the VEGF-induced angiogenesis. APPROACH AND RESULTS: SCUBE2 lentiviral overexpression in human ECs increased and short hairpin RNA knockdown inhibited VEGF-induced EC growth and capillary-like network formation on Matrigel. Like VEGF, endothelial SCUBE2 was upregulated by hypoxia-inducible factor-1α at both mRNA and protein levels. EC-specific Scube2 knockout mice were not defective in vascular development but showed impaired VEGF-induced neovascularization in implanted Matrigel plugs and recovery of blood flow after hind-limb ischemia. Coimmunoprecipitation and ligand-binding assays showed that SCUBE2 forms a complex with VEGF and VEGFR2, thus acting as a coreceptor to facilitate VEGF binding and augment VEGFR2 signal activity. SCUBE2 knockdown or genetic knockout suppressed and its overexpression promoted the VEGF-induced activation of downstream proangiogenic and proliferating signals, including VEGFR2 phosphorylation and mitogen-activated protein kinase or AKT activation. CONCLUSIONS: Endothelial SCUBE2 may be a novel coreceptor for VEGFR2 and potentiate VEGF-induced signaling in adult angiogenesis.


Subject(s)
Endothelial Cells/drug effects , Intercellular Signaling Peptides and Proteins/metabolism , Ischemia/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/agonists , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Genotype , Hindlimb , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Ischemia/genetics , Ischemia/physiopathology , Male , Membrane Proteins/genetics , Mice, Knockout , Phenotype , Phosphorylation , Protein Binding , RNA Interference , Signal Transduction/drug effects , Tissue Culture Techniques , Transfection
13.
J Bone Miner Res ; 30(7): 1255-67, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25639508

ABSTRACT

Signal peptide-CUB-EGF domain-containing protein 2 (SCUBE2) belongs to a secreted and membrane-tethered multidomain SCUBE protein family composed of three members found in vertebrates and mammals. Recent reports suggested that zebrafish scube2 could facilitate sonic hedgehog (Shh) signaling for proper development of slow muscle. However, whether SCUBE2 can regulate the signaling activity of two other hedgehog ligands (Ihh and Dhh), and the developmental relevance of the SCUBE2-induced hedgehog signaling in mammals remain poorly understood. In this study, we first showed that as compared with SCUBE1 or SCUBE3, SCUBE2 is the most potent modulator of IHH signaling in vitro. In addition, gain and loss-of-function studies demonstrated that SCUBE2 exerted an osteogenic function by enhancing Ihh-stimulated osteoblast differentiation in the mouse mesenchymal progenitor cells. Consistent with these in vitro studies and the prominent roles of Ihh in coordinating skeletogenesis, genetic ablation of Scube2 (-/-) caused defective endochondral bone formation and impaired Ihh-mediated chondrocyte differentiation and proliferation as well as osteoblast differentiation of -/- bone-marrow mesenchymal stromal-cell cultures. Our data demonstrate that Scube2 plays a key regulatory role in Ihh-dependent endochondral bone formation.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Osteogenesis , Adaptor Proteins, Signal Transducing , Alkaline Phosphatase/metabolism , Animals , Calcium-Binding Proteins , Cell Differentiation , Cell Proliferation , Chondrocytes/pathology , Gene Targeting , Growth Plate/metabolism , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/deficiency , Ligands , Mice , Mice, Knockout , NIH 3T3 Cells , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Solubility
14.
EMBO J ; 34(3): 294-306, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25452496

ABSTRACT

Transmembrane guanylyl cyclases (GCs), with activity regulated by peptide ligands and/or calcium-binding proteins, are essential for various physiological and sensory processes. The mode of activation of the GC subtype GC-G, which is expressed in neurons of the Grueneberg ganglion that respond to cool temperatures, has been elusive. In searching for appropriate stimuli to activate GC-G, we found that its enzymatic activity is directly stimulated by cool temperatures. In this context, it was observed that dimerization/oligomerization of GC-G, a process generally considered as critical for enzymatic activity of GCs, is strongly enhanced by coolness. Moreover, heterologous expression of GC-G in cultured cells rendered these cells responsive to coolness; thus, the protein might be a sensor for cool temperatures. This concept is supported by the observation of substantially reduced coolness-induced response of Grueneberg ganglion neurons and coolness-evoked ultrasonic vocalization in GC-G-deficient mouse pups. GC-G may be a novel thermosensory protein with functional implications for the Grueneberg ganglion, a sensory organ responding to cool temperatures.


Subject(s)
Calcium-Binding Proteins/metabolism , Cold Temperature , Guanylate Cyclase/metabolism , Membrane Proteins/metabolism , Neurons/metabolism , Protein Multimerization/physiology , Animals , Calcium-Binding Proteins/genetics , Guanylate Cyclase/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Neurons/cytology
15.
Arterioscler Thromb Vasc Biol ; 34(7): 1390-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24833801

ABSTRACT

OBJECTIVE: Signal peptide-CUB-EGF domain-containing protein 1 (SCUBE1), a secreted and surface-exposed glycoprotein on activated platelets, promotes platelet-platelet interaction and supports platelet-matrix adhesion. Its plasma level is a biomarker of platelet activation in acute thrombotic diseases. However, the exact roles of plasma SCUBE1 in vivo remain undefined. APPROACH AND RESULTS: We generated new mutant (Δ) mice lacking the soluble but retaining the membrane-bound form of SCUBE1. Plasma SCUBE1-depleted Δ/Δ mice showed normal hematologic and coagulant features and expression of major platelet receptors, but Δ/Δ platelet-rich plasma showed impaired platelet aggregation in response to ADP and collagen treatment. The addition of purified recombinant SCUBE1 protein restored the aggregation of platelets in Δ/Δ platelet-rich plasma and further enhanced platelet aggregation in +/+ platelet-rich plasma. Plasma deficiency of SCUBE1 diminished arterial thrombosis in mice and protected against lethal thromboembolism induced by collagen-epinephrine treatment. Last, antibodies directed against the epidermal growth factor-like repeats of SCUBE1, which are involved in trans-homophilic protein-protein interactions, protected mice against fatal thromboembolism without causing bleeding in vivo. CONCLUSIONS: We conclude that plasma SCUBE1 participates in platelet aggregation by bridging adjacent activated platelets in thrombosis. Blockade of soluble SCUBE1 might represent a novel antithrombotic strategy.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Intercellular Signaling Peptides and Proteins/blood , Platelet Aggregation , Pulmonary Embolism/prevention & control , Thrombosis/prevention & control , Animals , Antibodies, Monoclonal/pharmacology , Blood Coagulation/drug effects , Blood Platelets/drug effects , Calcium-Binding Proteins , Cell Shape , Disease Models, Animal , Fibrinolytic Agents/pharmacology , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , Platelet Aggregation/drug effects , Protein Structure, Tertiary , Pulmonary Embolism/blood , Pulmonary Embolism/genetics , Signal Transduction , Thrombosis/blood , Thrombosis/genetics , Time Factors
16.
J Cell Sci ; 127(Pt 1): 85-100, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24213532

ABSTRACT

Signal peptide-CUB-EGF domain-containing protein 2 (SCUBE2) belongs to a secreted and membrane-associated multi-domain SCUBE protein family. We previously demonstrated that SCUBE2 is a novel breast-tumor suppressor and could be a useful prognostic marker. However, the role of SCUBE2 in breast-cancer cell migration and invasion and how it is regulated during the epithelial-mesenchymal transition (EMT) remain undefined. In this study, we showed that ectopic SCUBE2 overexpression could enhance the formation of E-cadherin-containing adherens junctions by ß-catenin-SOX-mediated induction of forkhead box A1 (a positive regulator of E-cadherin) and upregulation of E-cadherin, which in turn led to epithelial transition and inhibited migration and invasion of aggressive MDA-MB-231 breast-carcinoma cells. SCUBE2 expression was repressed together with that of E-cadherin in TGF-ß-induced EMT; direct expression of SCUBE2 alone was sufficient to inhibit the TGF-ß-induced EMT. Furthermore, quantitative DNA methylation, methylation-specific PCR, and chromatin immunoprecipitation analyses revealed that SCUBE2 expression was inactivated by DNA hypermethylation at the CpG islands by recruiting and binding DNA methyltransferase 1 during TGF-ß-induced EMT. Together, our results suggest that SCUBE2 plays a key role in suppressing breast-carcinoma-cell mobility and invasiveness by increasing the formation of the epithelial E-cadherin-containing adherens junctions to promote epithelial differentiation and drive the reversal of EMT.


Subject(s)
Adherens Junctions/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Adaptor Proteins, Signal Transducing , Adherens Junctions/drug effects , Cadherins/genetics , Cadherins/metabolism , Calcium-Binding Proteins , Cell Line, Tumor , Cell Movement/drug effects , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Epithelial-Mesenchymal Transition/drug effects , Female , Genes, Reporter , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Luciferases/genetics , Luciferases/metabolism , Membrane Proteins/metabolism , SOX Transcription Factors/genetics , SOX Transcription Factors/metabolism , Signal Transduction , Transforming Growth Factor beta/pharmacology , beta Catenin/genetics , beta Catenin/metabolism
17.
J Biol Chem ; 286(30): 27039-47, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21652720

ABSTRACT

Signal peptide CUB (complement proteins C1r/C1s, Uegf, and Bmp 1)-EGF domain-containing protein 2 (SCUBE2) is a secreted, membrane-associated multidomain protein composed of five recognizable motifs: an NH(2)-terminal signal peptide sequence, nine copies of epidermal growth factor (EGF)-like repeats, a spacer region, three cysteine-rich repeats, and one CUB domain at the COOH terminus. Our previous clinical study showed that SCUBE2 may act as a novel breast tumor suppressor gene and serve as a useful prognostic marker. However, the specific domain responsible for its tumor suppressor activity and the precise mechanisms of its anti-tumor effect remain unknown. Using a combination of biochemical, molecular, and cell biology techniques, we further dissected the molecular functions and signal pathways mediated by the NH(2)-terminal EGF-like repeats or COOH-terminal CUB domain of SCUBE2. Independent overexpression of the NH(2)-terminal EGF-like repeats or COOH-terminal CUB domain resulted in suppression of MCF-7 breast cancer cell proliferation and reduced MCF-7 xenograft tumor growth in nude mice. Molecular and biochemical analyses revealed that the COOH-terminal CUB domain could directly bind to and antagonize bone morphogenetic protein activity in an autocrine manner, whereas the NH(2)-terminal EGF-like repeats could mediate cell-cell homophilic adhesions in a calcium-dependent fashion, interact with E-cadherin (a master tumor suppressor), and decrease the ß-catenin signaling pathway. Together, our data demonstrate that SCUBE2 has growth inhibitory effects through a coordinated regulation of two distinct mechanisms: antagonizing bone morphogenetic protein and suppressing the ß-catenin pathway in breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Membrane Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Breast Neoplasms/genetics , Calcium-Binding Proteins , Cell Line, Tumor , Female , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Peptide Mapping , Protein Structure, Tertiary , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics , beta Catenin/genetics , beta Catenin/metabolism
18.
Biochem J ; 422(1): 119-28, 2009 Jul 29.
Article in English | MEDLINE | ID: mdl-19480626

ABSTRACT

SCUBE2 [signal peptide, CUB domain, EGF (epidermal growth factor)-like protein 2] belongs to an evolutionarily conserved SCUBE protein family, which possesses domain organization characteristic of an N-terminal signal peptide sequence followed by nine EGF-like repeats, a spacer region, three cysteine-rich repeat motifs, and one CUB domain at the C-terminus. Despite several genetic analyses suggesting that the zebrafish orthologue of the mammalian SCUBE2 gene participates in HH (Hedgehog) signalling, the complete full-length cDNA and biochemical function for mammalian SCUBE2 on HH signalling remains uninvestigated. In the present study, we isolated the full-length cDNA and studied the role of human SCUBE2 in the HH signalling cascade. When overexpressed, recombinant human SCUBE2 manifests as a secreted surface-anchored glycoprotein. Deletion mapping analysis defines the critical role of the spacer region and/or cysteine-rich repeats for membrane association. Further biochemical analyses and functional reporter assays demonstrated that human SCUBE2 can specifically interact with SHH (Sonic Hedgehog) and SHH receptor PTCH1 (Patched-1), and enhance the SHH signalling activity within the cholesterol-rich raft microdomains of the plasma membranes. Together, our results reveal that human SCUBE2 is a novel positive component of the HH signal, acting upstream of ligand binding at the plasma membrane. Thus human SCUBE2 could play important roles in HH-related biology and pathology, such as during organ development and tumour progression.


Subject(s)
Membrane Glycoproteins/isolation & purification , Membrane Glycoproteins/metabolism , Membrane Proteins/isolation & purification , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Calcium-Binding Proteins , Caveolin 1/metabolism , DNA, Complementary/isolation & purification , Extracellular Matrix Proteins/chemistry , Glycosylation , Hedgehog Proteins/metabolism , Humans , Membrane Microdomains/metabolism , Membrane Proteins/chemistry , Mice , Molecular Sequence Data , NIH 3T3 Cells , Open Reading Frames/genetics , Patched Receptors , Patched-1 Receptor , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Cell Surface/metabolism , Repetitive Sequences, Amino Acid , Sequence Alignment , Signal Transduction , Zebrafish Proteins/chemistry
19.
Cancer Res ; 69(8): 3634-41, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19369267

ABSTRACT

Signal peptide-CUB-epidermal growth factor-like domain-containing protein 2 (SCUBE2), originally identified from the endothelium and several nonendothelial primary cell types, was recently shown to be expressed in invasive breast carcinomas. However, the protein localization and biological significance of SCUBE2 in breast cancer are unknown. In this report, we show by anti-SCUBE2 immunostaining that SCUBE2 is mainly expressed in vascular endothelial and mammary ductal epithelial cells in normal breast tissue. In addition, we observed positive staining for SCUBE2 in 55% (86 of 156) of primary breast tumors. Patients with positive SCUBE2 protein-expressing tumors had better prognosis than those with negative SCUBE2 protein-expressing tumors in terms of disease-free survival. Multivariate analysis confirmed SCUBE2 protein expression as an independent prognostic factor for disease-free survival. Furthermore, overexpression of ectopic SCUBE2 protein resulted in suppression of MCF-7 breast cancer cell proliferation and reduced MCF-7 xenograft tumor growth in nude mice. Molecular and biochemical analyses revealed that the COOH terminal region of SCUBE2 directly bound to and antagonized bone morphogenetic protein activity. Together, our results show for the first time that altered SCUBE2 expression is important in breast cancer progression and SCUBE2 may serve as a useful prognostic marker.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Membrane Proteins/biosynthesis , Adaptor Proteins, Signal Transducing , Animals , Antibodies/chemistry , Antibodies/immunology , Antibody Specificity , Calcium-Binding Proteins , Cell Growth Processes/physiology , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Matrix Metalloproteinase 2/metabolism , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Transplantation, Heterologous
20.
J Agric Food Chem ; 55(21): 8523-33, 2007 Oct 17.
Article in English | MEDLINE | ID: mdl-17894456

ABSTRACT

Chronic cardiovascular and neurodegenerative complications induced by hyperglycemia have been considered to be associated most relevantly with endothelial cell damages (ECD). The protective effects of the aqueous extract of Psidium guajava L. budding leaves (PE) on the ECD in human umbilical vein endothelial cell (HUVEC) model were investigated. Results revealed that glyoxal (GO) and methylglyoxal (MGO) resulting from the glycative and autoxidative reactions of the high blood sugar glucose (G) evoked a huge production of ROS and NO, which in turn increased the production of peroxynitrite, combined with the activation of the nuclear factor kappaB (NFkappaB), leading to cell apoptosis. High plasma glucose activated p38-MAPK, and high GO increased the expressions of p38-MAPK and JNK-MAPK, whereas high MGO levels induced the activity of ERK-MAPK. Glucose and dicarbonyl compounds were all found to be good inducers of intracellular PKCs, which together with MAPK acted as the upstream triggering factor to activate NFkappaB. Conclusively, high plasma glucose together with dicarbonyl compounds can trigger the signaling pathways of MAPK and PKC and induce cell apoptosis through ROS and peroxynitrite stimulation and finally by NFkappaB activation. Such effects of PE were ascribed to its high plant polyphenolic (PPP) contents, the latter being potent ROS inhibitors capable of blocking the glycation of proteins, which otherwise could have brought forth severe detrimental effects to the cells.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/drug effects , Plant Extracts/pharmacology , Plant Leaves/chemistry , Psidium/chemistry , Cell Survival/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , L-Lactate Dehydrogenase/metabolism , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Nitric Oxide/antagonists & inhibitors , Protein Kinase C/antagonists & inhibitors , Reactive Oxygen Species/antagonists & inhibitors , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...