Carboxypeptidase E (CPE) Inhibits the Secretion and Activity of Wnt3a

Total Page:16

File Type:pdf, Size:1020Kb

Carboxypeptidase E (CPE) Inhibits the Secretion and Activity of Wnt3a Oncogene (2016) 35, 6416–6428 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 0950-9232/16 www.nature.com/onc ORIGINAL ARTICLE Carboxypeptidase E (CPE) inhibits the secretion and activity of Wnt3a N Skalka1,4, M Caspi1,4, L Lahav-Ariel1, YP Loh2, K Hirschberg3 and R Rosin-Arbesfeld1 The Wnt pathway has essential roles in cell proliferation, cell fate determination and tumorigenesis by regulating the expression of a wide range of target genes. As a core signaling cascade, the canonical Wnt pathway is regulated at different levels by numerous proteins. We have previously shown that carboxypeptidase E (CPE) is a novel regulator of the canonical Wnt signaling pathway. Here, we show that CPE and the Wnt3a ligand are co-secreted from cells. We show that although the C'-terminal Lys residue of Wnt3a is critical for its activity and is important for the effect of CPE on the Wnt pathway, CPE does not execute its effect by removing this Wnt3a residue. Interestingly, CPE through its N'-terminal sequence, forms aggregates with Wnt3a and possible endoplasmic reticulum (ER) stress leading to its loss of function. Together, our current results provide a mechanistic insight into the way CPE regulates the canonical Wnt signaling pathway. Oncogene (2016) 35, 6416–6428; doi:10.1038/onc.2016.173; published online 4 July 2016 INTRODUCTION induced tumorigenicity, and may serve as a potential tool in the The Wnt/β-catenin signaling pathway has key roles in regulation design of novel colorectal cancer therapeutic strategies. of embryogenesis and adult homeostasis. However, aberrant constitutive activation of this pathway leads to uncontrolled cell proliferation, growth and survival, promoting the progression of RESULTS 1–3 various types of human cancers, especially colorectal cancer. In A novel Wnt3a fusion protein is secreted and active β the absence of a Wnt signal, the levels of -catenin, the key We have previously shown that full-length CPE downregulates the effector of the Wnt signaling pathway, are kept low by the β ‘β ’ Wnt/ -catenin signaling pathway and interacts with the Wnt3a -catenin destruction complex that contains, among others, the ligand both intracellularly and extracellularly.6 To further investi- tumor suppressor protein adenomatous polyposis coli (APC) and gate the relationship between CPE and Wnt3a, we have Axin. This complex promotes the phosphorylation of β-catenin at constructed two vectors encoding mWnt3a fused to the Cherry its N-terminal Ser/Thr residues by casein kinase 1a and glycogen protein either in its C' or N'-terminus (Cherry-Wnt3a and Wnt3a- synthase kinase 3β, marking it for ubiquitylation and subsequent Cherry, respectively). As shown in Figure 1a, Wnt3a-Cherry has a proteasomal degradation. Activation of the pathway by the Wnt ligand or by mutations in APC, Axin or β-catenin disrupts the similar expression pattern to that of the untagged Wnt3a degradation of the latter, resulting in cytoplasmic accumulation of expressed in L-Wnt3a cells. The reciprocal construct (Cherry- β-catenin that translocates into the nucleus where it associates Wnt3a) exhibited a more diffused pattern which resembles the with TCF transcription factors and upregulates the transcription of cherry protein cellular distribution (not shown), and western blot Wnt target genes. The Wnt pathway is tightly regulated, and analysis suggests that the cherry tag is cleaved off during protein uncontrolled changes in Wnt-induced transcription are thought to synthesis (Figure 1b). Both the Wnt3a-Cherry and the Wnt3a lie in the basis of tumorigenesis.1 Previously, we have found expressed from the Cherry-Wnt3a construct were secreted as seen that carboxypeptidase E (CPE), a pro-hormone processing exo- in Figure 1b (lower panel). To ensure that the different plasmids peptidase that cleaves its substrates’ C'-terminal Lysine or Arginine do not induce cell death and that total amount of proteins was residues,4,5 interacts with Wnt3a and decreases Wnt signaling loaded, the conditioned medium (CM) containing the secreted levels.6 In this study, we utilized N'-terminal and C'-terminal proteins was subjected to western blot analysis using an anti-RPL6 tagged Wnt3a fusion proteins and show that although the C'- antibody. In addition, to verify that the changes in constructs terminal Lys residue is crucial for Wnt3a's activity, CPE inhibits Wnt expression are not influenced by the position of the Cherry tag we signaling independently of its enzymatic activity and its ability to have reacted the same lysates to both anti-Wnt3a and anti-Cherry cleave the C'-terminal residue of Wnt3a. Our results further show antibodies (Supplementary Figure S1). Finally, we show that both that CPE reduces Wnt3a protein levels probably through overexpressed Wnt3a ligands are active although the Cherry- autophagy and that the N'-terminal region of CPE triggers the Wnt3a displays higher activity (Figure 1c). This finding could result aggregation of Wnt3a into insoluble cellular fractions. Taken from the Cherry tag being cleaved off as part of the signal peptide together, these results suggest that CPE has novel roles in Wnt- processing along the secretion route. In this scenario, the Wnt3a 1Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; 2Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA and 3Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel. Correspondence: Dr R Rosin-Arbesfeld, Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Ramat-Aviv, Tel-Aviv 69978, Israel. E-mail: [email protected] 4These authors contributed equally to this work. Received 26 November 2015; revised 26 February 2016; accepted 8 April 2016; published online 4 July 2016 Wnt signaling and carboxypeptidase E (CPE) N Skalka et al 6417 Figure 1. Wnt3a-Cherry is both secreted and active. (a) L-Wnt3a cells were transfected with Wnt3a-Cherry. Twenty-four hours later, the cells were fixed, permeabilized and stained with anti-Wnt3a antibodies. Cells were visualized by a confocal microscope. (b) HEK293T cells were transfected with the indicated vectors. Forty-eight hours later, medium was collected and cells were harvested. Lysates and cell media were subjected to SDS–PAGE analysis using anti-Wnt3a antibodies. Anti-actin (lysate) or anti-LRP6 (CM) was used as loading controls. Asterisks mark a degradation product. (c) HEK293T cells were co-transfected with the indicated vectors along with the TOPFLASH/FOPFLASH constructs. Forty-eight hours post transfection, cells were harvested and luciferase values were measured. Values are shown as mean ± s.d. from at least three independent experiments performed in triplicates. Asterisks denote statistical significance of cells transfected with the different Wnt expressing constructs as compared with control samples in an unpaired Student's t-test. *P ⩽ 0.05. **P ⩽ 0.008. C, cherry. ligand reverts to its native form and may be more active co-transfecting HEK293T cells. Figure 2c depicts the interaction compared to the tagged Wnt3a protein. between Wnt3a-Cherry and the full-length CPE protein. As expected, the truncated CPE (ΔN-CPE) did not bind Wnt3a 6 CPE binds and colocalizes with Wnt3a to the Golgi and vesicle-like (Figure 2c). The interaction between CPE and Wnt3a was also 6 structures shown in β-cells that abundantly express the CPE protein. The CPE is known to localize to the trans-Golgi network and secretory cells were transfected with Wnt3a-HA and subjected to immuno- vesicles.7,8 Thus, we examined the subcellular distribution of the precipitation (IP) using an anti-HA antibody (Figure 2d). Wnt3a-Cherry and the CPE-GFP proteins using both live-cell imaging and immunofluorescence. Our results show that the two The C'-terminal lysine of Wnt3a is critical for its signaling activity proteins are expressed in the Golgi apparatus and seem to CPE like other members of the CP family cleaves C'-terminal basic colocalize in secretory vesicles when overexpressed in COS-7 cells residues (Lysine or Arginine) in a broad spectrum of substrates.10 (Figure 2a and Supplementary Movie S1). Immunofluorescence Interestingly, it has been shown that carboxypeptidase Z (CPZ) experiments revealed that CPE and Wnt3a colocalize with the modulates Wnt signaling by removing the terminal arginine of the Golgi marker—Golgin-97 (Figure 2b). As β-catenin was also Wnt4 protein.11,12 As Wnt3a contains a Lysine residue at the reported to localize to the trans-Golgi network,9 we tested C'-terminal position, we decided to test whether this amino acid is whether CPE can direct β-catenin to the Golgi apparatus. important for the function of Wnt3a. Site-directed mutagenesis was However, our results show that the two proteins do not interact performed to construct a Wnt3a that either lacks its last lysine (Supplementary Figure S2A and not shown), and CPE does not (ΔK351) or contains a glutamic acid residue at this position (K351E). seem to sequester β-catenin toward a trans-Golgi network The mutated Wnt3a proteins show a similar expression pattern as localization (Supplementary Figure S2B). Next, we tested the the wild-type Wnt3a-Cherry, when expressed in COS-7 cells ability of Wnt3a-Cherry protein to interact with full-length CPE by (Figure 3a) and retain their ability to interact with full-length CPE © 2016 Macmillan Publishers Limited, part of Springer Nature. Oncogene (2016) 6416 – 6428 Wnt signaling and carboxypeptidase E (CPE) N Skalka et al 6418 Figure 2. CPE and Wnt3a colocalize and interact in vesicle-like structures. (a) COS-7 cells were co-transfected with CPE-GFP and Wnt3a-Cherry. Twenty-four hours later, the cells were fixed and images were taken using a confocal microscope. Enlargement shows vesicles containing Wnt3a and CPE proteins.
Recommended publications
  • 1 Evidence for Gliadin Antibodies As Causative Agents in Schizophrenia
    1 Evidence for gliadin antibodies as causative agents in schizophrenia. C.J.Carter PolygenicPathways, 20 Upper Maze Hill, Saint-Leonard’s on Sea, East Sussex, TN37 0LG [email protected] Tel: 0044 (0)1424 422201 I have no fax Abstract Antibodies to gliadin, a component of gluten, have frequently been reported in schizophrenia patients, and in some cases remission has been noted following the instigation of a gluten free diet. Gliadin is a highly immunogenic protein, and B cell epitopes along its entire immunogenic length are homologous to the products of numerous proteins relevant to schizophrenia (p = 0.012 to 3e-25). These include members of the DISC1 interactome, of glutamate, dopamine and neuregulin signalling networks, and of pathways involved in plasticity, dendritic growth or myelination. Antibodies to gliadin are likely to cross react with these key proteins, as has already been observed with synapsin 1 and calreticulin. Gliadin may thus be a causative agent in schizophrenia, under certain genetic and immunological conditions, producing its effects via antibody mediated knockdown of multiple proteins relevant to the disease process. Because of such homology, an autoimmune response may be sustained by the human antigens that resemble gliadin itself, a scenario supported by many reports of immune activation both in the brain and in lymphocytes in schizophrenia. Gluten free diets and removal of such antibodies may be of therapeutic benefit in certain cases of schizophrenia. 2 Introduction A number of studies from China, Norway, and the USA have reported the presence of gliadin antibodies in schizophrenia 1-5. Gliadin is a component of gluten, intolerance to which is implicated in coeliac disease 6.
    [Show full text]
  • Supplemental Materials Supplemental Table 1
    Electronic Supplementary Material (ESI) for RSC Advances. This journal is © The Royal Society of Chemistry 2016 Supplemental Materials Supplemental Table 1. The differentially expressed proteins from rat pancreas identified by proteomics (SAP vs. SO) No. Protein name Gene name ratio P value 1 Metallothionein Mt1m 3.35 6.34E-07 2 Neutrophil antibiotic peptide NP-2 Defa 3.3 8.39E-07 3 Ilf2 protein Ilf2 3.18 1.75E-06 4 Numb isoform o/o rCG 3.12 2.73E-06 5 Lysozyme Lyz2 3.01 5.63E-06 6 Glucagon Gcg 2.89 1.17E-05 7 Serine protease HTRA1 Htra1 2.75 2.97E-05 8 Alpha 2 macroglobulin cardiac isoform (Fragment) 2.75 2.97E-05 9 Myosin IF (Predicted) Myo1f 2.65 5.53E-05 10 Neuroendocrine secretory protein 55 Gnas 2.61 7.60E-05 11 Matrix metallopeptidase 8 Mmp8 2.57 9.47E-05 12 Protein Tnks1bp1 Tnks1bp1 2.53 1.22E-04 13 Alpha-parvin Parva 2.47 1.78E-04 14 C4b-binding protein alpha chain C4bpa 2.42 2.53E-04 15 Protein KTI12 homolog Kti12 2.41 2.74E-04 16 Protein Rab11fip5 Rab11fip5 2.41 2.84E-04 17 Protein Mcpt1l3 Mcpt1l3 2.33 4.43E-04 18 Phospholipase B-like 1 Plbd1 2.33 4.76E-04 Aldehyde dehydrogenase (NAD), cytosolic 19 2.32 4.93E-04 (Fragments) 20 Protein Dpy19l2 Dpy19l2 2.3 5.68E-04 21 Regenerating islet-derived 3 alpha, isoform CRA_a Reg3a 2.27 6.74E-04 22 60S acidic ribosomal protein P1 Rplp1 2.26 7.22E-04 23 Serum albumin Alb 2.25 7.98E-04 24 Ribonuclease 4 Rnase4 2.24 8.25E-04 25 Cct-5 protein (Fragment) Cct5 2.24 8.52E-04 26 Protein S100-A9 S100a9 2.22 9.71E-04 27 Creatine kinase M-type Ckm 2.21 1.00E-03 28 Protein Larp4b Larp4b 2.18 1.25E-03
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Carboxypeptidase E: a Negative Regulator of the Canonical Wnt Signaling Pathway
    Oncogene (2013) 32, 2836–2847 & 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13 www.nature.com/onc ORIGINAL ARTICLE Carboxypeptidase E: a negative regulator of the canonical Wnt signaling pathway N Skalka1, M Caspi1, E Caspi1, YP Loh2 and R Rosin-Arbesfeld1 Aberrant activation of the canonical Wnt signal transduction pathway is involved in many diseases including cancer and is especially implicated in the development and progression of colorectal cancer. The key effector protein of the canonical Wnt pathway is b-catenin, which functions with T-cell factor/lymphoid enhancer factor to activate expression of Wnt target genes. In this study, we used a new functional screen based on cell survival in the presence of cDNAs encoding proteins that activate the Wnt pathway thus identifying novel Wnt signaling components. Here we identify carboxypeptidase E (|CPE) and its splice variant, DN-CPE, as novel regulators of the Wnt pathway. We show that whereas DN-CPE activates the Wnt signal, the full-length CPE (F-CPE) protein is an inhibitor of Wnt/b-catenin signaling. F-CPE forms a complex with the Wnt3a ligand and the Frizzled receptor. Moreover, F-CPE disrupts disheveled-induced signalosomes that are important for transducing the Wnt signal and reduces b-catenin protein levels and activity. Taken together, our data indicate that F-CPE and DN-CPE regulate the canonical Wnt signaling pathway negatively and positively, respectively, and demonstrate that this screening approach can be a rapid means for isolation of novel Wnt signaling components. Oncogene (2013) 32, 2836–2847; doi:10.1038/onc.2012.308; published online 23 July 2012 Keywords: Wnt signaling; carboxypeptidase E (CPE); b-catenin; functional screen INTRODUCTION target genes.
    [Show full text]
  • Novel Pathways in Fatty-Acid Induced Apoptosis in the Pancreatic Beta-Cell
    Novel pathways in fatty-acid induced apoptosis in the pancreatic beta-cell by KRISTIN DANIELLE JEFFREY B.Sc, University of Ottawa, 2004 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF MASTER OF SCIENCE in THE FACULTY OF GRADUATE STUDIES (Physiology) University of British Columbia January 2007 ©Kristin Danielle Jeffrey, 2007 Abstract Pancreatic P-cell death is a critical event in the pathogenesis of all forms of diabetes. Type 2 diabetes is caused by the combination of acquired factors such as elevated circulating fatty acids as well as genetic factors. In this study, we show that the free fatty acid palmitate increases markers of endoplasmic stress and apoptosis in pancreatic P-cells. Carboxypeptidase E (CPE), an enzyme involved in the processing of insulin, was identified as the major down-regulated protein spot during palmitate-induced apoptosis using Cy-dye 2D gel proteomics in both the MIN6 P-cell line and human islets. Using MIN6 cells treated with 1.5 mM palmitate complexed to BSA (6:1), a significant decrease in total carboxypeptidase E protein was confirmed through Western blots. The decrease in CPE was seen in the presence of palmitate at both low and high glucose and was not affected in high glucose alone or with the ER-stress inducer, thapsigargin. Palmitate-induced changes in carboxypeptidase E were present after 2 hours, while CHOP, a marker of ER-stress, was not expressed until after 6 hours of incubation, suggesting that the decrease in CPE occurs before ER-stress. This finding, together with experiments using protein synthesis inhibitors and RT-PCR suggested that CPE was likely regulated at the post-translational level.
    [Show full text]
  • Downloaded from Bioscientifica.Com at 09/25/2021 06:33:18PM Via Free Access 666 LO’DRISCOLL, P GAMMELL and Others
    665 Phenotypic and global gene expression profile changes between low passage and high passage MIN-6 cells Lorraine O’Driscoll*, Patrick Gammell*, Eadaoin McKiernan, Eoin Ryan, Per Bendix Jeppesen1, Sweta Rani and Martin Clynes National Institute for Cellular Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland 1Department of Endocrinology and Metabolism C, Aarhus University Hospital, Aarhus Sygehus THG, DK-Aarhus C, Denmark (Requests for offprints should be addressed to L O’Driscoll; Email: [email protected]) *(L O’Driscoll and P Gammell contributed equally to this work) Abstract The long-term potential to routinely use replacement b bioinformatics and real-time PCR technologies. Long-term cells/islets as cell therapy for type 1 diabetes relies on our culture was found to be associated with many phenotypic ability to culture such cells/islets, in vitro, while maintaining changes, including changes in growth rate and cellular their functional status. Previous b cell studies, by ourselves morphology, as well as loss of GSIS. Microarray analyses and other researchers, have indicated that the glucose- indicate expression of many mRNAs, including many stimulated insulin secretion (GSIS) phenotype is relatively involved in regulated secretion, adhesion and proliferation, unstable, in long-term culture. This study aimed to to be significantly affected by passaging/ long-term culture. investigate phenotypic and gene expression changes associ- Loss/reduced levels, in high passage cells, of certain ated with this loss of GSIS, using the MIN-6 cell line as transcripts associated with the mature b cell, together with model. Phenotypic differences between MIN-6(L, low increased levels of neuron/glia-associated mRNAs, suggest passage) and MIN-6(H, high passage) were determined by that, with time in culture, MIN-6 cells may revert to an early ELISA (assessing GSIS and cellular (pro)insulin content), (possibly multi-potential), poorly differentiated, ‘precursor- proliferation assays, phase contrast light microscopy and like’ cell type.
    [Show full text]
  • Five Decades of Research on Opioid Peptides: Current Knowledge and Unanswered Questions
    Molecular Pharmacology Fast Forward. Published on June 2, 2020 as DOI: 10.1124/mol.120.119388 This article has not been copyedited and formatted. The final version may differ from this version. File name: Opioid peptides v45 Date: 5/28/20 Review for Mol Pharm Special Issue celebrating 50 years of INRC Five decades of research on opioid peptides: Current knowledge and unanswered questions Lloyd D. Fricker1, Elyssa B. Margolis2, Ivone Gomes3, Lakshmi A. Devi3 1Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; E-mail: [email protected] 2Department of Neurology, UCSF Weill Institute for Neurosciences, 675 Nelson Rising Lane, San Francisco, CA 94143, USA; E-mail: [email protected] 3Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, Annenberg Downloaded from Building, One Gustave L. Levy Place, New York, NY 10029, USA; E-mail: [email protected] Running Title: Opioid peptides molpharm.aspetjournals.org Contact info for corresponding author(s): Lloyd Fricker, Ph.D. Department of Molecular Pharmacology Albert Einstein College of Medicine 1300 Morris Park Ave Bronx, NY 10461 Office: 718-430-4225 FAX: 718-430-8922 at ASPET Journals on October 1, 2021 Email: [email protected] Footnotes: The writing of the manuscript was funded in part by NIH grants DA008863 and NS026880 (to LAD) and AA026609 (to EBM). List of nonstandard abbreviations: ACTH Adrenocorticotrophic hormone AgRP Agouti-related peptide (AgRP) α-MSH Alpha-melanocyte stimulating hormone CART Cocaine- and amphetamine-regulated transcript CLIP Corticotropin-like intermediate lobe peptide DAMGO D-Ala2, N-MePhe4, Gly-ol]-enkephalin DOR Delta opioid receptor DPDPE [D-Pen2,D- Pen5]-enkephalin KOR Kappa opioid receptor MOR Mu opioid receptor PDYN Prodynorphin PENK Proenkephalin PET Positron-emission tomography PNOC Pronociceptin POMC Proopiomelanocortin 1 Molecular Pharmacology Fast Forward.
    [Show full text]
  • Supplemental Table 1
    Symbol Gene name MIN6.EXO MIN6.M1 MIN6.M2 MIN6.M3 MIN6.M4 A2m alpha-2-macroglobulin A2m Acat1 acetyl-Coenzyme A acetyltransferase 1 Acat1 Acly ATP citrate lyase Acly Acly Acly Act Actin Act Act Act Act Aga aspartylglucosaminidase Aga Ahcy S-adenosylhomocysteine hydrolase Ahcy Alb Albumin Alb Alb Alb Aldoa aldolase A, fructose-bisphosphate Aldoa Anxa5 Annexin A5 Anxa5 AP1 Adaptor-related protein complex AP1 AP2 Adaptor protein complex AP2 Arf1 ADP-ribosylation factor 1 Arf1 Atp1a1 ATPase Na/K transpoting Atp1a1 ATP1b1 Na/K ATPase beta subunit ATP1b1 ATP6V1 ATPase, H+ transporting.. ATP6V1 ATP6v1 ATP6v1 Banf1 Barrier to autointegration factor Banf1 Basp1 brain abundant, memrane signal protein 1 Basp1 C3 complement C3 C3 C3 C3 C4 Complement C4 C4 C4 C4 Calm2 calmodulin 2 (phosphorylase kinase, delta) Calm2 Capn5 Calpain 5 Capn5 Capn5 Cct5 chaperonin subunit 5 Cct5 Cct8 chaperonin subunit 8 Cct8 CD147 basigin CD147 CD63 CD63 CD63 CD81 CD81 CD81 CD81 CD81 CD81 CD81 CD82 CD82 CD82 CD82 CD90.2 thy1.2 CD90.2 CD98 Slc3a2 CD98 CD98 Cdc42 Cell division cycle 42 Cdc42 Cfl1 Cofilin 1 Cfl1 Cfl1 Chmp4b chromatin modifying protein 4B Chmp4b Chmp5 chromatin modifying protein 5 Chmp5 Clta clathrin, light polypeptide A Clta Cltc Clathrin Hc Cltc Cltc Cltc Cltc Clu clusterin Clu Col16a1 collagen 16a1 Col16a1 Col2 Collagen type II Col2a1 Col2 Col6 Collagen type VI alpha 3 Col6a3 Col6 CpE carboxypeptidase E CpE CpE CpE, CpH CpE CpE Cspg4 Chondroitin sulfate proteoglycan 4 Cspg4 CyCAP Cyclophilin C-associated protein CyCAP CyCAP Dnpep aspartyl aminopeptidase Dnpep Dstn destrin Dstn EDIL3 EGF-like repeat discoidin.
    [Show full text]
  • Proteolytic Cleavage—Mechanisms, Function
    Review Cite This: Chem. Rev. 2018, 118, 1137−1168 pubs.acs.org/CR Proteolytic CleavageMechanisms, Function, and “Omic” Approaches for a Near-Ubiquitous Posttranslational Modification Theo Klein,†,⊥ Ulrich Eckhard,†,§ Antoine Dufour,†,¶ Nestor Solis,† and Christopher M. Overall*,†,‡ † ‡ Life Sciences Institute, Department of Oral Biological and Medical Sciences, and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada ABSTRACT: Proteases enzymatically hydrolyze peptide bonds in substrate proteins, resulting in a widespread, irreversible posttranslational modification of the protein’s structure and biological function. Often regarded as a mere degradative mechanism in destruction of proteins or turnover in maintaining physiological homeostasis, recent research in the field of degradomics has led to the recognition of two main yet unexpected concepts. First, that targeted, limited proteolytic cleavage events by a wide repertoire of proteases are pivotal regulators of most, if not all, physiological and pathological processes. Second, an unexpected in vivo abundance of stable cleaved proteins revealed pervasive, functionally relevant protein processing in normal and diseased tissuefrom 40 to 70% of proteins also occur in vivo as distinct stable proteoforms with undocumented N- or C- termini, meaning these proteoforms are stable functional cleavage products, most with unknown functional implications. In this Review, we discuss the structural biology aspects and mechanisms
    [Show full text]
  • And Insulin Signaling
    molecules Review The Antidiabetic Mechanisms of Polyphenols Related to Increased Glucagon-Like Peptide-1 (GLP1) and Insulin Signaling J. Abraham Domínguez Avila 1, Joaquín Rodrigo García 2, Gustavo A. González Aguilar 1 and Laura A. de la Rosa 3,* 1 Coordinación de Tecnología de Alimentos de Origen Vegetal, Centro de Investigación en Alimentación y Desarrollo A. C., Carretera a La Victoria km 0.6, AP 1735, Hermosillo 83304, Sonora, Mexico; [email protected], (J.A.D.A.); [email protected], (G.A.G.A.) 2 Departamento de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo Envolvente del PRONAF y Estocolmo s/n, Ciudad Juárez 32310, Chihuahua, Mexico; [email protected] 3 Departamento de Ciencias Químico-Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Anillo Envolvente del PRONAF y Estocolmo s/n, Ciudad Juárez 32310, Chihuahua, Mexico * Correspondence: [email protected]; Tel.: + 52-656-688-1800 (ext. 1563) Academic Editors: Solomon Habtemariam and Giovanni Lentini Received: 4 May 2017; Accepted: 26 May 2017; Published: 30 May 2017 Abstract: Type-2 diabetes mellitus (T2DM) is an endocrine disease related to impaired/absent insulin signaling. Dietary habits can either promote or mitigate the onset and severity of T2DM. Diets rich in fruits and vegetables have been correlated with a decreased incidence of T2DM, apparently due to their high polyphenol content. Polyphenols are compounds of plant origin with several documented bioactivities related to health promotion. The present review describes the antidiabetic effects of polyphenols, specifically related to the secretion and effects of insulin and glucagon-like peptide 1 (GLP1), an enteric hormone that stimulates postprandial insulin secretion.
    [Show full text]
  • Carboxypeptidase E Cytoplasmic Tail Mediates Localization of Synaptic
    JOURNAL OF NEUROCHEMISTRY | 2010 | 114 | 886–896 doi: 10.1111/j.1471-4159.2010.06820.x ,1 , ,1 *Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA Department of Neurosciences, University of Toledo, School of Medicine, Toledo, Ohio, USA àDepartment of Pharmacology, University of Michigan, School of Medicine, Ann Arbor, Michigan, USA Abstract tion. Yeast two-hybrid and pull-down experiments showed that How synaptic vesicles (SVs) are localized to the pre-active the CPE cytoplasmic tail interacted with c-adducin, which zone (5–200 nm beneath the active zone) in the nerve ter- binds actin enriched at the nerve terminal. Total internal minal, which may represent the slow response SV pool, is not reflective fluorescence (TIRF) microscopy using PC12 cells as fully understood. Electron microscopy revealed the number of a model showed that expression of GFP-CPEC15 reduced the SVs located in the pre-active zone, was significantly de- steady-state level of synaptophysin-mRFP containing synap- creased in hypothalamic neurons of carboxypeptidase E tic-like microvesicles accumulated in the area within 200 nm knockout (CPE-KO) mice compared with wild-type mice. from the sub-plasma membrane (TIRF zone). Our findings Additionally, we found K+-stimulated glutamate secretion from identify the CPE cytoplasmic tail, as a new mediator for the hypothalamic embryonic neurons was impaired in CPE-KO localization of SVs in the actin-rich pre-active zone in hypo- mice. Biochemical studies indicate that SVs from the hypo- thalamic neurons and the TIRF zone of PC12 cells.
    [Show full text]
  • Fibroblasts from the Human Skin Dermo-Hypodermal Junction Are
    cells Article Fibroblasts from the Human Skin Dermo-Hypodermal Junction are Distinct from Dermal Papillary and Reticular Fibroblasts and from Mesenchymal Stem Cells and Exhibit a Specific Molecular Profile Related to Extracellular Matrix Organization and Modeling Valérie Haydont 1,*, Véronique Neiveyans 1, Philippe Perez 1, Élodie Busson 2, 2 1, 3,4,5,6, , Jean-Jacques Lataillade , Daniel Asselineau y and Nicolas O. Fortunel y * 1 Advanced Research, L’Oréal Research and Innovation, 93600 Aulnay-sous-Bois, France; [email protected] (V.N.); [email protected] (P.P.); [email protected] (D.A.) 2 Department of Medical and Surgical Assistance to the Armed Forces, French Forces Biomedical Research Institute (IRBA), 91223 CEDEX Brétigny sur Orge, France; [email protected] (É.B.); [email protected] (J.-J.L.) 3 Laboratoire de Génomique et Radiobiologie de la Kératinopoïèse, Institut de Biologie François Jacob, CEA/DRF/IRCM, 91000 Evry, France 4 INSERM U967, 92260 Fontenay-aux-Roses, France 5 Université Paris-Diderot, 75013 Paris 7, France 6 Université Paris-Saclay, 78140 Paris 11, France * Correspondence: [email protected] (V.H.); [email protected] (N.O.F.); Tel.: +33-1-48-68-96-00 (V.H.); +33-1-60-87-34-92 or +33-1-60-87-34-98 (N.O.F.) These authors contributed equally to the work. y Received: 15 December 2019; Accepted: 24 January 2020; Published: 5 February 2020 Abstract: Human skin dermis contains fibroblast subpopulations in which characterization is crucial due to their roles in extracellular matrix (ECM) biology.
    [Show full text]