ARHGEF7 Antibody

Total Page:16

File Type:pdf, Size:1020Kb

ARHGEF7 Antibody Product Datasheet ARHGEF7 Antibody Catalog No: #AB32163 Orders: [email protected] Description Support: [email protected] Product Name ARHGEF7 Antibody Host Species Rabbit Clonality Polyclonal Purification Antibodies were purified by affinity purification using immunogen. Applications WB IHC Species Reactivity Hu Ms Rt Specificity The antibody detects endogenous level of total ARHGEF7 protein. Immunogen Type Recombinant Protein Immunogen Description Recombinant protein of human ARHGEF7. Target Name ARHGEF7 Other Names ARHGEF7; BETA-PIX; COOL1; DKFZp686C12170; DKFZp761K1021 Accession No. Swiss-Prot:Q14155NCBI Gene ID:8874 SDS-PAGE MW 90KD Concentration 1.0mg/ml Formulation Supplied at 1.0mg/mL in phosphate buffered saline (without Mg2+ and Ca2+), pH 7.4, 150mM NaCl, 0.02% sodium azide and 50% glycerol. Storage Store at -20°C Application Details Western blotting: 1:500 - 1:2000 Immunohistochemistry: 1:50 - 1:100 Images Western blot analysis of extracts of various cell lines, using ARHGEF7 antibody. Background Cool/Pix proteins comprise a family of guanine nucleotide exchange factors (GEFs) localized to focal adhesions. The family consists of two isoforms, cool2/αpix and cool1/βPix, the latter having two splice variants that vary in their carboxy termini (1). Cool1/βPix, like other GEFs, has a DH (Dbl Address: 8400 Baltimore Ave. Suite 302 College Park MD 20740 USA http://www.abscitech.com 1 homology) domain, which allows binding of small GTPases and GDP/GTP exchange, and a PH (Pleckstrin homology) domain, which is important in regulating subcellular localization. Cool1/βPix also has an SH3 domain, which binds to the PAK kinase, a downstream effector of cdc42 and Rac (3,4). Phosphorylation of cool1/βPix by PAK2 downstream of MAPK signaling alters the localization of a complex containing PAK2 and cool-1/βPix, regulating formation of growth cones in response to growth factors (4). Growth factor induced activation of Rac1 via cool1/βPix was later shown to occur independently of subcellular localization (5). Endothelin-1 stimulation of mesangial cells stimulates the protein kinase A (PKA) pathway, resulting in translocation of cool-1/βPix and activation of cdc42 (6). Note: This product is for in vitro research use only and is not intended for use in humans or animals. Address: 8400 Baltimore Ave. Suite 302 College Park MD 20740 USA http://www.abscitech.com 2.
Recommended publications
  • Regulation of Cdc42 and Its Effectors in Epithelial Morphogenesis Franck Pichaud1,2,*, Rhian F
    © 2019. Published by The Company of Biologists Ltd | Journal of Cell Science (2019) 132, jcs217869. doi:10.1242/jcs.217869 REVIEW SUBJECT COLLECTION: ADHESION Regulation of Cdc42 and its effectors in epithelial morphogenesis Franck Pichaud1,2,*, Rhian F. Walther1 and Francisca Nunes de Almeida1 ABSTRACT An overview of Cdc42 Cdc42 – a member of the small Rho GTPase family – regulates cell Cdc42 was discovered in yeast and belongs to a large family of small – polarity across organisms from yeast to humans. It is an essential (20 30 kDa) GTP-binding proteins (Adams et al., 1990; Johnson regulator of polarized morphogenesis in epithelial cells, through and Pringle, 1990). It is part of the Ras-homologous Rho subfamily coordination of apical membrane morphogenesis, lumen formation and of GTPases, of which there are 20 members in humans, including junction maturation. In parallel, work in yeast and Caenorhabditis elegans the RhoA and Rac GTPases, (Hall, 2012). Rho, Rac and Cdc42 has provided important clues as to how this molecular switch can homologues are found in all eukaryotes, except for plants, which do generate and regulate polarity through localized activation or inhibition, not have a clear homologue for Cdc42. Together, the function of and cytoskeleton regulation. Recent studies have revealed how Rho GTPases influences most, if not all, cellular processes. important and complex these regulations can be during epithelial In the early 1990s, seminal work from Alan Hall and his morphogenesis. This complexity is mirrored by the fact that Cdc42 can collaborators identified Rho, Rac and Cdc42 as main regulators of exert its function through many effector proteins.
    [Show full text]
  • ARHGEF7 (B-PIX) Is Required for the Maintenance of Podocyte Architecture and Glomerular Function
    BASIC RESEARCH www.jasn.org ARHGEF7 (b-PIX) Is Required for the Maintenance of Podocyte Architecture and Glomerular Function Jun Matsuda, Mirela Maier, Lamine Aoudjit, Cindy Baldwin, and Tomoko Takano Division of Nephrology, McGill University Health Centre, Montreal, Quebec, Canada ABSTRACT Background Previous studies showed that Cdc42, a member of the prototypical Rho family of small GTPases and a regulator of the actin cytoskeleton, is critical for the normal development and health of podocytes. However, upstream regulatory mechanisms for Cdc42 activity in podocytes are largely unknown. Methods We used a proximity-based ligation assay, BioID, to identify guanine nucleotide exchange fac- tors that activate Cdc42 in immortalized human podocytes. We generated podocyte-specificARHGEF7 (commonly known as b-PIX) knockout mice by crossing b-PIX floxed mice with Podocin-Cre mice. Using shRNA, we established cultured mouse podocytes with b-PIX knockdown and their controls. Results We identified b-PIX as a predominant guanine nucleotide exchange factor that interacts with Cdc42 in human podocytes. Podocyte-specific b-PIX knockout mice developed progressive proteinuria and kidney failure with global or segmental glomerulosclerosis in adulthood. Glomerular podocyte density gradually decreased in podocyte-specific b-PIX knockout mice, indicating podocyte loss. Compared with controls, glomeruli from podocyte-specific b-PIX knockout mice and cultured mouse podocytes with b-PIX knockdown exhibited significant reduction in Cdc42 activity. Loss of b-PIX promoted podocyte apoptosis, which was mediated by the reduced activity of the prosurvival transcriptional regulator Yes-associated protein. Conclusions These findings indicate that b-PIX is required for the maintenance of podocyte architecture and glomerular function via Cdc42 and its downstream Yes-associated protein activities.
    [Show full text]
  • Redefining the Specificity of Phosphoinositide-Binding by Human
    bioRxiv preprint doi: https://doi.org/10.1101/2020.06.20.163253; this version posted June 21, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. Redefining the specificity of phosphoinositide-binding by human PH domain-containing proteins Nilmani Singh1†, Adriana Reyes-Ordoñez1†, Michael A. Compagnone1, Jesus F. Moreno Castillo1, Benjamin J. Leslie2, Taekjip Ha2,3,4,5, Jie Chen1* 1Department of Cell & Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801; 2Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205; 3Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218; 4Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205; 5Howard Hughes Medical Institute, Baltimore, MD 21205, USA †These authors contributed equally to this work. *Correspondence: [email protected]. bioRxiv preprint doi: https://doi.org/10.1101/2020.06.20.163253; this version posted June 21, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC 4.0 International license. ABSTRACT Pleckstrin homology (PH) domains are presumed to bind phosphoinositides (PIPs), but specific interaction with and regulation by PIPs for most PH domain-containing proteins are unclear. Here we employed a single-molecule pulldown assay to study interactions of lipid vesicles with full-length proteins in mammalian whole cell lysates.
    [Show full text]
  • Cdc42 Mediates Cancer Cell Chemotaxis in Perineural Invasion
    Author Manuscript Published OnlineFirst on February 21, 2020; DOI: 10.1158/1541-7786.MCR-19-0726 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Cdc42 mediates cancer cell chemotaxis in perineural invasion ______________________________________________________________________ Natalya Chernichenko1, Tatiana Omelchenko2, Sylvie Deborde1, Richard Bakst3, Shizhi He1, Chun-Hao Chen1, Laxmi Gusain1, Efsevia Vakiani4, Nora Katabi4, Alan Hall2*, Richard J Wong1 ______________________________________________________________________ 1Department of Surgery, 2Cell Biology Program, 4Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, 10021 3Department of Radiation Oncology Mount Sinai Hospital, New York, NY, 10029 * Deceased The authors declare that they each do not have any conflict of interest with the material in this manuscript. Correspondence: Richard J. Wong, MD Memorial Sloan-Kettering Cancer Center 1275 York Avenue, C-1069 New York, NY 10021 Office: (212) 639-7638 FAX: (212) 717-3302 Email: [email protected] Downloaded from mcr.aacrjournals.org on October 1, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on February 21, 2020; DOI: 10.1158/1541-7786.MCR-19-0726 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 2 Abstract Perineural invasion (PNI) is an ominous form of cancer progression along nerves associated with poor clinical outcome. Glial derived neurotrophic factor (GDNF) interacts with cancer cell RET receptors to enable PNI, but downstream events remain undefined. We demonstrate that GDNF leads to early activation of the GTPase Cdc42 in pancreatic cancer cells, but only delayed activation of RhoA and does not affect Rac1. Depletion of Cdc42 impairs pancreatic cancer cell chemotaxis towards GDNF and nerves.
    [Show full text]
  • The Tumor Suppressor SCRIB Is a Negative Modulator of the Wnt/-Catenin Signaling Pathway
    The Tumor Suppressor SCRIB is a Negative Modulator of the Wnt/β-Catenin Signaling Pathway Avais Daulat, Mônica Silveira Wagner, Alexandra Walton, Emilie Baudelet, Stéphane Audebert, Luc Camoin, Jean-Paul Borg To cite this version: Avais Daulat, Mônica Silveira Wagner, Alexandra Walton, Emilie Baudelet, Stéphane Audebert, et al.. The Tumor Suppressor SCRIB is a Negative Modulator of the Wnt/β-Catenin Signaling Pathway. Pro- teomics, Wiley-VCH Verlag, 2019, 19 (21-22), pp.1800487. 10.1002/pmic.201800487. hal-02518664 HAL Id: hal-02518664 https://hal.archives-ouvertes.fr/hal-02518664 Submitted on 7 Apr 2020 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. PROTEOMICS Page 2 of 35 1 2 3 1 The tumor suppressor SCRIB is a negative modulator of the Wnt/-catenin signaling 4 5 6 2 pathway 7 8 3 Avais M. Daulat1,§, Mônica Silveira Wagner1,§, Alexandra Walton1, Emilie Baudelet2, 9 10 4 Stéphane Audebert2, Luc Camoin2, #,*, Jean-Paul Borg1,2,#,* 11 12 13 5 14 15 6 16 17 7 18 19 8 20 21 For Peer Review 1 22 9 Centre de Recherche en Cancérologie de Marseille, Equipe
    [Show full text]
  • Genome-Wide DNA Methylation Profiles in Community Members Exposed to the World Trade Center Disaster
    International Journal of Environmental Research and Public Health Article Genome-Wide DNA Methylation Profiles in Community Members Exposed to the World Trade Center Disaster Alan A. Arslan 1,2,3,* , Stephanie Tuminello 2, Lei Yang 2, Yian Zhang 2, Nedim Durmus 4, Matija Snuderl 5, Adriana Heguy 5,6, Anne Zeleniuch-Jacquotte 2,3, Yongzhao Shao 2,3 and Joan Reibman 4 1 Department of Obstetrics and Gynecology, New York University Langone Health, New York, NY 10016, USA 2 Department of Population Health, New York University Langone Health, New York, NY 10016, USA; [email protected] (S.T.); [email protected] (L.Y.); [email protected] (Y.Z.); [email protected] (A.Z.-J.); [email protected] (Y.S.) 3 NYU Perlmutter Comprehensive Cancer Center, New York, NY 10016, USA 4 Department of Medicine, New York University Langone Health, New York, NY 10016, USA; [email protected] (N.D.); [email protected] (J.R.) 5 Department of Pathology, New York University Langone Health, New York, NY 10016, USA; [email protected] (M.S.); [email protected] (A.H.) 6 NYU Langone’s Genome Technology Center, New York, NY 10016, USA * Correspondence: [email protected] Received: 30 June 2020; Accepted: 25 July 2020; Published: 30 July 2020 Abstract: The primary goal of this pilot study was to assess feasibility of studies among local community members to address the hypothesis that complex exposures to the World Trade Center (WTC) dust and fumes resulted in long-term epigenetic changes. We enrolled 18 WTC-exposed cancer-free women from the WTC Environmental Health Center (WTC EHC) who agreed to donate blood samples during their standard clinical visits.
    [Show full text]
  • Differential Expression of PAK3 in Cancers of the Breast
    Differential expression of p21 (RAC1) activated kinase 3 in cancers of the breast. Shahan Mamoor, MS1 [email protected] East Islip, NY 11739 Breast cancer affects women at relatively high frequency1. We mined published microarray datasets2,3 to determine in an unbiased fashion and at the systems level genes most differentially expressed in the primary tumors of patients with breast cancer. We report here significant differential expression of the gene encoding p21 (RAC1) activated kinase 3, PAK3, when comparing primary tumors of the breast to the tissue of origin, the normal breast. PAK3 was also differentially expressed in the tumor cells of patients with triple negative breast cancer. PAK3 mRNA was present at significantly lower quantities in tumors of the breast as compared to normal breast tissue. Analysis of human survival data revealed that expression of PAK3 in primary tumors of the breast was correlated with post-progression survival in patients with luminal B subtype cancer, demonstrating a relationship between primary tumor expression of a differentially expressed gene and patient survival outcomes influenced by molecular subtype. PAK3 may be of relevance to initiation, maintenance or progression of cancers of the female breast. Keywords: breast cancer, PAK3, p21 (RAC1) activated kinase 3, systems biology of breast cancer, targeted therapeutics in breast cancer. 1 Invasive breast cancer is diagnosed in over a quarter of a million women in the United States each year1 and in 2018, breast cancer was the leading cause of cancer death in women worldwide4. While patients with localized breast cancer are provided a 99% 5-year survival rate, patients with regional breast cancer, cancer that has spread to lymph nodes or nearby structures, are provided an 86% 5-year survival rate5,6.
    [Show full text]
  • Molecular Cytogenetic and Phenotypic Characterization of Ring Chromosome 13 in Three Unrelated Patients
    Journal of Pediatric Genetics 2 (2013) 147–155 147 DOI 10.3233/PGE-13063 IOS Press Case Report Molecular cytogenetic and phenotypic characterization of ring chromosome 13 in three unrelated patients Inesse B. Abdallah-Bouhjara,b,*, Soumaya Mougou-Zerellia,b, Hanene Hannachia,b, Abir Gmidènea, Audrey Labalmec, Najla Soyahd, Damien Sanlavillec, Ali Saada,b and Hatem Elghezala,b aDepartment of Cytogenetics and Reproductive Biology, Farhat Hached University Teaching Hospital, Sousse, Tunisia bCommon Service Units for Research in Genetics, Sousse Faculty of Medicine, Avenue Mohamed Karoui, Sousse, Tunisia cDepartment of Genetics, University Hospitals of Lyon, Lyon, France dDepartment of Pediatrics, Farhat Hached University Teaching Hospital, Sousse, Tunisia Received 7 July 2013 Revised 13 October 2013 Accepted 5 November 2013 Abstract. We report on the cytogenetic and molecular investigations of constitutional de-novo ring chromosome 13s in three unrelated patients for better understanding and delineation of the phenotypic variability characterizing this genomic rearrangement. The patient’s karyotypes were as follows: 46,XY,r(13)(p11q34) dn for patients 1 and 2 and 46,XY,r(13)(p11q14) dn for patient 3, as a result of the deletion in the telomeric regions of chromosome 13. The patients were, therefore, monosomic for the segment 13q34 → 13qter; in addition, for patient 3, the deletion was larger, encompassing the segment 13q14 → 13qter. Fluorescence in situ hybridi- zation confirmed these rearrangement and array CGH technique showed the loss of at least 2.9 Mb on the short arm and 4.7 Mb on the long arm of the chromosome 13 in patient 2. Ring chromosome 13 (r(13)) is associated with several phenotypic features like intellectual disability, marked short stature, brain and heart defects, microcephaly and genital malformations in males, including undescended testes and hypospadias.
    [Show full text]
  • The Human Gene Connectome As a Map of Short Cuts for Morbid Allele Discovery
    The human gene connectome as a map of short cuts for morbid allele discovery Yuval Itana,1, Shen-Ying Zhanga,b, Guillaume Vogta,b, Avinash Abhyankara, Melina Hermana, Patrick Nitschkec, Dror Friedd, Lluis Quintana-Murcie, Laurent Abela,b, and Jean-Laurent Casanovaa,b,f aSt. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY 10065; bLaboratory of Human Genetics of Infectious Diseases, Necker Branch, Paris Descartes University, Institut National de la Santé et de la Recherche Médicale U980, Necker Medical School, 75015 Paris, France; cPlateforme Bioinformatique, Université Paris Descartes, 75116 Paris, France; dDepartment of Computer Science, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel; eUnit of Human Evolutionary Genetics, Centre National de la Recherche Scientifique, Unité de Recherche Associée 3012, Institut Pasteur, F-75015 Paris, France; and fPediatric Immunology-Hematology Unit, Necker Hospital for Sick Children, 75015 Paris, France Edited* by Bruce Beutler, University of Texas Southwestern Medical Center, Dallas, TX, and approved February 15, 2013 (received for review October 19, 2012) High-throughput genomic data reveal thousands of gene variants to detect a single mutated gene, with the other polymorphic genes per patient, and it is often difficult to determine which of these being of less interest. This goes some way to explaining why, variants underlies disease in a given individual. However, at the despite the abundance of NGS data, the discovery of disease- population level, there may be some degree of phenotypic homo- causing alleles from such data remains somewhat limited. geneity, with alterations of specific physiological pathways under- We developed the human gene connectome (HGC) to over- come this problem.
    [Show full text]
  • CYK4 Inhibits Rac1-Dependent PAK1 and ARHGEF7 Effector Pathways During Cytokinesis
    Published Online: 3 September, 2012 | Supp Info: http://doi.org/10.1083/jcb.201204107 JCB: Article Downloaded from jcb.rupress.org on May 9, 2019 CYK4 inhibits Rac1-dependent PAK1 and ARHGEF7 effector pathways during cytokinesis Ricardo Nunes Bastos, Xenia Penate, Michelle Bates, Dean Hammond, and Francis A. Barr Department of Biochemistry, University of Oxford, Oxford OX1 3QU, England, UK n mitosis, animal cells lose their adhesion to the sur- CYK4 negatively regulated Rac1 activity at the cell equa- rounding surfaces and become rounded. During mi- tor in anaphase. Cells expressing a CYK4 GAP mutant I totic exit, they reestablish these adhesions and at the had defects in cytokinesis and showed elevated staining same time physically contract and divide. How these com- for the cell adhesion marker vinculin. These defects could peting processes are spatially segregated at the cell cortex be rescued by depletion of ARHGEF7 and p21-activated remains mysterious. To address this question, we define kinase, Rac1-specific effector proteins required for cell the specific effector pathways used by RhoA and Rac1 adhesion. Based on these findings, we propose that CYK4 in mitotic cells. We demonstrate that the MKlp1–CYK4 GAP activity is required during anaphase to inhibit Rac1- centralspindlin complex is a guanosine triphosphatase– dependent effector pathways associated with control of activating protein (GAP) for Rac1 and not RhoA and that cell spreading and adhesion. Introduction Dividing cells undergo dramatic changes in shape during Kimura et al., 2000; Yüce et al., 2005). ECT2 targets to the mitosis and cytokinesis (Glotzer, 2005; Barr and Gruneberg, midpoint of the dividing cells through interactions with the 2007).
    [Show full text]
  • Rho Family Gtpases and Rho Gefs in Glucose Homeostasis
    cells Review Rho Family GTPases and Rho GEFs in Glucose Homeostasis Polly A. Machin 1, Elpida Tsonou 1,2, David C. Hornigold 2 and Heidi C. E. Welch 1,* 1 Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; [email protected] (P.A.M.); [email protected] (E.T.) 2 Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK; [email protected] * Correspondence: [email protected]; Tel.: +44-(0)1223-496-596 Abstract: Dysregulation of glucose homeostasis leading to metabolic syndrome and type 2 diabetes is the cause of an increasing world health crisis. New intriguing roles have emerged for Rho family GTPases and their Rho guanine nucleotide exchange factor (GEF) activators in the regulation of glucose homeostasis. This review summates the current knowledge, focusing in particular on the roles of Rho GEFs in the processes of glucose-stimulated insulin secretion by pancreatic β cells and insulin-stimulated glucose uptake into skeletal muscle and adipose tissues. We discuss the ten Rho GEFs that are known so far to regulate glucose homeostasis, nine of which are in mammals, and one is in yeast. Among the mammalian Rho GEFs, P-Rex1, Vav2, Vav3, Tiam1, Kalirin and Plekhg4 were shown to mediate the insulin-stimulated translocation of the glucose transporter GLUT4 to the plasma membrane and/or insulin-stimulated glucose uptake in skeletal muscle or adipose tissue. The Rho GEFs P-Rex1, Vav2, Tiam1 and β-PIX were found to control the glucose-stimulated release of insulin by pancreatic β cells.
    [Show full text]
  • The Adaptor Protein APPL2 Controls Glucose-Stimulated Insulin Secretion Via F-Actin Remodeling in Pancreatic Β-Cells
    The adaptor protein APPL2 controls glucose-stimulated insulin secretion via F-actin remodeling in pancreatic β-cells Baile Wanga,b,1, Huige Linc,1, Xiaomu Lid, Wenqi Luc, Jae Bum Kime, Aimin Xua,b,f,2, and Kenneth K. Y. Chengc,2 aState Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China; bDepartment of Medicine, The University of Hong Kong, Hong Kong, China; cDepartment of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; dDepartment of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China; eDepartment of Biological Sciences, Institute of Molecular Biology and Genetics, Center for Adipose Tissue Remodeling, Seoul National University, Seoul 08826, South Korea; and fDepartment of Pharmacology & Pharmacy, The University of Hong Kong, Hong Kong, China Edited by Melanie H. Cobb, University of Texas Southwestern Medical Center, Dallas, TX, and approved September 28, 2020 (received for review August 19, 2020) Filamentous actin (F-actin) cytoskeletal remodeling is critical for have been observed in islet β-cells of diabetic mice and patients glucose-stimulated insulin secretion (GSIS) in pancreatic β-cells, with type 2 diabetes, respectively (3, 4). Early studies indicated and its dysregulation causes type 2 diabetes. The adaptor protein that cortical F-actin acts as a barrier to prevent fusion of the APPL1 promotes first-phase GSIS by up-regulating soluble N-ethyl- insulin granule with the plasma membrane under basal glucose maleimide-sensitive factor attachment protein receptor (SNARE) condition (<5 mM) (5). In response to high glucose stimulation, protein expression. However, whether APPL2 (a close homology F-actin is depolymerized to allow movement and fusion of in- of APPL1 with the same domain organization) plays a role in β-cell sulin granules for exocytosis (6).
    [Show full text]