I Differential Binding and Structural Properties of the WWC Protein

Total Page:16

File Type:pdf, Size:1020Kb

I Differential Binding and Structural Properties of the WWC Protein Differential Binding and Structural Properties of the WWC Protein Family by Diego Javier Rodriguez A THESIS Submitted to Oregon State University Honors College in partial fulfillment of the requirements for the degrees of Honors Baccalaureate of Science in Biochemistry and Molecular Biology Honors Baccalaureate of Arts in Psychology (Honors Scholar) Presented June 4, 2020 Commencement June 2020 i AN ABSTRACT OF THE THESIS OF Diego Javier Rodriguez for the degrees of Honors Baccalaureate of Science in Biochemistry and Molecular Biology and Honors Baccalaureate of Arts in Psychology presented on June 4, 2020. Title: Differential Binding and Structural Properties of the WWC Protein Family Abstract approved: ____________________________________________________________ Afua Nyarko The WWC family consists of three proteins‐ KIBRA/WWC1, WWC2, and WWC3. The three proteins share a similar primary structure and interact with a common set of proteins. Each paralog contains two WW domains which recognize proline‐rich sequences of the type PPxY (P is proline, Y is tyrosine, x is any amino acid). WW domains adopt a three‐stranded antiparallel beta sheet fold, but the WWC proteins are “atypical” in that, the second WW domain (WW2) is unfolded. The WWC proteins are implicated in regulating cell proliferation, organ growth, cell migration, synaptic signaling and generally thought to perform overlapping functions in most tissues. However, in recent years, several studies point to unique functions for each paralog. This study presents data that suggest that the unique functions arise from differences in binding mechanisms. Constructs of the WW domains of KIBRA and WWC3 and the PPxY segments of four putative binding partners, Large tumor suppressor 1 (LATS1), Angiomotin-like 1 (AMOTL1), DENDRIN, and Synaptopodin (SYNPO) were cloned and over-expressed in E.coli. Binding affinities and binding-induced structural changes were determined by isothermal titration calorimetry (ITC), fluorescence spectroscopy (FS) and nuclear magnetic resonance spectroscopy (NMR). The ITC data show that the LATS1, DENDRIN, and AMOTL1 polypeptides bind the KIBRA WW domains with a higher affinity than the WWC3 WW domains. The FS and NMR data further show that the binding of LATS1 and SYNPO to KIBRA and WWC3 does not induce ii folding of the WW2 domain, however, mutant LATS1 and SYNPO constructs, designed to mimic the short linker between the DENDRIN PPxY sites, induced folding of the KIBRA and WWC3 WW2 domains. The results suggest that differences in binding affinities for partner proteins and/or binding-induced folding may contribute to the unique functions of the WWC proteins. Key Words: WWC family, WW domains, PPxY motif Corresponding e-mail address: [email protected] iii ©Copyright by Diego Javier Rodriguez June 4, 2020 iv Differential Binding and Structural Properties of the WWC Protein Family by Diego Javier Rodriguez A THESIS Submitted to Oregon State University Honors College in partial fulfillment of the requirements for the degrees of Honors Baccalaureate of Science in Biochemistry and Molecular Biology Honors Baccalaureate of Arts in Psychology (Honors Scholar) Presented June 4, 2020 Commencement June 2020 v Honors Baccalaureate of Science in Biochemistry and Molecular Biology project of Diego Javier Rodriguez presented on June 4, 2020 APPROVED: Afua Nyarko, Mentor, representing Department of Biochemistry and Biophysics Elisar Barbar, Committee Member, representing Department of Biochemistry and Biophysics Ethiene Kwok, Committee Member, representing Department of Biochemistry and Biophysics Patrick Reardon, Committee Member, representing Department of Biochemistry and Biophysics Toni Doolen, Dean, Oregon State University Honors College I understand that my project will become part of the permanent collection of Oregon State University, Honors College. My signature below authorizes release of my project to any reader upon request. Diego Javier Rodriguez, Author vi ACKNOWLEDGEMENT My development as a scientist and my work in this thesis, I owe to Dr. Nyarko and the Nyarko lab, for mentoring me and providing me a space to learn. Every member of the Nyarko Lab took a part of my development. Including the mentorship of Afua and the countless hours we spent working on my projects, posters, discussions, troubleshooting, and this thesis. The patient guidance of Kasie Baker, who was my first introduction into wet lab work, has been immeasurably important to this thesis and my development as a scientist. Not only did she aid me by running, collecting, and processing all the NMR data presented in this thesis, Kasie guided me through protein purification and how to conduct many biophysical experiments. Ethiene ‘Kathy’ Kwok also played an integral part of my development and guided me through the molecular cloning and additional biophysical techniques. And, Amber Vogel was always around for any help I needed, advice, and countless discussions on our projects- past, present, and future. In addition to the Nyarko Lab, the Dept. of Biophysics and Biochemistry provided immeasurable support. Whether, through the tireless work of my advisor Dr. Kari Van Zee or the many ways the department leadership, including Drs. Elisar Barbar, Andy Karplus, and Michael Freitag, helped me succeed. In addition, funding from the Cripp’s Undergraduate Experience Award, the Undergraduate Research and Scholarship Award, and the Honor’s College DeLoach Work Scholarship helped support me and my work presented in this thesis. I would also like to thank my parents for always giving me something when they had nothing. This work and my education would have never happened if it were not for them. Lastly, I want to thank my family and friends who helped support me, made me laugh, and motivated me to always work hard. i Table of Contents List of Figures ................................................................................................................................ iv List of Tables .................................................................................................................................. v List of Abbreviations ..................................................................................................................... vi 1. INTRODUCTION ................................................................................................................... 1 1.1. Functions of the WWC protein family ............................................................................. 1 KIBRA: .................................................................................................................................... 1 WWC2: .................................................................................................................................... 2 WWC3: .................................................................................................................................... 2 1.2. Structure of the WWC protein family .............................................................................. 3 1.3. PPxY-containing binding partners of the WWC proteins ................................................ 5 Dendrin: ................................................................................................................................... 5 Synaptopodin: .......................................................................................................................... 6 Large associated tumor suppressor 1: ...................................................................................... 6 Angiomotin-like 1: .................................................................................................................. 7 2. EXPERIMENTAL DESIGN ................................................................................................... 8 2.1. Cloning of Constructs....................................................................................................... 8 2.2. Recombinant Protein Production ..................................................................................... 9 2.3. Isothermal Titration Calorimetry ................................................................................... 10 2.4. Fluorescence Spectroscopy ............................................................................................ 10 2.5. Circular Dichroism ......................................................................................................... 10 2.6. Nuclear Magnetic Resonance Spectroscopy .................................................................. 11 2.7. X-Ray Crystallography Protein Preparation and Screening ........................................... 11 3. RESULTS .............................................................................................................................. 12 3.1. Design and characterization of constructs ...................................................................... 12 3.2. ITC studies of the WWCs-PPxY partner interactions .................................................... 13 3.3. ITC binding studies of LATS1 and SYNPO mutants .................................................... 14 3.4. Binding-induced folding of the KIBRA WW domains .................................................. 15 3.5. Binding-induced folding of WWC3 WW domains ........................................................ 18 4. DISCUSSION ........................................................................................................................ 19 4.1. KIBRA and
Recommended publications
  • PARSANA-DISSERTATION-2020.Pdf
    DECIPHERING TRANSCRIPTIONAL PATTERNS OF GENE REGULATION: A COMPUTATIONAL APPROACH by Princy Parsana A dissertation submitted to The Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, Maryland July, 2020 © 2020 Princy Parsana All rights reserved Abstract With rapid advancements in sequencing technology, we now have the ability to sequence the entire human genome, and to quantify expression of tens of thousands of genes from hundreds of individuals. This provides an extraordinary opportunity to learn phenotype relevant genomic patterns that can improve our understanding of molecular and cellular processes underlying a trait. The high dimensional nature of genomic data presents a range of computational and statistical challenges. This dissertation presents a compilation of projects that were driven by the motivation to efficiently capture gene regulatory patterns in the human transcriptome, while addressing statistical and computational challenges that accompany this data. We attempt to address two major difficulties in this domain: a) artifacts and noise in transcriptomic data, andb) limited statistical power. First, we present our work on investigating the effect of artifactual variation in gene expression data and its impact on trans-eQTL discovery. Here we performed an in-depth analysis of diverse pre-recorded covariates and latent confounders to understand their contribution to heterogeneity in gene expression measurements. Next, we discovered 673 trans-eQTLs across 16 human tissues using v6 data from the Genotype Tissue Expression (GTEx) project. Finally, we characterized two trait-associated trans-eQTLs; one in Skeletal Muscle and another in Thyroid. Second, we present a principal component based residualization method to correct gene expression measurements prior to reconstruction of co-expression networks.
    [Show full text]
  • Analysis of Trans Esnps Infers Regulatory Network Architecture
    Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer Submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the Graduate School of Arts and Sciences COLUMBIA UNIVERSITY 2014 © 2014 Anat Kreimer All rights reserved ABSTRACT Analysis of trans eSNPs infers regulatory network architecture Anat Kreimer eSNPs are genetic variants associated with transcript expression levels. The characteristics of such variants highlight their importance and present a unique opportunity for studying gene regulation. eSNPs affect most genes and their cell type specificity can shed light on different processes that are activated in each cell. They can identify functional variants by connecting SNPs that are implicated in disease to a molecular mechanism. Examining eSNPs that are associated with distal genes can provide insights regarding the inference of regulatory networks but also presents challenges due to the high statistical burden of multiple testing. Such association studies allow: simultaneous investigation of many gene expression phenotypes without assuming any prior knowledge and identification of unknown regulators of gene expression while uncovering directionality. This thesis will focus on such distal eSNPs to map regulatory interactions between different loci and expose the architecture of the regulatory network defined by such interactions. We develop novel computational approaches and apply them to genetics-genomics data in human. We go beyond pairwise interactions to define network motifs, including regulatory modules and bi-fan structures, showing them to be prevalent in real data and exposing distinct attributes of such arrangements. We project eSNP associations onto a protein-protein interaction network to expose topological properties of eSNPs and their targets and highlight different modes of distal regulation.
    [Show full text]
  • Hippo Signaling in Cancer: Lessons from Drosophila Models
    fcell-07-00085 May 22, 2019 Time: 18:12 # 1 REVIEW published: 24 May 2019 doi: 10.3389/fcell.2019.00085 Hippo Signaling in Cancer: Lessons From Drosophila Models Kirti Snigdha1, Karishma Sanjay Gangwani1, Gauri Vijay Lapalikar2, Amit Singh1,3,4,5 and Madhuri Kango-Singh1,3,4,5* 1 Department of Biology, University of Dayton, Dayton, OH, United States, 2 Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, United States, 3 Pre-Medical Programs, University of Dayton, Dayton, OH, United States, 4 Center for Tissue Regeneration and Engineering at Dayton, University of Dayton, Dayton, OH, United States, 5 Integrated Science and Engineering Center, University of Dayton, Dayton, OH, United States Hippo pathway was initially identified through genetic screens for genes regulating organ size in fruitflies. Recent studies have highlighted the role of Hippo signaling as a key regulator of homeostasis, and in tumorigenesis. Hippo pathway is comprised of genes that act as tumor suppressor genes like hippo (hpo) and warts (wts), and oncogenes like yorkie (yki). YAP and TAZ are two related mammalian homologs of Drosophila Yki that act as effectors of the Hippo pathway. Hippo signaling deficiency can cause Edited by: YAP- or TAZ-dependent oncogene addiction for cancer cells. YAP and TAZ are often SrinivasVinod Saladi, activated in human malignant cancers. These transcriptional regulators may initiate Harvard Medical School, tumorigenic changes in solid tumors by inducing cancer stem cells and proliferation, United States culminating in metastasis and chemo-resistance. Given the complex mechanisms (e.g., Reviewed by: Sirisha M. Cheedipudi, of the cancer microenvironment, and the extrinsic and intrinsic cues) that overpower University of Texas Health Science YAP/TAZ inhibition, the molecular roles of the Hippo pathway in tumor growth and Center at Houston, United States Rizaldy Paz Scott, progression remain poorly defined.
    [Show full text]
  • Systematic Identification of Recognition Motifs for the Hub Protein
    Published Online: 2 July, 2019 | Supp Info: http://doi.org/10.26508/lsa.201900366 Downloaded from life-science-alliance.org on 25 September, 2021 Research Article Systematic identification of recognition motifs for the hub protein LC8 Nathan Jespersen1 , Aidan Estelle1, Nathan Waugh1 , Norman E Davey2, Cecilia Blikstad3 , York-Christoph Ammon4, Anna Akhmanova4, Ylva Ivarsson3, David A Hendrix1,5, Elisar Barbar1 Hub proteins participate in cellular regulation by dynamic binding of extensively studied, including the dynein intermediate chain (IC) multiple proteins within interaction networks. The hub protein LC8 (Makokha et al, 2002; Benison et al, 2006; Nyarko & Barbar, 2011) and reversibly interacts with more than 100 partners through a flexible the transcription factor ASCIZ (Jurado et al, 2012; Zaytseva et al, 2014; pocket at its dimer interface. To explore the diversity of the LC8 Clark et al, 2018). In addition, expression patterns show that LC8 is partner pool, we screened for LC8 binding partners using a proteomic highly expressed across a wide variety of cell types (Petryszak et al, phage display library composed of peptides from the human pro- 2016) and is broadly distributed within individual cells (Chen et al, teome, which had no bias toward a known LC8 motif. Of the identified 2009; Wang et al, 2016). hits, we validated binding of 29 peptides using isothermal titration LC8 is an 89–amino acid homodimeric protein first identified as a calorimetry.Ofthe29peptides,19were entirely novel, and all had the subunit of the dynein motor complex. Colocalization and binding canonical TQT motif anchor. A striking observation is that numerous studies with dynein led to a common perception that LC8 functions peptides containing the TQT anchor do not bind LC8, indicating that as a dynein “cargo adaptor” to facilitate transport of dynein cargo residues outside of the anchor facilitate LC8 interactions.
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Protein Identities in Evs Isolated from U87-MG GBM Cells As Determined by NG LC-MS/MS
    Protein identities in EVs isolated from U87-MG GBM cells as determined by NG LC-MS/MS. No. Accession Description Σ Coverage Σ# Proteins Σ# Unique Peptides Σ# Peptides Σ# PSMs # AAs MW [kDa] calc. pI 1 A8MS94 Putative golgin subfamily A member 2-like protein 5 OS=Homo sapiens PE=5 SV=2 - [GG2L5_HUMAN] 100 1 1 7 88 110 12,03704523 5,681152344 2 P60660 Myosin light polypeptide 6 OS=Homo sapiens GN=MYL6 PE=1 SV=2 - [MYL6_HUMAN] 100 3 5 17 173 151 16,91913397 4,652832031 3 Q6ZYL4 General transcription factor IIH subunit 5 OS=Homo sapiens GN=GTF2H5 PE=1 SV=1 - [TF2H5_HUMAN] 98,59 1 1 4 13 71 8,048185945 4,652832031 4 P60709 Actin, cytoplasmic 1 OS=Homo sapiens GN=ACTB PE=1 SV=1 - [ACTB_HUMAN] 97,6 5 5 35 917 375 41,70973209 5,478027344 5 P13489 Ribonuclease inhibitor OS=Homo sapiens GN=RNH1 PE=1 SV=2 - [RINI_HUMAN] 96,75 1 12 37 173 461 49,94108966 4,817871094 6 P09382 Galectin-1 OS=Homo sapiens GN=LGALS1 PE=1 SV=2 - [LEG1_HUMAN] 96,3 1 7 14 283 135 14,70620005 5,503417969 7 P60174 Triosephosphate isomerase OS=Homo sapiens GN=TPI1 PE=1 SV=3 - [TPIS_HUMAN] 95,1 3 16 25 375 286 30,77169764 5,922363281 8 P04406 Glyceraldehyde-3-phosphate dehydrogenase OS=Homo sapiens GN=GAPDH PE=1 SV=3 - [G3P_HUMAN] 94,63 2 13 31 509 335 36,03039959 8,455566406 9 Q15185 Prostaglandin E synthase 3 OS=Homo sapiens GN=PTGES3 PE=1 SV=1 - [TEBP_HUMAN] 93,13 1 5 12 74 160 18,68541938 4,538574219 10 P09417 Dihydropteridine reductase OS=Homo sapiens GN=QDPR PE=1 SV=2 - [DHPR_HUMAN] 93,03 1 1 17 69 244 25,77302971 7,371582031 11 P01911 HLA class II histocompatibility antigen,
    [Show full text]
  • Increased ACTL6A Occupancy Within Mswi/SNF Chromatin Remodelers
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.22.435873; this version posted March 22, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Increased ACTL6A Occupancy Within mSWI/SNF Chromatin Remodelers Drives Human Squamous Cell Carcinoma Chiung-Ying Chang1,2,7, Zohar Shipony3,7, Ann Kuo1,2, Kyle M. Loh4,5, William J. Greenleaf3,6, Gerald R. Crabtree1,2,5* 1Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, USA. 2Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, USA. 3Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA. 4Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, USA. 5Department of Developmental Biology, Stanford University School of Medicine, Stanford, California 94305, USA. 6Department of Applied Physics, Stanford University, Stanford, California 94305, USA. 7These authors contributed equally. *Corresponding author: Gerald R. Crabtree, [email protected] Summary Mammalian SWI/SNF (BAF) chromatin remodelers play dosage-sensitive roles in many human malignancies and neurologic disorders. The gene encoding the BAF subunit, ACTL6A, is amplified at an early stage in the development of squamous cell carcinomas (SCCs), but its oncogenic role remains unclear. Here we demonstrate that ACTL6A overexpression leads to its stoichiometric assembly into BAF complexes and drives its interaction and engagement with specific regulatory regions in the genome. In normal epithelial cells, ACTL6A was sub-stoichiometric to other BAF subunits. However, increased ACTL6A levels by ectopic expression or in SCC cells led to near-saturation of ACTL6A within BAF complexes.
    [Show full text]
  • Landscape of Tumor Suppressor Long Noncoding Rnas in Breast Cancer Boran Pang1, Qin Wang2, Shipeng Ning2, Junqiang Wu2, Xingda Zhang2, Yanbo Chen2 and Shouping Xu2*
    Pang et al. Journal of Experimental & Clinical Cancer Research (2019) 38:79 https://doi.org/10.1186/s13046-019-1096-0 RESEARCH Open Access Landscape of tumor suppressor long noncoding RNAs in breast cancer Boran Pang1, Qin Wang2, Shipeng Ning2, Junqiang Wu2, Xingda Zhang2, Yanbo Chen2 and Shouping Xu2* Abstract Background: The landscape and biological functions of tumor suppressor long noncoding RNAs in breast cancer are still unknown. Methods: Data from whole transcriptome sequencing of 33 breast specimens in the Harbin Medical University Cancer Center cohort and The Cancer Genome Atlas was applied to identify and validate the landscape of tumor suppressor long noncoding RNAs, which was further validated by The Cancer Genome Atlas pancancer data including 33 cancer types and 12,839 patients. Next, the expression model, prognostic roles, potential biological functions and epigenetic regulation of tumor suppressor long noncoding RNAs were investigated and validated in the breast cancer and pancancer cohorts. Finally, EPB41L4A-AS2 was selected to validate our novel finding, and the tumor suppressive roles of EPB41L4A-AS2 in breast cancer were examined. Results: We identified and validated the landscape of tumor suppressor long noncoding RNAs in breast cancer. The expression of the identified long noncoding RNAs was downregulated in cancer tissue samples compared with normal tissue samples, and these long noncoding RNAs correlated with a favorable prognosis in breast cancer patients and the patients in the pancancer cohort. Multiple carcinogenesis-associated biological functions were predicted to be regulated negatively by these long noncoding RNAs. Moreover, these long noncoding RNAs were transcriptionally regulated by epigenetic modification, including DNA methylation and histone methylation modification.
    [Show full text]
  • Biological and Prognostic Significance of Chromosome 5Q Deletions in Myeloid Malignancies Aristoteles A.N
    Review Biological and Prognostic Significance of Chromosome 5q Deletions in Myeloid Malignancies Aristoteles A.N. Giagounidis,1Ulrich Germing,2 and Carlo Aul1 Abstract The presence of del(5q), either as the sole karyotypic abnormality or as part of a more complex karyotype, has distinct clinical implications for myelodysplastic syndromes (MDS) and acute myeloid leukemia. The 5qÀ syndrome, a subtype of low-riskMDS, is characterized by an isolated 5q deletion and <5% blasts in the bone marrow and can serve as a useful model for studying the role of 5q deletions in the pathogenesis and prognosis of myeloid malignancies. Recent clinical results with lenalidomide, an oral immunomodulatory drug, have shown durable erythroid responses, including transfusion independence and complete cytogenetic remissions in patients with del(5q) MDS with or without additional chromosomal abnormalities. These results indicate that lenalidomide can overcome the pathogenic effect of 5q deletion in MDS and restore bone marrow balance. The data provide important new insights into the pathobiology of 5q chromo- somal deletions in myeloid malignancies. Cytogenetic abnormalities are detected in the bone marrow of preponderance, refractory macrocytic anemia, normal or high over 50% of patients diagnosed with primary myelodysplastic platelet counts, hypolobulated megakaryocytes, and modest syndromes (MDS) or myeloid leukemias, and up to 80% of leukopenia (11, 14, 17). The prognosis is favorable in 5qÀ patients with secondary or therapy-related MDS (1, 2). These syndrome with relatively low risk of transformation to AML abnormalities can be characterized as being balanced or (11, 18). Although the limits of 5q deletions vary among unbalanced (3, 4). Balanced cytogenetic abnormalities include patients with 5qÀ syndrome, the most frequent deletion is reciprocal translocations, inversions, and insertions (3, 5, 6).
    [Show full text]
  • The Hippo Pathway Component Wwc2 Is a Key Regulator of Embryonic Development and Angiogenesis in Mice Anke Hermann1,Guangmingwu2, Pavel I
    Hermann et al. Cell Death and Disease (2021) 12:117 https://doi.org/10.1038/s41419-021-03409-0 Cell Death & Disease ARTICLE Open Access The Hippo pathway component Wwc2 is a key regulator of embryonic development and angiogenesis in mice Anke Hermann1,GuangmingWu2, Pavel I. Nedvetsky1,ViktoriaC.Brücher3, Charlotte Egbring3, Jakob Bonse1, Verena Höffken1, Dirk Oliver Wennmann1, Matthias Marks4,MichaelP.Krahn 1,HansSchöler5,PeterHeiduschka3, Hermann Pavenstädt1 and Joachim Kremerskothen1 Abstract The WW-and-C2-domain-containing (WWC) protein family is involved in the regulation of cell differentiation, cell proliferation, and organ growth control. As upstream components of the Hippo signaling pathway, WWC proteins activate the Large tumor suppressor (LATS) kinase that in turn phosphorylates Yes-associated protein (YAP) and its paralog Transcriptional coactivator-with-PDZ-binding motif (TAZ) preventing their nuclear import and transcriptional activity. Inhibition of WWC expression leads to downregulation of the Hippo pathway, increased expression of YAP/ TAZ target genes and enhanced organ growth. In mice, a ubiquitous Wwc1 knockout (KO) induces a mild neurological phenotype with no impact on embryogenesis or organ growth. In contrast, we could show here that ubiquitous deletion of Wwc2 in mice leads to early embryonic lethality. Wwc2 KO embryos display growth retardation, a disturbed placenta development, impaired vascularization, and finally embryonic death. A whole-transcriptome analysis of embryos lacking Wwc2 revealed a massive deregulation of gene expression with impact on cell fate determination, 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; cell metabolism, and angiogenesis. Consequently, a perinatal, endothelial-specific Wwc2 KO in mice led to disturbed vessel formation and vascular hypersprouting in the retina.
    [Show full text]
  • (P -Value<0.05, Fold Change≥1.4), 4 Vs. 0 Gy Irradiation
    Table S1: Significant differentially expressed genes (P -Value<0.05, Fold Change≥1.4), 4 vs. 0 Gy irradiation Genbank Fold Change P -Value Gene Symbol Description Accession Q9F8M7_CARHY (Q9F8M7) DTDP-glucose 4,6-dehydratase (Fragment), partial (9%) 6.70 0.017399678 THC2699065 [THC2719287] 5.53 0.003379195 BC013657 BC013657 Homo sapiens cDNA clone IMAGE:4152983, partial cds. [BC013657] 5.10 0.024641735 THC2750781 Ciliary dynein heavy chain 5 (Axonemal beta dynein heavy chain 5) (HL1). 4.07 0.04353262 DNAH5 [Source:Uniprot/SWISSPROT;Acc:Q8TE73] [ENST00000382416] 3.81 0.002855909 NM_145263 SPATA18 Homo sapiens spermatogenesis associated 18 homolog (rat) (SPATA18), mRNA [NM_145263] AA418814 zw01a02.s1 Soares_NhHMPu_S1 Homo sapiens cDNA clone IMAGE:767978 3', 3.69 0.03203913 AA418814 AA418814 mRNA sequence [AA418814] AL356953 leucine-rich repeat-containing G protein-coupled receptor 6 {Homo sapiens} (exp=0; 3.63 0.0277936 THC2705989 wgp=1; cg=0), partial (4%) [THC2752981] AA484677 ne64a07.s1 NCI_CGAP_Alv1 Homo sapiens cDNA clone IMAGE:909012, mRNA 3.63 0.027098073 AA484677 AA484677 sequence [AA484677] oe06h09.s1 NCI_CGAP_Ov2 Homo sapiens cDNA clone IMAGE:1385153, mRNA sequence 3.48 0.04468495 AA837799 AA837799 [AA837799] Homo sapiens hypothetical protein LOC340109, mRNA (cDNA clone IMAGE:5578073), partial 3.27 0.031178378 BC039509 LOC643401 cds. [BC039509] Homo sapiens Fas (TNF receptor superfamily, member 6) (FAS), transcript variant 1, mRNA 3.24 0.022156298 NM_000043 FAS [NM_000043] 3.20 0.021043295 A_32_P125056 BF803942 CM2-CI0135-021100-477-g08 CI0135 Homo sapiens cDNA, mRNA sequence 3.04 0.043389246 BF803942 BF803942 [BF803942] 3.03 0.002430239 NM_015920 RPS27L Homo sapiens ribosomal protein S27-like (RPS27L), mRNA [NM_015920] Homo sapiens tumor necrosis factor receptor superfamily, member 10c, decoy without an 2.98 0.021202829 NM_003841 TNFRSF10C intracellular domain (TNFRSF10C), mRNA [NM_003841] 2.97 0.03243901 AB002384 C6orf32 Homo sapiens mRNA for KIAA0386 gene, partial cds.
    [Show full text]
  • Single Cell Regulatory Landscape of the Mouse Kidney Highlights Cellular Differentiation Programs and Disease Targets
    ARTICLE https://doi.org/10.1038/s41467-021-22266-1 OPEN Single cell regulatory landscape of the mouse kidney highlights cellular differentiation programs and disease targets Zhen Miao 1,2,3,8, Michael S. Balzer 1,2,8, Ziyuan Ma 1,2,8, Hongbo Liu1,2, Junnan Wu 1,2, Rojesh Shrestha 1,2, Tamas Aranyi1,2, Amy Kwan4, Ayano Kondo 4, Marco Pontoglio 5, Junhyong Kim6, ✉ Mingyao Li 7, Klaus H. Kaestner2,4 & Katalin Susztak 1,2,4 1234567890():,; Determining the epigenetic program that generates unique cell types in the kidney is critical for understanding cell-type heterogeneity during tissue homeostasis and injury response. Here, we profile open chromatin and gene expression in developing and adult mouse kidneys at single cell resolution. We show critical reliance of gene expression on distal regulatory elements (enhancers). We reveal key cell type-specific transcription factors and major gene- regulatory circuits for kidney cells. Dynamic chromatin and expression changes during nephron progenitor differentiation demonstrates that podocyte commitment occurs early and is associated with sustained Foxl1 expression. Renal tubule cells follow a more complex differentiation, where Hfn4a is associated with proximal and Tfap2b with distal fate. Mapping single nucleotide variants associated with human kidney disease implicates critical cell types, developmental stages, genes, and regulatory mechanisms. The single cell multi-omics atlas reveals key chromatin remodeling events and gene expression dynamics associated with kidney development. 1 Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA. 2 Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
    [Show full text]