Phosphatidylinositol-3 Kinase/Akt and Fos Like Protein 1

Total Page:16

File Type:pdf, Size:1020Kb

Phosphatidylinositol-3 Kinase/Akt and Fos Like Protein 1 PHOSPHATIDYLINOSITOL-3 KINASE/AKT AND FOS LIKE PROTEIN 1 REGULATION OF TROPHOBLAST DIFFERENTIATION BY Lindsey N. Kent Submitted to the graduate degree program in Pathology and Laboratory Medicine and the Graduate Faculty of the University of Kansas in partial fulfillment of the requirements for the degree of Doctor of Philosophy. Graduate committee: ______________________________ Chair: Michael J. Soares, Ph.D. ______________________________ Dr. Linheng Li, Ph.D. ______________________________ Dr. Margaret G. Petroff, Ph.D. ______________________________ Dr. Jay L. Vivian, Ph.D. ______________________________ Dr. Michael W. Wolfe, Ph.D. Date defended: ________________ PHOSPHATIDYLINOSITOL-3 KINASE/AKT AND FOS LIKE PROTEIN 1 REGULATION OF TROPHOBLAST DIFFERENTIATION BY Lindsey N. Kent Submitted to the graduate degree program in Pathology and Laboratory Medicine and the Graduate Faculty of the University of Kansas in partial fulfillment of the requirements for the degree of Doctor of Philosophy. ______________________________ Chair: Michael J. Soares, Ph.D. Date approved: ________________ 2 ACKNOWLEDGEMENTS I would like to thank my family for all of their support, encouragement and enthusiasm. I would also like to acknowledge all members (past and present) of the Soares Laboratory, all of which taught me important lesions and that have made me a better scientist. I would like to thank Dr. Toshiro Konno for his instruction and advice with all of the histology and statistical analysis; Dr. Dong- Soo Lee for his help with the embryo transfer experiments and Dr. Mohammad Rumi for his help with the lentiviral and shRNA experiments as well as his advice that has greatly improved my molecular biology knowledge and technique. I would also like to thank Mrs. Stacy McClure for her advice and support. I would also like to thank my committee members for their help and advice in guiding my research project over the years. Most especially I would like to thank my mentor Dr. Michael Soares. I would never have considered graduate school without his support and a large part of my success is due directly to his guidance and encouragement. 3 ABSTRACT Hemochorial placentation is an effective strategy facilitating nutrient delivery and development of the fetus within the female reproductive tract. Remodeling of the uterine vasculature by trophoblast cells is one of the key processes necessary for a successful pregnancy. Several pathways have been implicated in the regulation of trophoblast invasion including the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway. In this research, we identified genes characteristic of trophoblast stem cell and differentiated states, including those involved in trophoblast invasion and vasculature remodeling. We also evaluated the role of the PI3K/AKT signaling pathway in the regulation of trophoblast cell differentiation. This investigation utilized the Rcho-1 trophoblast stem cells as a model system for trophoblast differentiation. These cells can be maintained in a trophoblast stem cell state or induced to differentiate into trophoblast giant cells, which have the capability to invade. mRNA expression profiling using microarrays provided insights about trophoblast stem cell renewal and differentiation. Genes linked to trophoblast invasion and/or vasculature remodeling were identified. These putative ‘invasion related genes’ were upregulated during differentiation, corresponding to an increase in PI3K/AKT signaling during differentiation. Using pharmacological inhibition of PI3K or AKT we determined that expression of several ‘invasion related genes’ was dependent on the PI3K/AKT signaling pathway. We also showed that the invasive ability of trophoblast cells was decreased when the PI3K/AKT signaling pathway was inhibited. Further analysis using lentiviral delivered shRNAs to 4 knockdown Akt1, Akt2 or Akt3 further validated the involvement of the PI3K/AKT pathway in the regulation of these events. Fos like antigen 1 (FOSL1 or FRA1) was upregulated during trophoblast cell differentiation and shown to be a candidate downstream mediator of the PI3K/AKT signaling pathway. Modulation of the PI3K/AKT signaling pathway resulted in a decrease in FRA1 protein level. Knockdown of FRA1 using lentiviral delivered shRNA resulted in a decrease in a subset of the PI3K/AKT sensitive ‘invasion related genes’ and decreased trophoblast invasion. In conclusion, the PI3K/AKT signaling pathway regulates trophoblast differentiation by regulating FRA1 and the expression of key genes involved in the invasion process. In this research, we have identified key factors contributing to the regulation of rodent hemochorial placentation. The conservation of these regulatory factors in the process of human placentation and their potential involvement in pregnancy-related diseases remain to be determined. 5 TABLE OF CONTENTS Abstract……………………………………………………………………………….…4 Table of Contents………………………………………………………………….…..6 List of Figures and Tables…………………………………………..…………..….10 CHAPTER 1: GENERAL INTRODUCTION………………………………………..13 General Introduction…………………………………………………………….…..14 Rodent Trophoblast Models…………………………………………….…....17 Early lineage decisions in the mammalian embryo………………….……..17 Placentation in the Rodent………………………………..…………..………17 Trophoblast Invasion and Vascular Remodeling…………………..……….23 Rcho-1 Trophoblast Stem Cells……………………………………..……….24 Overview of the Phosphatidylinositol-3 Kinase (PI3K)/AKT Signaling Pathway……………………………………………………..………….27 PI3K/AKT Pathway in Placental and Trophoblast Cell Development and Invasion…………………………………………………………………30 FOS Like Protein 1 (FOSL1 or FRA1)……………………………………….32 PI3K/AKT Signaling Pathway and FRA1…………………………...……....35 FRA1 in Placental and Trophoblast Cell Development……………………35 Objectives of our Research………………………………………………………...37 CHAPTER 2: PHOSPHATIDYLINOSITOL 3-KINASE MODULATION OF TROPHOBLAST CELL DIFFERENTIATION………………………………..…….41 Abstract………………………………………………………………………………..42 Introduction…………………………………………………………………….……..43 6 Materials and Methods………………………………………………………………46 Results…………………………………………………………………………..……..56 Identification of genes associated with trophoblast differentiation……………….56 Trophoblast stem-associated genes………………………………………...56 Trophoblast differentiation-associated genes……………………………....63 PI3K signaling and trophoblast differentiation………………………………………71 PI3K signaling: negatively regulated genes……………………………..…76 PI3K signaling: positively regulated genes…………………………………78 PI3K regulation of trophoblast Steroidogenesis…………………………....79 Discussion…………………………………………………………………………….84 Trophoblast stem cell-associated genes…………………………………………....84 Trophoblast differentiation-associated genes………………………………………88 PI3K signaling-sensitive genes……………………………………………………....88 CHAPTER 3: PHOSPHATIDYLINOSITOL 3-KINASE/AKT SIGNALING PATHWAY AND FOS LIKE ANTIGEN 1 IN THE REGULATION OF TROPHOBLAST INVASION AND VASCULAR REMODELING……………..…92 Abstract………………………………………………………………………………..93 Introduction……………………………………………………………………………95 Materials and Methods………………………………………………………………99 Results………………………………………………………………………………..109 The PI3K/AKT pathway and trophoblast cell invasion……………………………109 Trophoblast AKT is expressed, active, and sensitive to small molecule inhibition……………………………………………………………….109 7 Small molecule inhibition of PI3K and AKT in trophoblast cells decreases expression of “invasion-vascular remodeling related” genes……112 Small molecule inhibition of PI3K or AKT decreases MMP9 activity and invasive ability of trophoblast cells………………………………....112 AKT isoform regulation of the expression of ‘invasion-vascular remodeling related’ genes and the invasive ability of trophoblast…………….118 FRA1 and PI3K/AKT signaling……………………………………………………...123 FRA1 is expressed in trophoblast giant cells and invasive trophoblast cells…………………………………………………………………….123 PI3K/AKT signaling regulates FRA1……………………………………….128 FRA1 and trophoblast invasion……………………………………………………..133 FRA1 regulates expression of PI3K/AKT-sensitive ‘invasion-vascular remodeling related’ genes………………………………………..…133 FRA1 regulates in vitro trophoblast cell invasion…………………………133 FRA1 regulates in vivo trophoblast cell invasion…………………………140 Discussion…………………………………………………………………………...145 PI3K/AKT regulation of ‘invasion-vascular remodeling related’ genes…………145 AKT isoforms and trophoblast cell biology………………………………………...146 PI3K/AKT regulation of FRA1……………………………………………………….148 FRA1 and placentation………………………………………………………………149 CHAPTER 4: GENERAL DISCUSSION…………………………………………..151 I. Model……………………………………………………………………………….152 II. Regulation of trophoblast differentiation and invasion by the PI3K/AKT signaling pathway…………………………………………………………….....152 Activation of the PI3K/AKT signaling pathway in trophoblast cells……………..152 Isoform specific roles for AKT in trophoblast cells…………………………….….155 Akt isoforms in placental and embryonic development………..………...155 8 Akt isoforms in invasion……………………………………………………..157 PI3K/AKT mediators of trophoblast invasion……………………………………...158 Regulation of Fos like 1 by the PI3K/Akt signaling pathway…………….158 Crosstalk between PI3K/AKT/FRA1 and other signaling pathways and trophoblast invasion………………………………………………….160 The role of FRA1 in trophoblast invasion……………………………………….…161 Identifying targets of FRA1……………………………………………….…161 Potential binding partners of FRA1…………………………………………161 The function of FRA1 target genes in trophoblast invasion……………..162 Significance of the invasion and vascular remodeling related genes……….….163 III. Human pregnancy related disorders…………………………………………..164
Recommended publications
  • Acetyl Group Coordinated Progression Through the Catalytic Cycle of an Arylalkylamine N-Acetyltransferase
    RESEARCH ARTICLE Acetyl group coordinated progression through the catalytic cycle of an arylalkylamine N-acetyltransferase Adam A. Aboalroub, Ashleigh B. Bachman, Ziming Zhang, Dimitra Keramisanou, David J. Merkler, Ioannis Gelis* Department of Chemistry, University of South Florida, Tampa, Florida, United States of America * [email protected] a1111111111 a1111111111 a1111111111 Abstract a1111111111 a1111111111 The transfer of an acetyl group from acetyl-CoA to an acceptor amine is a ubiquitous bio- chemical transformation catalyzed by Gcn5-related N-acetyltransferases (GNATs). Although it is established that the reaction proceeds through a sequential ordered mecha- nism, the role of the acetyl group in driving the ordered formation of binary and ternary com- OPEN ACCESS plexes remains elusive. Herein, we show that CoA and acetyl-CoA alter the conformation of the substrate binding site of an arylalkylamine N-acetyltransferase (AANAT) to facilitate Citation: Aboalroub AA, Bachman AB, Zhang Z, Keramisanou D, Merkler DJ, Gelis I (2017) Acetyl interaction with acceptor substrates. However, it is the presence of the acetyl group within group coordinated progression through the the catalytic funnel that triggers high affinity binding. Acetyl group occupancy is relayed catalytic cycle of an arylalkylamine N- through a conserved salt bridge between the P-loop and the acceptor binding site, and is acetyltransferase. PLoS ONE 12(5): e0177270. manifested as differential dynamics in the CoA and acetyl-CoA-bound states. The capacity https://doi.org/10.1371/journal.pone.0177270 of the acetyl group carried by an acceptor to promote its tight binding even in the absence of Editor: Viswanathan V. Krishnan, California State CoA, but also its mutually exclusive position to the acetyl group of acetyl-CoA underscore its University Fresno, UNITED STATES importance in coordinating the progression of the catalytic cycle.
    [Show full text]
  • Supp Material.Pdf
    Legends for Supplemental Figures and Tables Figure S1. Expression of Tlx during retinogenesis. (A) Staged embryos were stained for β- galactosidase knocked into the Tlx locus to indicate Tlx expression. Tlx was expressed in the neural blast layer in the early phase of neural retina development (blue signal). (B) Expression of Tlx in neural retina was quantified using Q-PCR at multiple developmental stages. Figure S2. Expression of p27kip1 and cyclin D1 (Ccnd1) at various developmental stages in wild-type or Tlx-/- retinas. (A) Q-PCR analysis of p27kip1 mRNA expression. (B) Western blotting analysis of p27kip1 protein expression. (C) Q-PCR analysis of cyclin D1 mRNA expression. Figure S3. Q-PCR analysis of mRNA expression of Sf1 (A), Lrh1 (B), and Atn1 (C) in wild-type mouse retinas. RNAs from testis and liver were used as controls. Table S1. List of genes dysregulated both at E15.5 and P0 Tlx-/- retinas. Gene E15.5 P0 Cluste Gene Title Fold Fold r Name p-value p-value Change Change nuclear receptor subfamily 0, group B, Nr0b1 1.65 0.0024 2.99 0.0035 member 1 1 Pou4f3 1.91 0.0162 2.39 0.0031 POU domain, class 4, transcription factor 3 1 Tcfap2d 2.18 0.0000 2.37 0.0001 transcription factor AP-2, delta 1 Zic5 1.66 0.0002 2.02 0.0218 zinc finger protein of the cerebellum 5 1 Zfpm1 1.85 0.0030 1.88 0.0025 zinc finger protein, multitype 1 1 Pten 1.60 0.0155 1.82 0.0131 phospatase and tensin homolog 2 Itgb5 -1.85 0.0063 -1.85 0.0007 integrin beta 5 2 Gpr49 6.86 0.0001 15.16 0.0001 G protein-coupled receptor 49 3 Cmkor1 2.60 0.0007 2.72 0.0013
    [Show full text]
  • Exosomal Lncrna DOCK9-AS2 Derived from Cancer Stem Cell-Like
    Dai et al. Cell Death and Disease (2020) 11:743 https://doi.org/10.1038/s41419-020-02827-w Cell Death & Disease ARTICLE Open Access Exosomal lncRNA DOCK9-AS2 derived from cancer stem cell-like cells activated Wnt/β-catenin pathway to aggravate stemness, proliferation, migration, and invasion in papillary thyroid carcinoma Wencheng Dai1, Xiaoxia Jin2,LiangHan1, Haijing Huang3,ZhenhuaJi1, Xinjiang Xu1, Mingming Tang1,BinJiang1 and Weixian Chen1 Abstract Exosomal long non-coding RNAs (lncRNAs) are crucial factors that mediate the extracellular communication in tumor microenvironment. DOCK9 antisense RNA2 (DOCK9-AS2) is an exosomal lncRNA which has not been investigated in papillary thyroid carcinoma (PTC). Based on the result of differentially expressed lncRNAs in PTC via bioinformatics databases, we discovered that DOCK9-AS2 was upregulated in PTC, and presented elevation in plasma exosomes of PTC patients. Functionally, DOCK9-AS2 knockdown reduced proliferation, migration, invasion, epithelial-to- mesenchymal (EMT) and stemness in PTC cells. PTC-CSCs transmitted exosomal DOCK9-AS2 to improve stemness of PTC cells. Mechanistically, DOCK9-AS2 interacted with SP1 to induce catenin beta 1 (CTNNB1) transcription and 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; sponged microRNA-1972 (miR-1972) to upregulate CTNNB1, thereby activating Wnt/β-catenin pathway in PTC cells. In conclusion, PTC-CSCs-derived exosomal lncRNA DOCK9-AS2 activated Wnt/β-catenin pathway to aggravate PTC progression, indicating that DOCK9-AS2 was a potential target for therapies in PTC. Introduction Therefore, more efforts are required for the improvement of Papillary thyroid cancer (PTC) takes up around 80% of targeted therapy and diagnosis in PTC. thyroid cancer (TC) cases1. Treatment outcome of PTC is Long non-coding RNAs (lncRNAs) are known as tran- generally satisfactory, and with appropriate treatment, over scripts without protein-coding ability and consist over 200 95% of PTC patients can survive longer than 5 years2.
    [Show full text]
  • Supporting Information for Proteomics DOI 10.1002/Pmic.200400896
    Supporting Information for Proteomics DOI 10.1002/pmic.200400896 Odette Prat, Frdric Berenguer, Vronique Malard, Emmanuelle Tavan, Nicole Sage, Grard Steinmetz and Eric Quemeneur Transcriptomic and proteomic responses of human renal HEK293 cells to uranium toxicity ª 2004 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.proteomics-journal.de Table 1 : Differentially expressed genes in HEK293 cells treated with uranium at CI50 , CI30 and CI20. GENE ID GENE DESCRIPTION CI50 CI30 CI20 AANAT arylalkylamine N-acetyltransferase 1.66 AASDHPPT aminoadipate-semialdehyde dehydrogenase-phosphopantetheinyl transferase -1.73 ABCC8 ATP-binding cassette, sub-family C (CFTR/MRP), member 8 2.96 2.9 ABCF2 ATP-binding cassette, sub-family F (GCN20), member 2 1.86 ACAT2 acetyl-Coenzyme A acetyltransferase 2 (acetoacetyl Coenzyme A thiolase) -2.47 ACTB actin, beta -2.12 ACTR2 ARP2 actin-related protein 2 homolog (yeast) -1.94 ADAR adenosine deaminase, RNA-specific 1.87 1.92 ADNP activity-dependent neuroprotector -1.03 ADPRTL1 ADP-ribosyltransferase (NAD+; poly (ADP-ribose) polymerase)-like 1 1.48 2.31 2.1 AKAP1 A kinase (PRKA) anchor protein 1 1.59 AKR1C3 aldo-keto reductase family 1, member C3 -1.37 (3-alpha hydroxysteroid dehydrogenase, type II) ALS2CR3 amyotrophic lateral sclerosis 2 (juvenile) chromosome region, candidate 3 -1.21 APBB1 amyloid beta (A4) precursor protein-binding, family B, member 1 (Fe65) 4.41 APC adenomatosis polyposis coli -1.66 APP amyloid beta (A4) precursor protein (protease nexin-II, Alzheimer disease) -1.32 APPBP1 amyloid beta precursor
    [Show full text]
  • Supplemental Table S1
    Entrez Gene Symbol Gene Name Affymetrix EST Glomchip SAGE Stanford Literature HPA confirmed Gene ID Profiling profiling Profiling Profiling array profiling confirmed 1 2 A2M alpha-2-macroglobulin 0 0 0 1 0 2 10347 ABCA7 ATP-binding cassette, sub-family A (ABC1), member 7 1 0 0 0 0 3 10350 ABCA9 ATP-binding cassette, sub-family A (ABC1), member 9 1 0 0 0 0 4 10057 ABCC5 ATP-binding cassette, sub-family C (CFTR/MRP), member 5 1 0 0 0 0 5 10060 ABCC9 ATP-binding cassette, sub-family C (CFTR/MRP), member 9 1 0 0 0 0 6 79575 ABHD8 abhydrolase domain containing 8 1 0 0 0 0 7 51225 ABI3 ABI gene family, member 3 1 0 1 0 0 8 29 ABR active BCR-related gene 1 0 0 0 0 9 25841 ABTB2 ankyrin repeat and BTB (POZ) domain containing 2 1 0 1 0 0 10 30 ACAA1 acetyl-Coenzyme A acyltransferase 1 (peroxisomal 3-oxoacyl-Coenzyme A thiol 0 1 0 0 0 11 43 ACHE acetylcholinesterase (Yt blood group) 1 0 0 0 0 12 58 ACTA1 actin, alpha 1, skeletal muscle 0 1 0 0 0 13 60 ACTB actin, beta 01000 1 14 71 ACTG1 actin, gamma 1 0 1 0 0 0 15 81 ACTN4 actinin, alpha 4 0 0 1 1 1 10700177 16 10096 ACTR3 ARP3 actin-related protein 3 homolog (yeast) 0 1 0 0 0 17 94 ACVRL1 activin A receptor type II-like 1 1 0 1 0 0 18 8038 ADAM12 ADAM metallopeptidase domain 12 (meltrin alpha) 1 0 0 0 0 19 8751 ADAM15 ADAM metallopeptidase domain 15 (metargidin) 1 0 0 0 0 20 8728 ADAM19 ADAM metallopeptidase domain 19 (meltrin beta) 1 0 0 0 0 21 81792 ADAMTS12 ADAM metallopeptidase with thrombospondin type 1 motif, 12 1 0 0 0 0 22 9507 ADAMTS4 ADAM metallopeptidase with thrombospondin type 1
    [Show full text]
  • Integrated Bioinformatic Analysis of RNA Binding Proteins in Hepatocellular Carcinoma
    www.aging-us.com AGING 2021, Vol. 13, No. 2 Research Paper Integrated bioinformatic analysis of RNA binding proteins in hepatocellular carcinoma Ling Wang1,*, Zhen Zhang2,3,*, Yuan Li1, Yanyan Wan1, Baocai Xing1,& 1Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital and Institute, Beijing 100142, China 2Department of Gastroenterological Surgery, Peking University People’s Hospital, Beijing 100044, China 3Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People’s Hospital, Beijing 100044, China *Equal contribution Correspondence to: Baocai Xing; email: [email protected] Keywords: RNA binding protein, hepatocellular carcinoma, biomarker, transcriptomics, proteomics Received: June 8, 2020 Accepted: November 3, 2020 Published: December 19, 2020 Copyright: © 2020 Wang et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. ABSTRACT RNA binding proteins (RBPs) are aberrantly expressed in a tissue-specific manner across many tumors. These proteins, which play a vital role in post-transcriptional gene regulation, are involved in RNA splicing, maturation, transport, stability, degradation, and translation. We set out to establish an accurate risk score model based on RBPs to estimate prognosis in hepatocellular carcinoma (HCC). RNA-sequencing data, proteomic data and corresponding clinical information were acquired from the Cancer Genome Atlas database and the Clinical Proteomic Tumor Analysis Consortium database respectively. We identified 406 differentially expressed RBPs between HCC tumor and normal tissues at the transcriptional and protein level.
    [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]
  • T-Brain Regulates Archenteron Induction Signal 5207 Range of Amplification
    Development 129, 5205-5216 (2002) 5205 Printed in Great Britain © The Company of Biologists Limited 2002 DEV5034 T-brain homologue (HpTb) is involved in the archenteron induction signals of micromere descendant cells in the sea urchin embryo Takuya Fuchikami1, Keiko Mitsunaga-Nakatsubo1, Shonan Amemiya2, Toshiya Hosomi1, Takashi Watanabe1, Daisuke Kurokawa1,*, Miho Kataoka1, Yoshito Harada3, Nori Satoh3, Shinichiro Kusunoki4, Kazuko Takata1, Taishin Shimotori1, Takashi Yamamoto1, Naoaki Sakamoto1, Hiraku Shimada1 and Koji Akasaka1,† 1Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Higashi-Hiroshima 739-8526, Japan 2Department of Integrated Biosciences, Graduate School of Frontier Sciences, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan 3Department of Zoology, Graduate School of Science, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan 4LSL, Nerima-ku, Tokyo 178-0061, Japan *Present address: Evolutionary Regeneration Biology Group, RIKEN Center for Developmental Biology, Kobe 650-0047, Japan †Author for correspondence (e-mail: [email protected]) Accepted 30 July 2002 SUMMARY Signals from micromere descendants play a crucial role in cells, the initial specification of primary mesenchyme cells, sea urchin development. In this study, we demonstrate that or the specification of endoderm. HpTb expression is these micromere descendants express HpTb, a T-brain controlled by nuclear localization of β-catenin, suggesting homolog of Hemicentrotus pulcherrimus. HpTb is expressed that
    [Show full text]
  • CXCR4 Pathway Retards Muscle Atrophy During Cancer Cachexia
    Oncogene (2016) 35, 6212–6222 © 2016 Macmillan Publishers Limited, part of Springer Nature. All rights reserved 0950-9232/16 www.nature.com/onc ORIGINAL ARTICLE Activation of the SDF1/CXCR4 pathway retards muscle atrophy during cancer cachexia GB Martinelli1, D Olivari1, AD Re Cecconi1, L Talamini1, L Ottoboni2, SH Lecker3, C Stretch4, VE Baracos4, OF Bathe5, A Resovi6, R Giavazzi1, L Cervo7 and R Piccirillo1 Cancer cachexia is a life-threatening syndrome that affects most patients with advanced cancers and causes severe body weight loss, with rapid depletion of skeletal muscle. No treatment is available. We analyzed microarray data sets to identify a subset of genes whose expression is specifically altered in cachectic muscles of Yoshida hepatoma-bearing rodents but not in those with diabetes, disuse, uremia or fasting. Ingenuity Pathways Analysis indicated that three genes belonging to the C-X-C motif chemokine receptor 4 (CXCR4) pathway were downregulated only in muscles atrophying because of cancer: stromal cell-derived factor 1 (SDF1), adenylate cyclase 7 (ADCY7), and p21 protein-activated kinase 1 (PAK1). Notably, we found that, in the Rectus Abdominis muscle of cancer patients, the expression of SDF1 and CXCR4 was inversely correlated with that of two ubiquitin ligases induced in muscle wasting, atrogin-1 and MuRF1, suggesting a possible clinical relevance of this pathway. The expression of all main SDF1 isoforms (α, β, γ) also declined in Tibialis Anterior muscle from cachectic mice bearing murine colon adenocarcinoma or human renal cancer and drugs with anticachexia properties restored their expression. Overexpressing genes of this pathway (that is, SDF1 or CXCR4) in cachectic muscles increased the fiber area by 20%, protecting them from wasting.
    [Show full text]
  • A Rac/Cdc42 Exchange Factor Complex Promotes Formation of Lateral filopodia and Blood Vessel Lumen Morphogenesis
    ARTICLE Received 1 Oct 2014 | Accepted 26 Apr 2015 | Published 1 Jul 2015 DOI: 10.1038/ncomms8286 OPEN A Rac/Cdc42 exchange factor complex promotes formation of lateral filopodia and blood vessel lumen morphogenesis Sabu Abraham1,w,*, Margherita Scarcia2,w,*, Richard D. Bagshaw3,w,*, Kathryn McMahon2,w, Gary Grant2, Tracey Harvey2,w, Maggie Yeo1, Filomena O.G. Esteves2, Helene H. Thygesen2,w, Pamela F. Jones4, Valerie Speirs2, Andrew M. Hanby2, Peter J. Selby2, Mihaela Lorger2, T. Neil Dear4,w, Tony Pawson3,z, Christopher J. Marshall1 & Georgia Mavria2 During angiogenesis, Rho-GTPases influence endothelial cell migration and cell–cell adhesion; however it is not known whether they control formation of vessel lumens, which are essential for blood flow. Here, using an organotypic system that recapitulates distinct stages of VEGF-dependent angiogenesis, we show that lumen formation requires early cytoskeletal remodelling and lateral cell–cell contacts, mediated through the RAC1 guanine nucleotide exchange factor (GEF) DOCK4 (dedicator of cytokinesis 4). DOCK4 signalling is necessary for lateral filopodial protrusions and tubule remodelling prior to lumen formation, whereas proximal, tip filopodia persist in the absence of DOCK4. VEGF-dependent Rac activation via DOCK4 is necessary for CDC42 activation to signal filopodia formation and depends on the activation of RHOG through the RHOG GEF, SGEF. VEGF promotes interaction of DOCK4 with the CDC42 GEF DOCK9. These studies identify a novel Rho-family GTPase activation cascade for the formation of endothelial cell filopodial protrusions necessary for tubule remodelling, thereby influencing subsequent stages of lumen morphogenesis. 1 Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
    [Show full text]
  • Genome-Wide Association and Transcriptome Studies Identify Candidate Genes and Pathways for Feed Conversion Ratio in Pigs
    Miao et al. BMC Genomics (2021) 22:294 https://doi.org/10.1186/s12864-021-07570-w RESEARCH ARTICLE Open Access Genome-wide association and transcriptome studies identify candidate genes and pathways for feed conversion ratio in pigs Yuanxin Miao1,2,3, Quanshun Mei1,2, Chuanke Fu1,2, Mingxing Liao1,2,4, Yan Liu1,2, Xuewen Xu1,2, Xinyun Li1,2, Shuhong Zhao1,2 and Tao Xiang1,2* Abstract Background: The feed conversion ratio (FCR) is an important productive trait that greatly affects profits in the pig industry. Elucidating the genetic mechanisms underpinning FCR may promote more efficient improvement of FCR through artificial selection. In this study, we integrated a genome-wide association study (GWAS) with transcriptome analyses of different tissues in Yorkshire pigs (YY) with the aim of identifying key genes and signalling pathways associated with FCR. Results: A total of 61 significant single nucleotide polymorphisms (SNPs) were detected by GWAS in YY. All of these SNPs were located on porcine chromosome (SSC) 5, and the covered region was considered a quantitative trait locus (QTL) region for FCR. Some genes distributed around these significant SNPs were considered as candidates for regulating FCR, including TPH2, FAR2, IRAK3, YARS2, GRIP1, FRS2, CNOT2 and TRHDE. According to transcriptome analyses in the hypothalamus, TPH2 exhibits the potential to regulate intestinal motility through serotonergic synapse and oxytocin signalling pathways. In addition, GRIP1 may be involved in glutamatergic and GABAergic signalling pathways, which regulate FCR by affecting appetite in pigs. Moreover, GRIP1, FRS2, CNOT2,andTRHDE may regulate metabolism in various tissues through a thyroid hormone signalling pathway.
    [Show full text]
  • Predicting Clinical Response to Treatment with a Soluble Tnf-Antagonist Or Tnf, Or a Tnf Receptor Agonist
    (19) TZZ _ __T (11) EP 2 192 197 A1 (12) EUROPEAN PATENT APPLICATION (43) Date of publication: (51) Int Cl.: 02.06.2010 Bulletin 2010/22 C12Q 1/68 (2006.01) (21) Application number: 08170119.5 (22) Date of filing: 27.11.2008 (84) Designated Contracting States: (72) Inventor: The designation of the inventor has not AT BE BG CH CY CZ DE DK EE ES FI FR GB GR yet been filed HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR (74) Representative: Habets, Winand Designated Extension States: Life Science Patents AL BA MK RS PO Box 5096 6130 PB Sittard (NL) (71) Applicant: Vereniging voor Christelijk Hoger Onderwijs, Wetenschappelijk Onderzoek en Patiëntenzorg 1081 HV Amsterdam (NL) (54) Predicting clinical response to treatment with a soluble tnf-antagonist or tnf, or a tnf receptor agonist (57) The invention relates to methods for predicting a clinical response to a therapy with a soluble TNF antagonist, TNF or a TNF receptor agonist and a kit for use in said methods. EP 2 192 197 A1 Printed by Jouve, 75001 PARIS (FR) EP 2 192 197 A1 Description [0001] The invention relates to methods for predicting a clinical response to a treatment with a soluble TNF antagonist, with TNF or a TNF receptor agonist using expression levels of genes of the Type I INF pathway and a kit for use in said 5 methods. In another aspect, the invention relates to a method for evaluating a pharmacological effect of a treatment with a soluble TNF antagonist, TNF or a TNF receptor agonist.
    [Show full text]