Developmental Toxicity of Polycyclic Aromatic Hydrocarbons: Defining Mechanisms with Systems-Based Transcriptional Profiling

Total Page:16

File Type:pdf, Size:1020Kb

Developmental Toxicity of Polycyclic Aromatic Hydrocarbons: Defining Mechanisms with Systems-Based Transcriptional Profiling AN ABSTRACT OF THE DISSERTATION OF Britton C. Goodale for the degree of Doctor of Philosophy in Toxicology presented on August 12, 2013. Title: Developmental Toxicity of Polycyclic Aromatic Hydrocarbons: Defining Mechanisms with Systems-based Transcriptional Profiling Abstract approved:___________________________________________________________________________________ Robert L. Tanguay Polycyclic aromatic hydrocarbons (PAHs) are ubiquitous in the environment as components of fossil fuels and by-products of combustion. Defining toxicity mechanisms for this large family of multi-ring structures and substituted derivatives is a substantial challenge. Several PAHs, such as benzo(a)pyrene (BaP), are mutagenic, toxic to wildlife, and classified as probable carcinogens to humans. PAHs are present in the environment both in the gaseous phase as well as associated with particulates, and exposures occur via complex mixtures; combustion emissions contain PAHs along with many other contaminants. Cardiac dysfunction and adverse birth outcomes associated with exposure to airborne PAHs suggest that this family of compounds may have non-mutagenic biological activities that affect human health. Some PAHs exert toxic effects via binding the aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor that mediates transcription of many downstream target genes, including cytochrome P450 metabolizing enzymes. Unlike planar halogenated hydrocarbons, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), PAHs are readily metabolized by CYP1A, CYP1B1 and other enzymes, which create reactive intermediates and/or facilitate excretion. Mechanisms of PAH toxicity therefore include canonical AHR signaling, induction of oxidative stress, and other lesser-understood activities that do not require the AHR. We employed zebrafish as a model to rapidly assess developmental toxicity, global transcriptional responses and AHR activation in embryos exposed to parent and oxygenated PAHs (OPAHs). Using comparative analysis of mRNA expression profiles from microarrays with embryos exposed to benz(a)anthracene (BAA), dibenzothiophene (DBT) and pyrene (PYR), we identified expression biomarkers and disrupted biological processes that precede developmental abnormalities. These transcriptional responses were associated with PAH body burdens in the embryos detected by GC-MS. We found that uptake data were essential for discerning molecular pathways from dose-related differences, and identified two primary toxicity profiles. While BAA disrupted transcripts involved in vasculogenesis, DBT and PYR misregulated ion homeostasis and muscle-related genes. NfKB signaling was predicted to be involved in both responses, but canonical AHR signaling was only activated by BAA. In order to study the role of the AHR in mediating toxicity of PAHs, we developed an AHR2 mutant zebrafish line, which has a mutation in the transactivation domain of AHR2. We used AHR agonists TCDD and leflunomide as toxicological probes to characterize AHR activity in the mutant line, and determined that the mutants were functionally null. Finally, we used AHR2 deficient zebrafish embryos to investigate mechanisms by which two four-ring OPAHs induced developmental effects. 1,9 benz-10-anthrone (BEZO) and benz(a)anthracene-7,12-dione (7,12-B[a]AQ) both caused malformations in developing embryos, but they differentially induced CYP1A expression. Despite this difference, the toxicity produced from both compounds was AHR2-dependent. We used mRNA-seq to compare the transcriptional profiles of BEZO and 7,12-B[a]AQ, and identified transcriptional networks that will be investigated further to determine how ligands differentially modulate AHR activity. We also discovered novel transcripts that are potentially important mediators of AHR toxic effects. Comparison across all five parent and OPAHs highlighted clusters of genes that, surprisingly, were similarly expressed in response to the OPAHs, DBT and PYR. These commonly-regulated transcripts may be important to consider when investigating toxicity of PAH mixtures. Together, these studies show that PAHs act via different transcriptional mechanisms, but can be categorized based on transcriptional profiles and differential AHR activation. The clusters of transcripts identified may be involved in common pathways; further investigation of transcription factors and coactivators that interact with mixexpressed genes is a promising area of research for elucidating diverse functions of the AHR. ©Copyright by Britton C. Goodale August 12, 2013 All Rights Reserved Developmental Toxicity of Polycyclic Aromatic Hydrocarbons: Defining Mechanisms with Systems-based Transcriptional Profiling by Britton C. Goodale A DISSERTATION submitted to Oregon State University in partial fulfillment of the requirements for the degree of Doctor of Philosophy Presented August 12, 2013 Commencement June 2014 Doctor of Philosophy dissertation of Britton C. Goodale presented on August 12, 2013. APPROVED: _________________________________________________________________________________________________________ Major Professor, representing Toxicology _________________________________________________________________________________________________________ Head of the Department of Environmental and Molecular Toxicology _________________________________________________________________________________________________________ Dean of the Graduate School I understand that my dissertation will become part of the permanent collection of Oregon State University libraries. My signature below authorizes release of my dissertation to any reader upon request. _______________________________________________________________________________________________________ Britton C. Goodale, Author ACKNOWLEDGEMENTS The completion of this dissertation has by no means been a solitary effort, and I am grateful to the many people who made it possible. I would like to express my sincere gratitude to my graduate mentor, Robert Tanguay, for giving me the opportunity to join the lab and pursue what were, five years ago, fairly specific interests in toxicology. Thank you for sharing your endless excitement about new ideas, and for broadening my perspective and enthusiasm for this field of research more than I could have imagined. None of the work presented here would have been possible without the support I had during my time at OSU, and I am appreciative of the excellent resources that the Tanguay lab has provided. I would like to thank the many members of the Tanguay lab who have helped me over the years: Jane La Du, for her humor and for always being there to help in a pinch; Kate Saili, Tamara Tal, Lisa Truong, Galen Miller and Sumitra Sengupta for providing training in the lab, and for their mentorship and friendship; Andrea Knecht, Mike Simonich, and Annika Swanson for their help and patience with the OPAH studies; Leah Wehmas, Derik Haggard, Siba Das and Cory Gerlach for editing, experiment assistance, and helping me through this final year with excellent company in the lab; Sean Bugel for sharing the office with the window and the challenge assay, providing thoughtful answers to my many questions, and for the excellent discussions; Jill Franzosa for encouragement and advice, both in the lab and on the climbing wall; and Margaret Corvi for helping me keep perspective, making the OPAH body burden studies possible, and being an inspiration to work with. All of the studies in this dissertation were truly made possible by the SARL staff, and I am thankful to Carrie Barton and Cari Buchner for their dedication to the zebrafish colony, particularly the AHR2 mutants (they will some day express their gratitude, too, I’m sure). Thank you to Greg Gonnerman, Chapell Miller for screening help, and to all of the other staff who have kept SARL running. I would like to thank others at OSU for their support; I am grateful to my committee members Daniel Sudakin, Michael Freitag, Kim Anderson, Barbara Taylor and Jeffrey Greenwood for their wisdom and guidance of this project. The Anderson lab welcomed me to OSU, and I can’t imagine my time here without them. Thank you to the many folks who helped me with chemistry questions, compound requests, advised me on environmental relevance for my studies, and gave me good excuses to go to campus. I thank the EMT department for making my graduate work possible, especially department head Craig Marcus for helping me accomplish many goals, and all of the EMT office staff for making things run smoothly. I am especially grateful to my friends who, from near and far, supported me through every step of graduate school. Thank you for being my family when I arrived, exploring the west coast with me, and making Oregon home. Since graduate work feels, much of the time, like a process of many failures, successes in other arenas were an essential part of life the past five years. Thank you to everyone who buoyed my sanity with triumphant climbs, bikes, and foraging adventures. I am so thankful to Erik Endrulat, for his support through this endeavor, and for gently reminding me to think about balance. I was unable to conduct a controlled study on the topic, but nevertheless attribute my perplexing decrease in flu susceptibility the past two years to the good food and abundance of laughter I’ve enjoyed with him. Most of all, I am forever grateful to my family, for their love and support through this entire journey, especially when I chose to move across the country. To my sister, Jane: thank you for your
Recommended publications
  • Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model
    Downloaded from http://www.jimmunol.org/ by guest on September 25, 2021 T + is online at: average * The Journal of Immunology , 34 of which you can access for free at: 2016; 197:1477-1488; Prepublished online 1 July from submission to initial decision 4 weeks from acceptance to publication 2016; doi: 10.4049/jimmunol.1600589 http://www.jimmunol.org/content/197/4/1477 Molecular Profile of Tumor-Specific CD8 Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A. Waugh, Sonia M. Leach, Brandon L. Moore, Tullia C. Bruno, Jonathan D. Buhrman and Jill E. Slansky J Immunol cites 95 articles Submit online. Every submission reviewed by practicing scientists ? is published twice each month by Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts http://jimmunol.org/subscription Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html http://www.jimmunol.org/content/suppl/2016/07/01/jimmunol.160058 9.DCSupplemental This article http://www.jimmunol.org/content/197/4/1477.full#ref-list-1 Information about subscribing to The JI No Triage! Fast Publication! Rapid Reviews! 30 days* Why • • • Material References Permissions Email Alerts Subscription Supplementary The Journal of Immunology The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2016 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. This information is current as of September 25, 2021. The Journal of Immunology Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model Katherine A.
    [Show full text]
  • Joint Gene Network Construction by Single-Cell RNA Sequencing Data
    bioRxiv preprint doi: https://doi.org/10.1101/2021.07.14.452387; this version posted July 14, 2021. 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 4.0 International license. Biometrics xx, 1–24 DOI: pending...... xxxx 2021 Joint Gene Network Construction by Single-Cell RNA Sequencing Data Meichen Dong1, Yiping He2, Yuchao Jiang 1,3, Fei Zou1,3,∗ 1Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, U.S.A. 2Department of Pathology, Duke University, Durham, North Carolina, U.S.A. 3Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, U.S.A. email: [email protected] Summary: In contrast to differential gene expression analysis at single gene level, gene regulatory networks (GRN) analysis depicts complex transcriptomic interactions among genes for better understandings of underlying genetic architectures of human diseases and traits. Recently, single-cell RNA sequencing (scRNA-seq) data has started to be used for constructing GRNs at a much finer resolution than bulk RNA-seq data and microarray data. However, scRNA- seq data are inherently sparse which hinders direct application of the popular Gaussian graphical models (GGMs). Furthermore, most existing approaches for constructing GRNs with scRNA-seq data only consider gene networks under one condition. To better understand GRNs under different but related conditions with single-cell resolution, we propose to construct Joint Gene Networks with scRNA-seq data (JGNsc) using the GGMs framework.
    [Show full text]
  • Magnesium-Protoporphyrin Chelatase of Rhodobacter
    Proc. Natl. Acad. Sci. USA Vol. 92, pp. 1941-1944, March 1995 Biochemistry Magnesium-protoporphyrin chelatase of Rhodobacter sphaeroides: Reconstitution of activity by combining the products of the bchH, -I, and -D genes expressed in Escherichia coli (protoporphyrin IX/tetrapyrrole/chlorophyll/bacteriochlorophyll/photosynthesis) LUCIEN C. D. GIBSON*, ROBERT D. WILLOWSt, C. GAMINI KANNANGARAt, DITER VON WETTSTEINt, AND C. NEIL HUNTER* *Krebs Institute for Biomolecular Research and Robert Hill Institute for Photosynthesis, Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, S10 2TN, United Kingdom; and tCarlsberg Laboratory, Department of Physiology, Gamle Carlsberg Vej 10, DK-2500 Copenhagen Valby, Denmark Contributed by Diter von Wettstein, November 14, 1994 ABSTRACT Magnesium-protoporphyrin chelatase lies at Escherichia coli and demonstrate that the extracts of the E. coli the branch point of the heme and (bacterio)chlorophyll bio- transformants can convert Mg-protoporphyrin IX to Mg- synthetic pathways. In this work, the photosynthetic bacte- protoporphyrin monomethyl ester (20, 21). Apart from posi- rium Rhodobacter sphaeroides has been used as a model system tively identifying bchM as the gene encoding the Mg- for the study of this reaction. The bchH and the bchI and -D protoporphyrin methyltransferase, this work opens up the genes from R. sphaeroides were expressed in Escherichia coli. possibility of extending this approach to other parts of the When cell-free extracts from strains expressing BchH, BchI, pathway. In this paper, we report the expression of the genes and BchD were combined, the mixture was able to catalyze the bchH, -I, and -D from R. sphaeroides in E. coli: extracts from insertion of Mg into protoporphyrin IX in an ATP-dependent these transformants, when combined in vitro, are highly active manner.
    [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]
  • Haploinsufficiency of Cardiac Myosin Binding Protein-C in the Development of Hypertrophic Cardiomyopathy
    Loyola University Chicago Loyola eCommons Dissertations Theses and Dissertations 2014 Haploinsufficiency of Cardiac Myosin Binding Protein-C in the Development of Hypertrophic Cardiomyopathy David Barefield Loyola University Chicago Follow this and additional works at: https://ecommons.luc.edu/luc_diss Part of the Physiology Commons Recommended Citation Barefield, David, "Haploinsufficiency of Cardiac Myosin Binding Protein-C in the Development of Hypertrophic Cardiomyopathy" (2014). Dissertations. 1249. https://ecommons.luc.edu/luc_diss/1249 This Dissertation is brought to you for free and open access by the Theses and Dissertations at Loyola eCommons. It has been accepted for inclusion in Dissertations by an authorized administrator of Loyola eCommons. For more information, please contact [email protected]. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 3.0 License. Copyright © 2014 David Barefield LOYOLA UNIVERSITY CHICAGO HAPLOINSUFFICIENCY OF CARDIAC MYOSIN BINDING PROTEIN-C IN THE DEVELOPMENT OF HYPERTROPHIC CARDIOMYOPATHY A DISSERTATION SUBMITTED TO THE FACULTY OF THE GRADUATE SCHOOL IN CANDIDACY FOR THE DEGREE OF DOCTOR OF PHILOSOPHY PROGRAM IN CELL AND MOLECULAR PHYSIOLOGY BY DAVID YEOMANS BAREFIELD CHICAGO, IL AUGUST 2014 Copyright by David Yeomans Barefield, 2014 All Rights Reserved. ii ACKNOWLEDGEMENTS The completion of this work would not have been possible without the support of excellent mentors, colleagues, friends, and family. I give tremendous thanks to my mentor, Dr. Sakthivel Sadayappan, who has facilitated my growth as a scientist and as a human being over the past five years. I would like to thank my dissertation committee: Drs. Pieter de Tombe, Kenneth Byron, Leanne Cribbs, Kyle Henderson, and Christine Seidman for their erudite guidance of my project and my development as a scientist.
    [Show full text]
  • Analysis and Interpretation of Next-Generation Sequencing Data for the Identification of Genetic Variants Involved in Cardiovascular Malformation
    Analysis and interpretation of next-generation sequencing data for the identification of genetic variants involved in cardiovascular malformation Darren T. Houniet For the degree of Doctor of Philosophy Newcastle University Faculty of Medical Sciences Institute of Genetic Medicine February 2013 Abstract Congenital cardiovascular malformation (CVM) affects 7/1000 live births. Approximately 20% of cases are caused by chromosomal and syndromic conditions. Rare Mendelian families segregating particular forms of CVM have also been described. Among the remaining 80% of non-syndromic cases, there is a familial predisposition implicating as yet unidentified genetic factors. Since the reproductive consequences to an individual of CVM are usually severe, evolutionary considerations suggest predisposing variants are likely to be rare. The overall aim of my PhD was to use next generation sequencing (NGS) methods to identify such rare, potentially disease causing variants in CVM. First, I developed a novel approach to calculate the sensitivity and specificity of NGS data in detecting variants using publicly available population frequency data. My aim was to provide a method that would yield sound estimates of the quality of a sequencing experiment without the need for additional genotyping in the sequenced samples. I developed such a method and demonstrated that it provided comparable results to methods using microarray data as a reference. Furthermore, I evaluated different variant calling pipelines and showed that they have a large effect on sensitivity and specificity. Following this, the NovoAlign-Samtools and BWA-Dindel pipelines were used to identify single base substitution and indel variants in three pedigrees, where predisposition to a different disease appears to segregate following an autosomal dominant mode of inheritance.
    [Show full text]
  • AOP 131: Aryl Hydrocarbon Receptor Activation Leading to Uroporphyria
    Organisation for Economic Co-operation and Development DOCUMENT CODE For Official Use English - Or. English 1 January 1990 AOP 131: Aryl hydrocarbon receptor activation leading to uroporphyria Short Title: AHR activation-uroporphyria This document was approved by the Extended Advisory Group on Molecular Screening and Toxicogenomics in June 2018. The Working Group of the National Coordinators of the Test Guidelines Programme and the Working Party on Hazard Assessment are invited to review and endorse the AOP by 29 March 2019. Magdalini Sachana, Administrator, Hazard Assessment, [email protected], +(33- 1) 85 55 64 23 Nathalie Delrue, Administrator, Test Guidelines, [email protected], +(33-1) 45 24 98 44 This document, as well as any data and map included herein, are without prejudice to the status of or sovereignty over any territory, to the delimitation of international frontiers and boundaries and to the name of any territory, city or area. 2 │ Foreword This Adverse Outcome Pathway (AOP) on Aryl hydrocarbon receptor activation leading to uroporphyria, has been developed under the auspices of the OECD AOP Development Programme, overseen by the Extended Advisory Group on Molecular Screening and Toxicogenomics (EAGMST), which is an advisory group under the Working Group of the National Coordinators for the Test Guidelines Programme (WNT). The AOP has been reviewed internally by the EAGMST, externally by experts nominated by the WNT, and has been endorsed by the WNT and the Working Party on Hazard Assessment (WPHA) in xxxxx. Through endorsement of this AOP, the WNT and the WPHA express confidence in the scientific review process that the AOP has undergone and accept the recommendation of the EAGMST that the AOP be disseminated publicly.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2016/0289762 A1 KOH Et Al
    US 201602897.62A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2016/0289762 A1 KOH et al. (43) Pub. Date: Oct. 6, 2016 (54) METHODS FOR PROFILIING AND Publication Classification QUANTITATING CELL-FREE RNA (51) Int. Cl. (71) Applicant: The Board of Trustees of the Leland CI2O I/68 (2006.01) Stanford Junior University, Palo Alto, (52) U.S. Cl. CA (US) CPC ....... CI2O 1/6883 (2013.01); C12O 2600/112 (2013.01); C12O 2600/118 (2013.01); C12O (72) Inventors: Lian Chye Winston KOH, Stanford, 2600/158 (2013.01) CA (US); Stephen R. QUAKE, Stanford, CA (US); Hei-Mun Christina FAN, Fremont, CA (US); Wenying (57) ABSTRACT PAN, Stanford, CA (US) The invention generally relates to methods for assessing a (21) Appl. No.: 15/034,746 neurological disorder by characterizing circulating nucleic acids in a blood sample. According to certain embodiments, (22) PCT Filed: Nov. 6, 2014 methods for S. a Nial disorder include (86). PCT No.: PCT/US2O14/064355 obtaining RNA present in a blood sample of a patient Suspected of having a neurological disorder, determining a S 371 (c)(1), level of RNA present in the sample that is specific to brain (2) Date: May 5, 2016 tissue, comparing the sample level of RNA to a reference O O level of RNA specific to brain tissue, determining whether a Related U.S. Application Data difference exists between the sample level and the reference (60) Provisional application No. 61/900,927, filed on Nov. level, and indicating a neurological disorder if a difference 6, 2013.
    [Show full text]
  • Protein Network Analyses of Pulmonary Endothelial Cells In
    www.nature.com/scientificreports OPEN Protein network analyses of pulmonary endothelial cells in chronic thromboembolic pulmonary hypertension Sarath Babu Nukala1,8,9*, Olga Tura‑Ceide3,4,5,9, Giancarlo Aldini1, Valérie F. E. D. Smolders2,3, Isabel Blanco3,4, Victor I. Peinado3,4, Manuel Castell6, Joan Albert Barber3,4, Alessandra Altomare1, Giovanna Baron1, Marina Carini1, Marta Cascante2,7,9 & Alfonsina D’Amato1,9* Chronic thromboembolic pulmonary hypertension (CTEPH) is a vascular disease characterized by the presence of organized thromboembolic material in pulmonary arteries leading to increased vascular resistance, heart failure and death. Dysfunction of endothelial cells is involved in CTEPH. The present study describes for the frst time the molecular processes underlying endothelial dysfunction in the development of the CTEPH. The advanced analytical approach and the protein network analyses of patient derived CTEPH endothelial cells allowed the quantitation of 3258 proteins. The 673 diferentially regulated proteins were associated with functional and disease protein network modules. The protein network analyses resulted in the characterization of dysregulated pathways associated with endothelial dysfunction, such as mitochondrial dysfunction, oxidative phosphorylation, sirtuin signaling, infammatory response, oxidative stress and fatty acid metabolism related pathways. In addition, the quantifcation of advanced oxidation protein products, total protein carbonyl content, and intracellular reactive oxygen species resulted increased
    [Show full text]
  • On Tuning the Fluorescence Emission of Porphyrin Free Bases Bonded to the Pore Walls of Organo-Modified Silica
    Molecules 2014, 19, 2261-2285; doi:10.3390/molecules19022261 OPEN ACCESS molecules ISSN 1420-3049 www.mdpi.com/journal/molecules Article On Tuning the Fluorescence Emission of Porphyrin Free Bases Bonded to the Pore Walls of Organo-Modified Silica Rosa I. Y. Quiroz-Segoviano 1, Iris N. Serratos 1, Fernando Rojas-González 1, Salvador R. Tello-Solís 1, Rebeca Sosa-Fonseca 2, Obdulia Medina-Juárez 1, Carmina Menchaca-Campos 3 and Miguel A. García-Sánchez 1,* 1 Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco 186, Vicentina, D. F. 09340, Mexico 2 Departamento de Física, Universidad Autónoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco 186, Vicentina, D. F. 09340, Mexico 3 Centro de Investigación en Ingeniería y Ciencias Aplicadas, UAEM, Av. Universidad 1001, Col. Chamilpa, C.P. 62209, Cuernavaca Mor., Mexico * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +52-55-5804-4677; Fax: +52-55-5804-4666. Received: 24 December 2013; in revised form: 29 January 2014 / Accepted: 7 February 2014 / Published: 21 February 2014 Abstract: A sol-gel methodology has been duly developed in order to perform a controlled covalent coupling of tetrapyrrole macrocycles (e.g., porphyrins, phthalocyanines, naphthalocyanines, chlorophyll, etc.) to the pores of metal oxide networks. The resulting absorption and emission spectra intensities in the UV-VIS-NIR range have been found to depend on the polarity existing inside the pores of the network; in turn, this polarization can be tuned through the attachment of organic substituents to the tetrapyrrrole macrocycles before bonding them to the pore network.
    [Show full text]
  • AMPK–Akt Double-Negative Feedback Loop in Breast Cancer Cells Regulates Their Adaptation to Matrix Deprivation Manipa Saha1, Saurav Kumar1, Shoiab Bukhari1, Sai A
    Published OnlineFirst January 16, 2018; DOI: 10.1158/0008-5472.CAN-17-2090 Cancer Tumor Biology and Immunology Research AMPK–Akt Double-Negative Feedback Loop in Breast Cancer Cells Regulates Their Adaptation to Matrix Deprivation Manipa Saha1, Saurav Kumar1, Shoiab Bukhari1, Sai A. Balaji1, Prashant Kumar2, Sravanth K. Hindupur1, and Annapoorni Rangarajan1 Abstract Cell detachment from the extracellular matrix triggers anoi- of the pAkthigh/pAMPKlow state. Clinical specimens of primary kis. Disseminated tumor cells must adapt to survive matrix and metastatic breast cancer displayed an Akt-associated gene deprivation, while still retaining the ability to attach at sec- expression signature, whereas circulating breast tumor cells ondary sites and reinitiate cell division. In this study, we displayed an elevated AMPK-dependent gene expression signa- elucidatemechanismsthatenablereversiblematrixattachment ture. Our work establishes a double-negative feedback loop by breast cancer cells. Matrix deprival triggered AMPK activity between Akt and AMPK to control the switch between matrix- and concomitantly inhibited AKT activity by upregulating the attached and matrix-detached states needed to coordinate cell Akt phosphatase PHLPP2. The resultant pAMPKhigh/pAktlow growth and survival during metastasis. state was critical for cell survival in suspension, as PHLPP2 Significance: These findings reveal a molecular switch that silencing also increased anoikis while impairing autophagy regulates cancer cell survival during metastatic dissemination, and metastasis. In contrast, matrix reattachment led to Akt- with the potential to identify targets to prevent metastasis in mediated AMPK inactivation via PP2C-a-mediated restoration breast cancer. Cancer Res; 78(6); 1497–510. Ó2018 AACR. Introduction vival, and metabolism, and plays a major role in tumor progres- sion (4).
    [Show full text]
  • HSP90-Incorporating Chaperome Networks As Biosensor for Disease-Related Pathways in Patient- Specific Midbrain Dopamine Neurons
    ARTICLE DOI: 10.1038/s41467-018-06486-6 OPEN HSP90-incorporating chaperome networks as biosensor for disease-related pathways in patient- specific midbrain dopamine neurons Sarah Kishinevsky1,2,3,4, Tai Wang3, Anna Rodina3, Sun Young Chung1,2, Chao Xu3, John Philip5, Tony Taldone3, Suhasini Joshi3, Mary L. Alpaugh3,14, Alexander Bolaender3, Simon Gutbier6, Davinder Sandhu7, Faranak Fattahi1,2, Bastian Zimmer1,2, Smit K. Shah3, Elizabeth Chang5, Carmen Inda3,15, John Koren 3rd3,16, Nathalie G. Saurat1,2, Marcel Leist 6, Steven S. Gross7, Venkatraman E. Seshan8, Christine Klein9, Mark J. Tomishima1,2,10, Hediye Erdjument-Bromage11,12, Thomas A. Neubert 11,12, Ronald C. Henrickson5, 1234567890():,; Gabriela Chiosis3,13 & Lorenz Studer1,2 Environmental and genetic risk factors contribute to Parkinson’s Disease (PD) pathogenesis and the associated midbrain dopamine (mDA) neuron loss. Here, we identify early PD pathogenic events by developing methodology that utilizes recent innovations in human pluripotent stem cells (hPSC) and chemical sensors of HSP90-incorporating chaperome networks. We show that events triggered by PD-related genetic or toxic stimuli alter the neuronal proteome, thereby altering the stress-specific chaperome networks, which produce changes detected by chemical sensors. Through this method we identify STAT3 and NF-κB signaling activation as examples of genetic stress, and phospho-tyrosine hydroxylase (TH) activation as an example of toxic stress-induced pathways in PD neurons. Importantly, pharmacological inhibition of the stress chaperome network reversed abnormal phospho- STAT3 signaling and phospho-TH-related dopamine levels and rescued PD neuron viability. The use of chemical sensors of chaperome networks on hPSC-derived lineages may present a general strategy to identify molecular events associated with neurodegenerative diseases.
    [Show full text]