Transcriptome Profiling and Molecular Therapeutic Advances in Cystic Fibrosis: Recent Insights

Genes - Tập 10 Số 3 - Trang 180
Justin E. Ideozu1,2,3, Xi Zhang1,2,3, Susanna A. McColley1,2, Hara Levy1,2,3
1Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
2Feinberg School of Medicine at Northwestern University Chicago, Chicago, IL 60611, USA
3Human Molecular Genetics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA

Tóm tắt

In cystic fibrosis (CF), mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene disrupt the capacity of the encoded protein to function as a channel to transport chloride ions and water across cell membranes. The consequences are deleterious, system-wide, and immensely variable, even among patients with the same CFTR genotype. This underscores the need to characterize the mechanisms contributing to CF pathophysiology. Gene replacement and gene editing therapies have been pursued intensively and are expected to provide a one-time treatment for CF. However, gene replacement therapy is limited by the lack of efficient vectors to deliver functional copies of CFTR to cells without immunological complications, while gene editing technologies such as CRISPR/Cas9 are still in their infancy, mainly useful in somatic cells and limited by off-target insertions. Small molecule treatments targeted at potentiating or correcting CFTR have shown clinical benefits, but they are limited to a few CFTR mutations and insufficient to overcome challenges related to clinical heterogeneity. Transcriptome profiling approaches have emerged as robust tools capable of characterizing phenotypic variability and revealing novel molecular targets with therapeutic potential for CF. We summarize current insights gained through transcriptome profiling approaches in CF studies and recent advances in molecular therapeutics.

Từ khóa


Tài liệu tham khảo

Madacsy, 2018, Cystic fibrosis of the pancreas: The role of CFTR channel in the regulation of intracellular Ca(2+) signaling and mitochondrial function in the exocrine pancreas, Front. Physiol., 9, 1585, 10.3389/fphys.2018.01585

Riordan, 1989, Identification of the cystic fibrosis gene: Cloning and characterization of complementary DNA, Science, 245, 1066, 10.1126/science.2475911

Veit, 2016, From CFTR biology toward combinatorial pharmacotherapy: Expanded classification of cystic fibrosis mutations, Mol. Biol. Cell, 27, 424, 10.1091/mbc.e14-04-0935

Fajac, 2017, New treatments targeting the basic defects in cystic fibrosis, Presse Med., 46, E165, 10.1016/j.lpm.2017.01.024

Carithers, 2015, The genotype-tissue expression (GTEx) project, Biopreserv. Biobank., 13, 307, 10.1089/bio.2015.29031.hmm

Burney, 2012, Gene therapy for the treatment of cystic fibrosis, Appl. Clin. Genet., 5, 29

Cooney, A.L., McCray, P.B., and Sinn, P.L. (2018). Cystic fibrosis gene therapy: Looking back, looking forward. Genes (Basel), 9.

Mekus, 2000, Categories of ΔF508 homozygous cystic fibrosis twin and sibling pairs with distinct phenotypic characteristics, Twin Res., 3, 277, 10.1375/136905200320565256

Corvol, 2015, Genome-wide association meta-analysis identifies five modifier loci of lung disease severity in cystic fibrosis, Nat. Commun., 6, 8382, 10.1038/ncomms9382

Kormann, M.S.D., Dewerth, A., Eichner, F., Baskaran, P., Hector, A., Regamey, N., Hartl, D., Handgretinger, R., and Antony, J.S. (2017). Transcriptomic profile of cystic fibrosis patients identifies type I interferon response and ribosomal stalk proteins as potential modifiers of disease severity. PLoS ONE, 12.

Worgall, 2005, Similarity of gene expression patterns in human alveolar macrophages in response to Pseudomonas aeruginosa and Burkholderia cepacia, Infect. Immun., 73, 5262, 10.1128/IAI.73.8.5262-5268.2005

Wright, 2006, Respiratory epithelial gene expression in patients with mild and severe cystic fibrosis lung disease, Am. J. Respir. Cell Mol. Biol., 35, 327, 10.1165/rcmb.2005-0359OC

Verhaeghe, 2007, Role of IKK and ERK pathways in intrinsic inflammation of cystic fibrosis airways, Biochem. Pharmacol., 73, 1982, 10.1016/j.bcp.2007.03.019

Ribeiro, C.M., Hurd, H., Wu, Y., Martino, M.E., Jones, L., Brighton, B., Boucher, R.C., and O’Neal, W.K. (2009). Azithromycin treatment alters gene expression in inflammatory, lipid metabolism, and cell cycle pathways in well-differentiated human airway epithelia. PLoS ONE, 4.

Ogilvie, 2011, Differential global gene expression in cystic fibrosis nasal and bronchial epithelium, Genomics, 98, 327, 10.1016/j.ygeno.2011.06.008

Hampton, 2012, Does the ΔF508-CFTR mutation induce a proinflammatory response in human airway epithelial cells?, Am. J. Physiol. Lung Cell. Mol. Physiol., 303, L509, 10.1152/ajplung.00226.2011

Levy, 2012, Transcriptional signatures as a disease-specific and predictive inflammatory biomarker for type 1 diabetes, Genes Immun., 13, 593, 10.1038/gene.2012.41

Clarke, 2013, Changes in transcriptome of native nasal epithelium expressing F508del-CFTR and intersecting data from comparable studies, Respir. Res., 14, 38, 10.1186/1465-9921-14-38

Mayer, 2013, Rescue of dysfunctional autophagy attenuates hyperinflammatory responses from cystic fibrosis cells, J. Immunol., 190, 1227, 10.4049/jimmunol.1201404

Stanke, 2014, The CF-modifying gene EHF promotes p.Phe508del-CFTR residual function by altering protein glycosylation and trafficking in epithelial cells, Eur. J. Hum. Genet., 22, 660, 10.1038/ejhg.2013.209

Chesne, J., Danger, R., Botturi, K., Reynaud-Gaubert, M., Mussot, S., Stern, M., Danner-Boucher, I., Mornex, J.F., Pison, C., and Dromer, C. (2014). Systematic analysis of blood cell transcriptome in end-stage chronic respiratory diseases. PLoS ONE, 9.

Voisin, 2014, Oxidative stress modulates the expression of genes involved in cell survival in ΔF508 cystic fibrosis airway epithelial cells, Physiol. Genomics, 46, 634, 10.1152/physiolgenomics.00003.2014

Gallins, 2015, Gene expression in transformed lymphocytes reveals variation in endomembrane and HLA pathways modifying cystic fibrosis pulmonary phenotypes, Am. J. Hum. Genet., 96, 318, 10.1016/j.ajhg.2014.12.022

Zeitlin, 2017, Digitoxin for airway inflammation in cystic fibrosis: Preliminary assessment of safety, pharmacokinetics, and dose finding, Ann. Am. Thorac. Soc., 14, 220, 10.1513/AnnalsATS.201608-649OC

Polineni, 2017, Airway mucosal host defense is key to genomic regulation of cystic fibrosis lung disease severity, Am. J. Respir. Crit. Care Med., 197, 79, 10.1164/rccm.201701-0134OC

Jiang, K., Poppenberg, K.E., Wong, L., Chen, Y., Borowitz, D., Goetz, D., Sheehan, D., Frederick, C., Tutino, V.M., and Meng, H. (2018). RNA sequencing data from neutrophils of patients with cystic fibrosis reveals potential for developing biomarkers for pulmonary exacerbations. J. Cyst. Fibros.

Levy, 2019, Identification of molecular signatures of cystic fibrosis disease status using plasma-based functional genomics, Physiol. Genom., 51, 27, 10.1152/physiolgenomics.00109.2018

Casamassimi, A., Federico, A., Rienzo, M., Esposito, S., and Ciccodicola, A. (2017). Transcriptome profiling in human diseases: New advances and perspectives. Int. J. Mol. Sci., 18.

Kasoju, 2017, Transcriptomics of human multipotent mesenchymal stromal cells: Retrospective analysis and future prospects, Biotechnol. Adv., 35, 407, 10.1016/j.biotechadv.2017.04.005

Shi, 2017, Gene expression profiling and functional analysis reveals that p53 pathway-related gene expression is highly activated in cancer cells treated by cold atmospheric plasma-activated medium, PeerJ, 5, e3751, 10.7717/peerj.3751

Westermann, A.J., Barquist, L., and Vogel, J. (2017). Resolving host–pathogen interactions by dual RNA-seq. PLoS Pathogens, 13.

Thomas, 2003, Comparative analyses of multi-species sequences from targeted genomic regions, Nature, 424, 788, 10.1038/nature01858

Collawn, 2014, CFTR and lung homeostasis, Am. J. Physiol. Lung Cell. Mol. Physiol., 307, L917, 10.1152/ajplung.00326.2014

Byrne, 2016, Pulmonary macrophages: A new therapeutic pathway in fibrosing lung disease?, Trends Mol. Med., 22, 303, 10.1016/j.molmed.2016.02.004

Ratner, 2012, Immune responses in cystic fibrosis: Are they intrinsically defective?, Am. J. Respir. Cell Mol. Biol., 46, 715, 10.1165/rcmb.2011-0399RT

Del Porto, P., Cifani, N., Guarnieri, S., Di Domenico, E.G., Mariggio, M.A., Spadaro, F., Guglietta, S., Anile, M., Venuta, F., and Quattrucci, S. (2011). Dysfunctional CFTR alters the bactericidal activity of human macrophages against Pseudomonas aeruginosa. PLoS ONE, 6.

Ideozu, J.E., Zhang, X., Pan, A., Ashrafi, Z., Woods, K.J., Hessner, M.J., Simpson, P., and Levy, H. (2017). Increased expression of plasma-induced ABCC1 mRNA in cystic fibrosis. Int. J. Mol. Sci., 18.

Kiel, 2010, Engineering signal transduction pathways, Cell, 140, 33, 10.1016/j.cell.2009.12.028

Reilly, 2017, Targeting the PI3K/AKT/mTOR signalling pathway in cystic fibrosis, Sci. Rep., 7, 7642, 10.1038/s41598-017-06588-z

Glick, 2010, Autophagy: Cellular and molecular mechanisms, J. Pathol., 221, 3, 10.1002/path.2697

Strubberg, 2018, CFTR modulates Wnt/β-catenin signaling and stem cell proliferation in murine intestine, Cell. Mol. Gastroenterol. Hepatol., 5, 253, 10.1016/j.jcmgh.2017.11.013

Bodas, 2010, The NF-κβ signaling in cystic fibrosis lung disease: Pathophysiology and therapeutic potential, Discov. Med., 9, 346

Liu, 2017, Nf-κβ signaling in inflammation, Signal Transduct. Target. Ther., 2, 17023, 10.1038/sigtrans.2017.23

Verhaeghe, 2007, Early inflammation in the airways of a cystic fibrosis foetus, J. Cyst. Fibros., 6, 304, 10.1016/j.jcf.2006.12.001

Quon, 2016, New and emerging targeted therapies for cystic fibrosis, BMJ, 352, i859, 10.1136/bmj.i859

Schwank, 2013, Functional repair of CFTR by CRISPR/cas9 in intestinal stem cell organoids of cystic fibrosis patients, Cell Stem Cell, 13, 653, 10.1016/j.stem.2013.11.002

Roy, 2018, CRISPR/cascade 9-mediated genome editing-challenges and opportunities, Front. Genet., 9, 240, 10.3389/fgene.2018.00240

Hadida, 2009, Rescue of CF airway epithelial cell function in vitro by a CFTR potentiator, vx-770, Proc. Natl. Acad. Sci. USA, 106, 18825, 10.1073/pnas.0904709106

Koscielny, 2017, Open targets: A platform for therapeutic target identification and validation, Nucleic Acids Res., 45, D985, 10.1093/nar/gkw1055

Beringer, 2012, Pharmacokinetics of doxycycline in adults with cystic fibrosis, Antimicrob. Agents Chemother., 56, 70, 10.1128/AAC.05710-11

Hermes, 2017, Prospective, randomized, double-blind, parallel-group, comparative effectiveness clinical trial comparing a powder vehicle compound of vitamin d with an oil vehicle compound in adults with cystic fibrosis, J. Parenter. Enteral Nutr., 41, 952, 10.1177/0148607116629673

Dalakas, 2018, Immunotherapy in myasthenia gravis in the era of biologics, Nat. Rev. Neurol., 15, 113, 10.1038/s41582-018-0110-z

Narozna, B., Langwinski, W., and Szczepankiewicz, A. (2017). Non-coding RNAs in pediatric airway diseases. Genes, 8.

Glasgow, 2018, Non-coding RNA in cystic fibrosis, Biochem. Soc. Trans., 46, 619, 10.1042/BST20170469

Hammond, 2015, An overview of microRNAs, Adv. Drug Deliv. Rev., 87, 3, 10.1016/j.addr.2015.05.001

Peng, 2016, The role of microRNAs in human cancer, Signal Transduct. Target. Ther., 1, 15004, 10.1038/sigtrans.2015.4

Stolzenburg, 2018, The role of microRNAs in chronic respiratory disease: Recent insights, Biol. Chem., 399, 219, 10.1515/hsz-2017-0249

Oglesby, 2017, MiRNA expression in cystic fibrosis bronchial epithelial cells, Methods Mol. Biol., 1509, 57, 10.1007/978-1-4939-6524-3_7

Bhattacharyya, 2011, Elevated miR-155 promotes inflammation in cystic fibrosis by driving hyperexpression of interleukin-8, J. Biol. Chem., 286, 11604, 10.1074/jbc.M110.198390

Oglesby, 2010, MiR-126 is downregulated in cystic fibrosis airway epithelial cells and regulates TOM1 expression, J. Immunol., 184, 1702, 10.4049/jimmunol.0902669

Palazzo, 2015, Non-coding RNA: What is functional and what is junk?, Front. Genet., 6, 2, 10.3389/fgene.2015.00002

Ponting, 2009, Evolution and functions of long noncoding RNAs, Cell, 136, 629, 10.1016/j.cell.2009.02.006

Xie, 2015, NcRNA-regulated immune response and its role in inflammatory lung diseases, Am. J. Physiol. Lung Cell. Mol. Physiol., 309, L1076, 10.1152/ajplung.00286.2015

Saayman, 2016, Long non-coding RNA BGas regulates the cystic fibrosis transmembrane conductance regulator, Mol. Ther., 24, 1351, 10.1038/mt.2016.112

McKiernan, 2014, Long noncoding RNA are aberrantly expressed in vivo in the cystic fibrosis bronchial epithelium, Int. J. Biochem. Cell Biol., 52, 184, 10.1016/j.biocel.2014.02.022

Balloy, 2017, Bronchial epithelial cells from cystic fibrosis patients express a specific long non-coding RNA signature upon Pseudomonas aeruginosa infection, Front. Cell Infect. Microbiol., 7, 218, 10.3389/fcimb.2017.00218

Kamei, 2019, Integrative expression analysis identifies a novel interplay between CFTR and linc-SUMF1-2 that involves CF-associated gene dysregulation, Biochem. Biophys. Res. Commun., 509, 521, 10.1016/j.bbrc.2018.12.152

Li, 2017, Noncoding RNAs and their potential therapeutic applications in tissue engineering, Engineering, 3, 3, 10.1016/J.ENG.2017.01.005

Fabbri, E., Tamanini, A., Jakova, T., Gasparello, J., Manicardi, A., Corradini, R., Sabbioni, G., Finotti, A., Borgatti, M., and Lampronti, I. (2017). A peptide nucleic acid against microRNA miR-145-5p enhances the expression of the cystic fibrosis transmembrane conductance regulator (CFTR) in calu-3 cells. Molecules, 23.

Donaldson, 2016, Alternative RNA splicing: Contribution to pain and potential therapeutic strategy, Drug Discov. Today, 21, 1787, 10.1016/j.drudis.2016.06.017

Cieply, 2015, Functional roles of alternative splicing factors in human disease, Wiley Interdiscip. Rev. RNA, 6, 311, 10.1002/wrna.1276

Kim, 2018, Alternative splicing isoforms in health and disease, Pflugers Arch., 470, 995, 10.1007/s00424-018-2136-x

Niu, L., Huang, W., Umbach, D.M., and Li, L. (2014). Iuta: A tool for effectively detecting differential isoform usage from RNA-seq data. BMC Genom., 15.

Stricker, T.P., Brown, C.D., Bandlamudi, C., McNerney, M., Kittler, R., Montoya, V., Peterson, A., Grossman, R., and White, K.P. (2017). Robust stratification of breast cancer subtypes using differential patterns of transcript isoform expression. PLoS Genet., 13.

Khatoon, 2014, Introduction to RNA-seq and its applications to drug discovery and development, Drug Dev. Res., 75, 324, 10.1002/ddr.21215

Snouwaert, 1992, An animal-model for cystic-fibrosis made by gene targeting, Science, 257, 1083, 10.1126/science.257.5073.1083

Mou, 2015, Personalized medicine for cystic fibrosis: Establishing human model systems, Pediatr. Pulmonol., 50, S14, 10.1002/ppul.23233

Harris, 1997, Towards an ovine model of cystic fibrosis, Hum. Mol. Genet., 6, 2191, 10.1093/hmg/6.13.2191

Rogers, 2008, Production of CFTR-null and CFTR-ΔF508 heterozygous pigs by adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer, J. Clin. Invest., 118, 1571, 10.1172/JCI34773

Rosen, 2018, Animal and model systems for studying cystic fibrosis, J. Cyst. Fibros., 17, S28, 10.1016/j.jcf.2017.09.001

Palatnik, A., Ye, S., Kendziorski, C., Iden, M., Zigman, J.S., Hessner, M.J., and Rader, J.S. (2017). Identification of a serum-induced transcriptional signature associated with metastatic cervical cancer. PLoS ONE, 12.

Kaldunski, 2010, Identification of a serum-induced transcriptional signature associated with type 1 diabetes in the biobreeding rat, Diabetes, 59, 2375, 10.2337/db10-0372

Kanter, 2015, Single cell transcriptomics: Methods and applications, Front. Oncol., 5, 53, 10.3389/fonc.2015.00053

Chen, 2018, Profiling tumor infiltrating immune cells with cibersort, Methods Mol. Biol., 1711, 243, 10.1007/978-1-4939-7493-1_12

Bolen, C.R., Uduman, M., and Kleinstein, S.H. (2011). Cell subset prediction for blood genomic studies. BMC Bioinform., 12.

Aran, 2017, Xcell: Digitally portraying the tissue cellular heterogeneity landscape, Genome Biol., 18, 220, 10.1186/s13059-017-1349-1

Plasschaert, 2018, A single-cell atlas of the airway epithelium reveals the CFTR-rich pulmonary ionocyte, Nature, 560, 377, 10.1038/s41586-018-0394-6