0
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      “Shocking” the System to Achieve Efficient Gene Targeting in Primary Human Airway Epithelia

      editorial

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The ability to knock out specific genes to gain an understanding of their function is a mainstay of modern research, and the use of technologies to achieve this goal to study pulmonary biology is no exception. Homologous recombination in embryonic stem cells, antisense oligonucleotides, sh/siRNA, and CRISPR-mediated deletions are widely known and used. Most of these methods will perform well in various model systems and cell lines, with the classic method, homologous recombination in embryonic stem cells, being highly useful in animals. However, for translational questions, interspecies and/or cell line differences often require confirmation of functional findings in primary human cells. Primary human airway cultures are a highly valued model to study pulmonary biology, and the application of robust gene knockdown technologies to these cells is a valuable method to foster our knowledge in this field. Unfortunately, as with other primary cell models, primary airway cells generally remain persistently defiant to easy genetic manipulation. Many examples of successful gene manipulation in human airway cells can be found, including from our own laboratories. However, in practice, although a method might work well for one gene in one set of studies, it is not necessarily an indication that it will be applicable for a different gene in separate experiments. Furthermore, a method that seems to work efficiently with cells from one human donor might suddenly not work effectively when attempted in cells from another donor. This is true for all types of technologies, including the use of 1) viral vectors (lentivirus, adenovirus, and/or retroviral) to express knockdown components, such as sh/siRNAs, CRISPR, and/or anti-sense species; 2) standard lipofection methods; and 3) electroporation. An easy-to-use, robust, economical, versatile, and efficient method to knock down genes in primary human airway cells would represent an important advance in the field. As a step forward, the article by Koh and colleagues (pp. 373–381) in this issue of the Journal (1) describes the optimization of a method for targeting genes in primary human bronchial epithelial cells (HBECs). They used electroporation of the necessary components for CRISPR-mediated deletion (guide RNA [gRNA] sequences and recombinant Cas9 complexes) in HBECs, which was nicely demonstrated using a well-described model of the regulation of MUC5AC (mucin 5AC) by SPEDF (SAM Pointed Domain Containing ETS Transcription Factor) (2, 3). Knockdown of SPEDF is predicted from the literature to dramatically reduce the upregulation of MUC5AC expression resulting from treatment of HBECs with IL-13, a central mediator of allergic asthma. Applying a variety of experimental conditions, the authors were able to demonstrate near-complete loss of SPEDF in HBEC cultures using their electroporation method. Targeting of SPDEF with this technology was accompanied by the expected loss of IL-13–mediated 1) upregulation of MUC5AC and downregulation of MUC5B expression, 2) induction of goblet cell differentiation, and 3) impairment of mucociliary clearance. Although the science was mostly confirmatory of previous studies regarding SPDEF-dependent function, the optimized method provides a useful paradigm for other laboratories to follow. Although the usual caveats with CRISPRs apply (e.g., gRNAs do not always work as predicted), Koh and colleagues were able to demonstrate that gRNAs shown to be efficient in relevant cell line models are also effective in HBECs after electroporation of the CRISPR/gRNA complexes (1). Prescreening of gRNA sequences is still recommended. Once a gRNA is identified, it is synthesized and simply mixed with the recombinant Cas9 to form a complex in the buffer recommended for electroporation. Varying the timing of the electroporation and the concentrations of the components altered the efficiency in predictable ways, suggesting that the degree of gene knockdown could be manipulated to the experimenter’s advantage. Hence, the method might be also useful for studies evaluating the relationship between the degree of gene expression and functional responses. Importantly, a similar method has also recently been described by Rapiteanu and colleagues (4), indicating that this approach can be robust across laboratories; indeed, we are finding similar usefulness of the technique in our own laboratories. Nevertheless, the target gene still seems to matter, and success may remain out of reach for difficult-to-target genes whose functions are necessary to maintain the integrity of the HBECs as they differentiate in culture. The technique still has some disadvantages, including cost, and time will tell if it becomes a standard method for studying airway pathophysiology. Nonetheless, the method optimized by Koh and colleagues (1) significantly contributes to the growing arsenal of new technologies that will drive the pulmonary biology field forward.

          Related collections

          Most cited references4

          • Record: found
          • Abstract: found
          • Article: not found

          SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production.

          Various acute and chronic inflammatory stimuli increase the number and activity of pulmonary mucus-producing goblet cells, and goblet cell hyperplasia and excess mucus production are central to the pathogenesis of chronic pulmonary diseases. However, little is known about the transcriptional programs that regulate goblet cell differentiation. Here, we show that SAM-pointed domain-containing Ets-like factor (SPDEF) controls a transcriptional program critical for pulmonary goblet cell differentiation in mice. Initial cell-lineage-tracing analysis identified nonciliated secretory epithelial cells, known as Clara cells, as the progenitors of goblet cells induced by pulmonary allergen exposure in vivo. Furthermore, in vivo expression of SPDEF in Clara cells caused rapid and reversible goblet cell differentiation in the absence of cell proliferation. This was associated with enhanced expression of genes regulating goblet cell differentiation and protein glycosylation, including forkhead box A3 (Foxa3), anterior gradient 2 (Agr2), and glucosaminyl (N-acetyl) transferase 3, mucin type (Gcnt3). Consistent with these findings, levels of SPDEF and FOXA3 were increased in mouse goblet cells after sensitization with pulmonary allergen, and the proteins were colocalized in goblet cells lining the airways of patients with chronic lung diseases. Deletion of the mouse Spdef gene resulted in the absence of goblet cells in tracheal/laryngeal submucosal glands and in the conducting airway epithelium after pulmonary allergen exposure in vivo. These data show that SPDEF plays a critical role in regulating a transcriptional network mediating the goblet cell differentiation and mucus hyperproduction associated with chronic pulmonary disorders.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            SPDEF regulates goblet cell hyperplasia in the airway epithelium.

            Goblet cell hyperplasia and mucous hypersecretion contribute to the pathogenesis of chronic pulmonary diseases including cystic fibrosis, asthma, and chronic obstructive pulmonary disease. In the present work, mouse SAM pointed domain-containing ETS transcription factor (SPDEF) mRNA and protein were detected in subsets of epithelial cells lining the trachea, bronchi, and tracheal glands. SPDEF interacted with the C-terminal domain of thyroid transcription factor 1, activating transcription of genes expressed selectively in airway epithelial cells, including Sftpa, Scgb1a1, Foxj1, and Sox17. Expression of Spdef in the respiratory epithelium of adult transgenic mice caused goblet cell hyperplasia, inducing both acidic and neutral mucins in vivo, and stainined for both acidic and neutral mucins in vivo. SPDEF expression was increased at sites of goblet cell hyperplasia caused by IL-13 and dust mite allergen in a process that was dependent upon STAT-6. SPDEF was induced following intratracheal allergen exposure and after Th2 cytokine stimulation and was sufficient to cause goblet cell differentiation of Clara cells in vivo.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Efficient RNP-directed Human Gene Targeting Reveals SPDEF Is Required for IL-13–induced Mucostasis

              Primary human bronchial epithelial cell (HBEC) cultures are a useful model for studies of lung health and major airway diseases. However, mechanistic studies have been limited by our ability to selectively disrupt specific genes in these cells. Here we optimize methods for gene targeting in HBECs by direct delivery of single guide RNA (sgRNA) and rCas9 (recombinant Cas9) complexes by electroporation, without a requirement for plasmids, viruses, or antibiotic selection. Variations in the method of delivery, sgRNA and rCas9 concentrations, and sgRNA sequences all had effects on targeting efficiency, allowing for predictable control of the extent of gene targeting and for near-complete disruption of gene expression. To demonstrate the value of this system, we targeted SPDEF , which encodes a transcription factor previously shown to be essential for the differentiation of MUC5AC-producing goblet cells in mouse models of asthma. Targeting SPDEF led to proportional decreases in MUC5AC expression in HBECs stimulated with IL-13, a central mediator of allergic asthma. Near-complete targeting of SPDEF abolished IL-13–induced MUC5AC expression and goblet cell differentiation. In addition, targeting of SPDEF prevented IL-13–induced impairment of mucociliary clearance, which is likely to be an important contributor to airway obstruction, morbidity, and mortality in asthma. We conclude that direct delivery of sgRNA and rCas9 complexes allows for predictable and efficient gene targeting and enables mechanistic studies of disease-relevant pathways in primary HBECs.
                Bookmark

                Author and article information

                Journal
                Am J Respir Cell Mol Biol
                Am. J. Respir. Cell Mol. Biol
                ajrcmb
                American Journal of Respiratory Cell and Molecular Biology
                American Thoracic Society
                1044-1549
                1535-4989
                March 2020
                March 2020
                March 2020
                : 62
                : 3
                : 279-280
                Affiliations
                [ 1 ]Department of Medicine
                [ 2 ]Cystic Fibrosis Research Center

                The University of North Carolina at Chapel Hill

                Chapel Hill, North Carolina

                and
                [ 3 ]Department of Medicine

                Department of Cell Biology and Physiology
                Article
                2019-0360ED
                10.1165/rcmb.2019-0360ED
                7055690
                31633992
                dc8b6a60-ed06-47a0-b13a-0cfc9835bb30
                Copyright © 2020 by the American Thoracic Society

                This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial No Derivatives License 4.0 ( http://creativecommons.org/licenses/by-nc-nd/4.0/). For commercial usage and reprints, please contact Diane Gern ( dgern@ 123456thoracic.org ).

                History
                Page count
                Figures: 0, Tables: 0, Pages: 2
                Categories
                Editorials

                Comments

                Comment on this article