2,181
Views
16
CrossRef citations to date
0
Altmetric
Editorial

CLCA1 and TMEM16A: the link towards a potential cure for airway diseases

Abstract

The hallmark traits of chronic obstructive airway diseases are inflammation, airway constriction due to hyperreactivity and mucus overproduction. The current common treatments for asthma and chronic obstructive pulmonary disease target the first two traits with none currently targeting mucus overproduction. The main source of obstructive mucus production is mucus cell metaplasia (MCM), the transdifferentiation of airway epithelial cells into mucus-producing goblet cells, in the small airways. Our current understanding of MCM is profusely incomplete. Few of the molecular players involved in driving MCM in humans have been identified and for many of those that have, their functions and mechanisms are unknown. This fact has limited the development of therapeutics that target mucus overproduction by inhibiting MCM. Current work in the field is aiming to change that.

The main source of obstructive mucus production is mucus cell metaplasia (MCM), the transdifferentiation of airway epithelial cells into mucus-producing goblet cells, in the small airways. MCM results in the upregulated expression of mucin genes that is driven by inflammatory mediators Citation[1–3]. MCM is triggered by IL-13 and IL-4, TH2 cytokines produced by inflammatory immune cells in response to allergens, pathogens and other insults Citation[4–6]. Consequently, asthma therapeutics targeting IL-13, for example, anti-IL-13 antibodies (lebrikizumab and tralokinumab) and anti-IL-13 receptor antibodies (dupilumab), have been developed and have reached clinical trials. Some have been shown partially effective within some subgroups Citation[7,8] or to have no effect on severe asthmatics Citation[9]. Additionally, initial studies also reported some adverse side effects associated with the use of these therapeutics, likely because of the fact that IL-13 also plays immunoregulatory roles. For example, IL-13 suppresses TH17 cytokine production in an IL-10-dependent manner and thus may play a role in TH17-associated autoimmune diseases. Accordingly, some patients who were administered IL-13-targeted biologics had significantly increased detrimental events involving musculoskeletal and autoimmune-related diseases Citation[7,10]. These observations give rise to three main points: targeting the IL-13 pathway could be effective for treating asthma and chronic obstructive pulmonary disease (COPD); 2) IL-13 may not be the optimal target since it is involved in numerous important processes and 3) identifying therapeutically manipulatable targets specific to the MCM pathway and understanding their mechanism of function is crucial.

Towards that end, recent work in this field has identified a number of new potential molecular targets in this pathway for the treatment of chronic inflammatory airways diseases (e.g., SPDEF Citation[11], MARCKS Citation[12], etc.). In this editorial, I will focus on three of them, each of which represents a different molecular class: a kinase (MAPK13/p38δ); a calcium-activated chloride channel regulator 1 (CLCA1) and a calcium-activated chloride channel (TMEM16A), paying particular attention to the emerging importance of the last two.

CLCA1, the human ortholog of mouse CLCA3/Gob-5, has had a long and highly undefined association with chronic inflammatory airway diseases. The expression of CLCA1 is highly upregulated in response to IL-13 in both mouse Citation[13] and human models Citation[14,15] of MCM. It is also highly expressed in COPD airways Citation[15]. A factor contributing to the confusion regarding CLCA1 function has been observations from Clca3 (the mouse ortholog of CLCA1) knockout mice, which suggest that it does not play a role in IL-13-induced mucus overproduction or Cl secretion Citation[13,16]. This is likely due to functional redundancy due to the fact that mice encode at least seven different CLCA proteins Citation[13,17]. The situation in humans appears to be much simpler, as they encode only three CLCA proteins. Overexpression of CLCA1 alone in human airway epithelial cells can drive mucus production, and siRNA knockdown of CLCA1 in primary airway cell models of IL-13-induced MCM prevents mucus production Citation[15]. This puts CLCA1 function in the middle of the IL-13 to mucus pathway. CLCA1 is a secreted protein which can be found in isolated mucin granules Citation[18] and is proteolytically cleaved into two fragments during secretion Citation[19]. This secreted protein can be detected in bronchoalveolar lavage fluid from asthmatic patients Citation[20], and recently it was demonstrated that CLCA1 protein levels in sputum are an accurate biomarker of asthma endotypes (either TH2-low or TH2-high) Citation[21].

Although the expression of CLCA1 has been associated with airway diseases, understanding the role that it plays in development of disease has lagged far behind. This is largely due to the fact that CLCA1 belongs to a unique family of proteins whose function and mechanism of action were largely unknown until recently. Originally identified over 20 years ago, CLCA proteins were originally annotated as calcium-activated chloride channels since cells display increased calcium-dependent chloride currents when these proteins are expressed. However, subsequent in vivo and in vitro studies indicated that these proteins are largely secreted soluble proteins that do not contain the numerous transmembrane domains required to form a channel Citation[16,17,20] and subsequent analysis indicated these were general properties of the CLCA family Citation[17]. Consequently, it was demonstrated that CLCA1 was able to mediate calcium-dependent chloride currents by activating an endogenous calcium-activated chloride channel in human cells Citation[19,22].

How then does CLCA1 activate calcium-activated chloride channels (CaCCs)? We recently demonstrated that full-length CLCA1 is unable to activate CaCCs and that cleavage of the protein is required to activate it Citation[19]. The protease responsible is a novel matrix metalloprotease-like domain located within the N-terminus of CLCA1 itself. This self-cleavage then allows the N-terminal fragment of CLCA1 to interact with and activate its target CaCC. What is the identity of that target CaCC and how does CLCA1 activate it? We recently elucidated that CLCA1 activates the CaCC TMEM16A and that it can do so in a paracrine fashion Citation[23]. This last point is important, given the observation that secreted CLCA1 is highly abundant under inflammatory conditions in the airway. CLCA1 directly engages TMEM16A on the surface of cells, and this interaction appears to stabilize the surface expression of TMEM16A, increasing the number of channels on the surface, and consequently increasing the overall cellular currents through these channels.

These recent exciting results have uncovered many details regarding the function of CLCA1 and how it activates calcium-dependent chloride currents through TMEM16A. Several questions remain however, with the most prominent ones regarding whether or not the CLCA1-TMEM16A interaction contributes to the development of chronic inflammatory airways diseases, and if so, how? There are several lines of evidence that would suggest that these two molecules act synergistically. For example, the expression of both CLCA1 and TMEM16A is upregulated by the inflammatory cytokines IL-4 and IL-13, with this expression largely limited to goblet cells in the airway Citation[15,24,25]. In addition, both CLCA1 and TMEM16A have been tied to mucus production in some way. As mentioned above, overexpression of CLCA1 appears to be central to mucus overproduction. Likewise, TMEM16A activity has been linked to mucus secretion, as TMEM16A inhibitors block ATP-stimulated mucus secretion in IL-13-treated human bronchial epithelial cells Citation[26]. What is unknown at this time, however, is whether or not the CLCA1-TMEM16A interaction plays a central or an auxiliary role in MCM. We previously observed that IL-13-driven overexpression of CLCA1 in human airway epithelial cells activates a signaling cascade involving MAPK13 and that inhibitors to MAPK13 developed by structure-based drug design are able to block the IL-13-driven mucus production Citation[15,27]. It is unknown at this time whether or not the CLCA1-mediated activation of TMEM16A directly drives the activation of MAPK13 and the subsequent signaling cascade leading to the expression of MUC5AC, or whether this function of CLCA1 is just an important auxiliary function in the pathway. It is well known that anion channel activity is required for secreted mucins (MUC5AC and MUC5B) to function properly in a mucosal immunity and mucocilliary clearance capacity Citation[28]. Mucin proteins are secreted in dense, dehydrated granules and require anion channel passage of chloride and bicarbonate ions to ensure proper hydration, salination and pH control Citation[29]. This is exemplified by the disease cystic fibrosis, where the impaired function of the anion channel CFTR produces mucus that does not function properly and causes disease. In this regard, TMEM16A has been shown to carry both chloride and bicarbonate anions Citation[30]. Thus, regardless of whether or not the CLCA1-TMEM16A interaction is required within the signaling pathway driving MCM, it more than likely does play a role in ensuring that the mucus produced will function properly. This would be similar to the role that has recently been attributed to the Cl channel SLC26A9, whose activity is increased by IL-13 stimulation and is essential to prevent mucus plugging in the setting of MCM Citation[31].

In conclusion, we have learned much recently regarding the function of CLCA1 and mechanistically how it activates chloride currents through TMEM16A. However, much more information will be required before one can develop asthma and COPD treatments that target this duo. First and foremost among those is defining the role of the CLCA1-TMEM16A interaction within the MCM pathway, that is does it drive mucus production and disease, or does it solely play a protective role in contributing to improved mucus hydration and clearance. In addition, more structural data for both proteins is required. Currently, there are no structural data for any portion of CLCA1, and although the groundbreaking crystal structure of a fungal ortholog of TMEM16A was recently reported and provided several enormous insights into the structure and function of this protein Citation[32], the structure of a more highly related mammalian ortholog will be needed to effectively design specific TMEM16A-targeting drugs. Furthermore, the precise molecular determinants that govern the CLCA1-TMEM16A interaction remain to be determined. These future studies will determine if the CLCA1-TMEM16A interaction should be targeted therapeutically for asthma and COPD, and if so, how.

Financial & competing interests disclosure

This work was supported by NIH R01-HL119813, American Lung Association RG-196051 and a CIMED Pilot and Feasibility grant received by TJ Brett. The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

References

  • Rose MC, Voynow JA. Respiratory tract mucin genes and mucin glycoproteins in health and disease. Physiol Rev 2006;86:245-78
  • Thai P, Loukoianov A, Wachi S, Wu R. Regulation of airway mucin gene expression. Annu Rev Physiol 2008;70:405-29
  • Evans CM, Koo JS. Airway mucus: the good, the bad, the sticky. Pharmacol Ther 2009;121:332-48
  • Grunig G, Warnock M, Wakil AE, et al. Requirement for IL-13 independently of IL-4 in experimental asthma. Science 1998;282:2261-3
  • Wills-Karp M, Luyimbazi J, Xu X, et al. Interleukin-13: central mediator of allergic asthma. Science 1998;282:2258-61
  • Kuperman D A, Huang X, Koth LL, et al. Direct effects of interleukin-13 on epithelial cells cause airway hyperreactivity and mucus overproduction in asthma. Nat Med 2002;8:885-9
  • Corren J, Lemanske RF, Hanania NA, et al. Lebrikizumab treatment in adults with asthma. N Engl J Med 2011;365:1088-98
  • Gauvreau GM, Boulet LP, Cockcroft DW, et al. Effects of interleukin-13 blockade on allergen-induced airway responses in mild atopic asthma. Am J Respir Crit Care Med 2011;183:1007-14
  • De Boever EH, Ashman C, Cahn AP, et al. Efficacy and safety of an anti-IL-13 mAb in patients with severe asthma: a randomized trial. J Allergy Clin Immunol 2014;133:989-96
  • Singh D, Kane B, Molfino NA, et al. A phase 1 study evaluating the pharmacokinetics, safety and tolerability of repeat dosing with a human IL-13 antibody (CAT-354) in subjects with asthma. BMC Pulm Med 2010;10:3
  • Chen G, Korfhagen TR, Xu Y, et al. SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J Clin Invest 2009;119:2914-24
  • Singer M, Martin LD, Vargaftig BB, et al. A MARCKS-related peptide blocks mucus hypersecretion in a mouse model of asthma. Nat Med 2004;10:193-6
  • Patel AC, Morton JD, Kim EY, et al. Genetic segregation of airway disease traits despite redundancy of calcium-activated chloride channel family members. Physiol Genomics 2006;25:502-13
  • Woodruff PG, Boushey HA, Dolganov GM, et al. Genome-wide profiling identifies epithelial cell genes associated with asthma and with treatment response to corticosteroids. Proc Natl Acad Sci USA 2007;104:15858-63
  • Alevy YG, Patel AC, Romero AG, et al. IL-13-induced airway mucus production is attenuated by MAPK13 inhibition. J Clin Invest 2012;122:4555-68
  • Mundhenk L, Johannesson B, Anagnostopoulou P, et al. mCLCA3 does not contribute to calcium-activated chloride conductance in murine airways. Am J Respir Cell Mol Biol 2012;47:87-93
  • Patel AC, Brett TJ, Holtzman MJ. The role of CLCA proteins in inflammatory airway disease. Annu Rev Physiol 2009;71:425-49
  • Raiford KL, Park J, Lin KW, et al. Mucin granule-associated proteins in human bronchial epithelial cells: the airway goblet cell “granulome”. Respir Res 2011;12:118
  • Yurtsever Z, Sala-Rabanal M, Randolph DT, et al. Self-cleavage of human CLCA1 protein by a novel internal metalloprotease domain controls calcium-activated chloride channel activation. J Biol Chem 2012;287:42138-49
  • Gibson A, Lewis AP, Affleck K, et al. hCLCA1 and mCLCA3 are secreted non-integral membrane proteins and therefore are not ion channels. J Biol Chem 2005;280:27205-12
  • Peters MC, Mekonnen ZK, Yuan S, et al. Measures of gene expression in sputum cells can identify TH2-high and TH2-low subtypes of asthma. J Allergy Clin Immunol 2014;133:388-94
  • Hamann M, Gibson A, Davies N, et al. Human ClCa1 modulates anionic conduction of calcium-dependent chloride currents. J Physiol 2009;587:2255-74
  • Sala-Rabanal M, Yurtsever Z, Nichols CG, Brett TJ. Secreted CLCA1 modulates TMEM16A to activate Ca(2+)-dependent chloride currents in human cells. Elife 2015;4
  • Caputo A, Caci E, Ferrera L, et al. TMEM16A, a membrane protein associated with calcium-dependent chloride channel activity. Science 2008;322:590-4
  • Scudieri P, Caci E, Bruno S, et al. Association of TMEM16A chloride channel overexpression with airway goblet cell metaplasia. J Physiol 2012;590:6141-55
  • Huang F, Zhang H, Wu M, et al. Calcium-activated chloride channel TMEM16A modulates mucin secretion and airway smooth muscle contraction. Proc Natl Acad Sci USA 2012;109:16354-9
  • Yurtsever Z, Scheaffer SM, Romero AG, et al. The crystal structure of phosphorylated MAPK13 reveals common structural features and differences in p38 MAPK family activation. Acta Crystallogr D Biol Crystallogr 2015;71:790-9
  • Ghosh A, Boucher RC, Tarran R. Airway hydration and COPD. Cell Mol Life Sci 2015. [Epub ahead of print]
  • Fahy JV, Dickey BF. Airway mucus function and dysfunction. N Engl J Med 2010;363:2233-47
  • Jung J, Nam JH, Park HW, et al. Dynamic modulation of ANO1/TMEM16A HCO3(-) permeability by Ca2+/calmodulin. Proc Natl Acad Sci USA 2013;110:360-5
  • Anagnostopoulou P, Riederer B, Duerr J, et al. SLC26A9-mediated chloride secretion prevents mucus obstruction in airway inflammation. J Clin Invest 2012;122:3629-34
  • Brunner JD, Lim NK, Schenck S, et al. X-ray structure of a calcium-activated TMEM16 lipid scramblase. Nature 2014;516:207-12

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.