Abstract

Editorial FocusDysregulation of ion transport in the lung epithelium infected with SARS-CoV-2Laura A. Dada, Olga Vagin, and Jacob I. SznajderLaura A. DadaNorthwestern University Feinberg School of Medicine, Chicago, IllinoisSearch for more papers by this author, Olga VaginDepartment of Physiology, David Geffen School of Medicine, University of California Los Angeles and Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, CaliforniaSearch for more papers by this author, and Jacob I. SznajderNorthwestern University Feinberg School of Medicine, Chicago, IllinoisCorrespondence: J. I. Sznajder ([email protected]).Search for more papers by this authorPublished Online:22 Jun 2021https://doi.org/10.1152/ajplung.00170.2021This is the final version - click for previous versionMoreSectionsPDF (262 KB)Download PDF ToolsExport citationAdd to favoritesGet permissionsTrack citations ShareShare onFacebookTwitterLinkedInWeChat As of April 14, 2021, ∼138 million people worldwide and ∼31 million in the United States have been diagnosed with COVID-19 disease, and ∼3 million people worldwide and ∼565,000 people in the United States have died (https://coronavirus.jhu.edu). Most patients with SARS-CoV-2 infection are asymptomatic or have mild disease; however, some patients develop severe pneumonia and acute respiratory distress syndrome (ARDS) requiring mechanical ventilation and even lung transplant (1–5). In some of these patients, the disease triggers an exuberant inflammatory condition, termed “cytokine storm” due to dysregulated immune response, which can lead to multi-organ failure and increased fatality rates (6, 7).Pulmonary edema is a hallmark of ARDS, characterized by disruption of the alveolo-capillary barrier and flooding of the airspaces with proteinaceous fluid and inflammatory cells (8–10). The upper and lower respiratory epithelium are lined by a thin fluid layer, referred to as airway surface liquid and alveolar lining fluid, respectively (11). Their composition is maintained by tightly regulated processes of secretion and absorption mediated by ion channels and pumps of the respiratory epithelial cells. Lung injury induces changes in the alveolar-capillary barrier, dysregulation of epithelial Na,K-ATPase, epithelial sodium channel (ENaC), and cystic fibrosis membrane conductance regulator (CFTR), leading to the accumulation of edema and impaired alveolar fluid clearance (11–16). Two recent articles have discussed the role of transepithelial ion transport during SARS-CoV-2 infection of the respiratory tract.The first article by Abdel Hameid et al. (17) proposes that SARS-CoV-2 may alter evolutionary conserved second messenger signaling cascades via activation of G protein-coupled receptors (GPCRs) or by directly modulating G protein signaling. Based on the well-known reciprocal relationship between ENaC and CFTR (18, 19), the authors speculate that stimulation of GPCR signaling leads to activation of CFTR-mediated Cl− transport, which may overwhelm absorptive pathways such as ENaC-dependent Na+ uptake. This process would trigger a pathophysiological cascade, leading to accumulation of lung edema, which is observed in the more severe cases of patients with COVID-19 and ARDS. CFTR is regulated via the cAMP/PKA pathway (20), a pathway known to be hijacked by microorganisms, such as cholera toxin, which by activating the adenylate cyclase triggers chloride secretion through CFTR (21). The authors also propose a role for exchange protein directly activated by cAMP (EPAC1) pathway, which is an alternative cAMP effector that interacts with CFTR through the Na+/H+ exchanger regulatory factor 1 (NHERF1) (22). The EPAC1 pathway has been reported to play a role in MERS-CoV and SARS-CoV infections (23). However, it is well known that viral infections cause ENaC inhibition by mechanisms not involving GPCR activation. For example, the influenza M2 protein, which functions as a proton-conducting channel, decreases ENaC and CFTR activity by causing the degradation of these transport proteins (24). In this case, only the ENaC would contribute to the dysregulation of fluid homeostasis. The authors acknowledge the speculative nature of their reasoning as they do not show any relation between SARS-CoV-2 infection and the function and/or expression levels of CFTR.The study by Kryvenko and Vadász (25) focuses on the effects of lung injury, including SARS-CoV-2 infection, on Na,K-ATPase downregulation. There is significant evidence that links the downregulation of the Na,K-ATPase to the disruption of the alveolar barrier in models of lung injury since this ion transporter and adhesion molecule is required for normal alveolar epithelial function (12, 15, 16, 26–28). Therefore, the authors propose that the decrease of Na,K-ATPase abundance at the plasma membrane of alveolar epithelial cells contributes to the impairment of alveolar epithelial function due to SARS-CoV-2-infection. Furthermore, the authors propose that the disruption of the alveolo-capillary barrier leads to persistence of lung injury, which contributes to the extrapulmonary manifestations of COVID-19. Several publications report decreases in mRNA and total protein level of Na,K-ATPase subunits in SARS-CoV-2-infected cells and in postmortem lung tissue samples from patients with COVID-19 (29–32). These data suggest decreased transcription and translation of the Na,K-ATPase. Moreover, the authors present a convincing analysis of the cellular processes affected by SARS-CoV-2 infection, which impair the maturation of Na,K-ATPase molecules and their delivery to the cell plasma membrane. Particularly, data reporting that SARS-CoV-2 infection causes ER stress (32–35) suggest impairment of chaperone-assisted folding of transmembrane proteins, including the Na,K-ATPase, in the ER lumen. In addition, the SARS-CoV-2 spike protein is extensively glycosylated and highjacks the host glycosylation and glycan-dependent protein folding machinery, which could disrupt the Na,K-ATPase maturation that critically depends on the glycosylation of one of its subunits (36). Also, SARS-CoV-2 infection interferes with signaling cascades that normally regulate the plasma membrane Na,K-ATPase abundance by promoting its endocytosis.Both articles discuss potential contributions of the ion transport proteins to the pathophysiology of acute lung injury and respiratory distress in patients with severe COVID-19 disease. However, they fail to discuss that in severe cases of SARS-CoV-2, the cytokine storm may lead to increased cell death causing a “leaky” epithelium (37, 38). It has been described that cytokines such as IL-1β, IL-6, and TNFα, are increased in the lungs of patients with SARS-CoV-2 and may lead to destabilization of CFTR, ENaC, and Na,K-ATPase (37). Along with these data, it has been reported that dexamethasone is associated with improved outcomes in patients with COVID-19 disease (39). Dexamethasone is known to upregulate ion transport proteins, including ENaC, CFTR, and the Na,K-ATPase (40–42), suggesting a role for ion-transport mechanisms in the pathophysiology and outcome of patients with ARDS due to SARS-CoV-2 infection. As mentioned above, during viral pneumonitis and ARDS, the alveolar epithelial barrier is disrupted by overwhelming epithelial cell death and also by dysregulation of the lung epithelial ion transport (43). We reason that SARS-CoV-2 infection alters the cellular processes that impair the function of ion transport proteins, such as CFTR, ENaC, and the Na,K-ATPase, which results in hypoxia and hypercapnia, further impairing ion-transport mechanisms (37, 38).Although not much data are available on the mechanisms regulating the repair of the lungs after the acute phase of COVID-19, they are probably similar to lung repair after influenza pneumonitis or other severe causes of lung injury where ion-transport mechanisms are paramount. If the lung is not overwhelmingly injured, necessitating lung transplant, the alveolar epithelium must regain normal function after SARS-CoV-2-induced injury for the patients to survive.GRANTSThis work was supported by the National Institutes of Health (NIH) National Heart, Lung, and Blood Institute Grants HL-147070, HL-71643, and HL-154998 (to L.A.D. and J.I.S.) and the NIH National Institute on Aging Grant PO1-AG049665 (to L.A.D. and J.I.S.).DISCLOSURESNo conflicts of interest, financial or otherwise, are declared by the authors.AUTHOR CONTRIBUTIONSL.A.D., O.V., and J.I.S. drafted manuscript; edited and revised manuscript; and approved final version of manuscript.REFERENCES1. Bharat A, Querrey M, Markov NS, Kim S, Kurihara C, Garza-Castillon R, Manerikar A, Shilatifard A, Tomic R, Politanska Y, Abdala-Valencia H, Yeldandi AV, Lomasney JW, Misharin AV, Budinger GRS. Lung transplantation for patients with severe COVID-19. Sci Transl Med 12: eabe4282, 2020. doi:10.1126/scitranslmed.abe4282. Crossref | PubMed | ISI | Google Scholar2. Grant RA, Morales-Nebreda L, Markov NS, Swaminathan S, Querrey M, Guzman ER; The NU SCRIPT Study Investigators, et al. Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia. Nature 590: 635–641, 2021. doi:10.1038/s41586-020-03148-w. Crossref | PubMed | ISI | Google Scholar3. Mangalmurti NS, Reilly JP, Cines DB, Meyer NJ, Hunter CA, Vaughan AE. COVID-19–associated acute respiratory distress syndrome clarified: a vascular endotype? Am J Respir Crit Care Med 202: 750–753, 2020. doi:10.1164/rccm.202006-2598LE. Crossref | PubMed | ISI | Google Scholar4. Richardson S, Hirsch JS, Narasimhan M, Crawford JM, McGinn T, Davidson KW, Barnaby DP, Becker LB, Chelico JD, Cohen SL, Cookingham J, Coppa K, Diefenbach MA, Dominello AJ, Duer-Hefele J, Falzon L, Gitlin J, Hajizadeh N, Harvin TG, Hirschwerk DA, Kim EJ, Kozel ZM, Marrast LM, Mogavero JN, Osorio GA, Qiu M, Zanos TP; The Northwell COVID-19 Research Consortium. Presenting characteristics, comorbidities, and outcomes among 5700 patients hospitalized with COVID-19 in the New York City area. JAMA 323: 2052–2059, 2020 [Erratum in JAMA 323: 2098, 2020]. doi:10.1001/jama.2020.6775. Crossref | PubMed | ISI | Google Scholar5. Yang X, Yu Y, Xu J, Shu H, Xia J, Liu H, Wu Y, Zhang L, Yu Z, Fang M, Yu T, Wang Y, Pan S, Zou X, Yuan S, Shang Y. Clinical course and outcomes of critically ill patients with SARS-CoV-2 pneumonia in Wuhan, China: a single-centered, retrospective, observational study. Lancet Respir Med 8: 475–481, 2020 [Erratum in Lancet Respir Med 8: e26, 2020]. doi:10.1016/S2213-2600(20)30079-5. Crossref | PubMed | ISI | Google Scholar6. Mangalmurti N, Hunter CA. Cytokine storms: understanding COVID. Immunity 53: 19–25, 2020. doi:10.1016/j.immuni.2020.06.017. Crossref | PubMed | ISI | Google Scholar7. Soy M, Keser G, Atagündüz P, Tabak F, Atagündüz I, Kayhan S. Cytokine storm in COVID-19: pathogenesis and overview of anti-inflammatory agents used in treatment. Clin Rheumatol 39: 2085–2094, 2020. doi:10.1007/s10067-020-05190-5. Crossref | PubMed | ISI | Google Scholar8. Calfee CS, Delucchi K, Parsons PE, Thompson BT, Ware LB, Matthay MA; NHLBI ARDS Network. Subphenotypes in acute respiratory distress syndrome: latent class analysis of data from two randomised controlled trials. Lancet Respir Med 2: 611–620, 2014. doi:10.1016/S2213-2600(14)70097-9. Crossref | PubMed | ISI | Google Scholar9. Herrero R, Sanchez G, Lorente JA. New insights into the mechanisms of pulmonary edema in acute lung injury. Ann Transl Med 6: 32–32, 2018. doi:10.21037/atm.2017.12.18. Crossref | PubMed | ISI | Google Scholar10. Sznajder JI. Alveolar edema must be cleared for the acute respiratory distress syndrome patient to survive. Am J Respir Crit Care Med 163: 1293–1294, 2001. doi:10.1164/ajrccm.163.6.ed1801d. Crossref | PubMed | ISI | Google Scholar11. Peteranderl C, Sznajder JI, Herold S, Lecuona E. Inflammatory responses regulating alveolar ion transport during pulmonary infections. Front Immunol 8: 446, 2017. doi:10.3389/fimmu.2017.00446. Crossref | PubMed | ISI | Google Scholar12. Brazee PL, Morales-Nebreda L, Magnani ND, Garcia JG, Misharin AV, Ridge KM, Budinger GRS, Iwai K, Dada LA, Sznajder JI. Linear ubiquitin assembly complex regulates lung epithelial-driven responses during influenza infection. J Clin Invest 130: 1301–1314, 2020. doi:10.1172/JCI128368. Crossref | PubMed | ISI | Google Scholar13. Brazee PL, Soni PN, Tokhtaeva E, Magnani N, Yemelyanov A, Perlman HR, Ridge KM, Sznajder JI, Vagin O, Dada LA. FXYD5 Is an essential mediator of the inflammatory response during. Front Immunol 8: 623, 2017. doi:10.3389/fimmu.2017.00623. Crossref | PubMed | ISI | Google Scholar14. Mutlu GM, Adir Y, Jameel M, Akhmedov AT, Welch L, Dumasius V, Meng FJ, Zabner J, Koenig C, Lewis ER, Balagani R, Traver G, Sznajder JI, Factor P. Interdependency of beta-adrenergic receptors and CFTR in regulation of alveolar active Na+ transport. Circ Res 96: 999–1005, 2005. doi:10.1161/01.RES.0000164554.21993.AC. Crossref | PubMed | ISI | Google Scholar15. Peteranderl C, Morales-Nebreda L, Selvakumar B, Lecuona E, Vadasz I, Morty RE, Schmoldt C, Bespalowa J, Wolff T, Pleschka S, Mayer K, Gattenloehner S, Fink L, Lohmeyer J, Seeger W, Sznajder JI, Mutlu GM, Budinger GR, Herold S. Macrophage-epithelial paracrine crosstalk inhibits lung edema clearance during influenza infection. J Clin Invest 126: 1566–1580, 2016. doi:10.1172/JCI83931. Crossref | PubMed | ISI | Google Scholar16. Vadasz I, Sznajder JI. Gas exchange disturbances regulate alveolar fluid clearance during acute lung injury. Front Immunol 8: 757, 2017. doi:10.3389/fimmu.2017.00757. Crossref | PubMed | ISI | Google Scholar17. Abdel Hameid R, Cormet-Boyaka E, Kuebler WM, Uddin M, Berdiev BK. SARS-CoV-2 may hijack GPCR signaling pathways to dysregulate lung ion and fluid transport. Am J Physiol Lung Cell Mol Physiol 320: L430–L435, 2021. doi:10.1152/ajplung.00499.2020. Link | ISI | Google Scholar18. Matalon S, Bartoszewski R, Collawn JF. Role of epithelial sodium channels in the regulation of lung fluid homeostasis. Am J Physiol Lung Cell Mol Physiol 309: L1229–L1238, 2015. doi:10.1152/ajplung.00319.2015. Link | ISI | Google Scholar19. Stutts MJ, Canessa CM, Olsen JC, Hamrick M, Cohn JA, Rossier BC, Boucher RC. CFTR as a cAMP-dependent regulator of sodium channels. Science 269: 847–850, 1995. doi:10.1126/science.7543698. Crossref | PubMed | ISI | Google Scholar20. Monterisi S, Casavola V, Zaccolo M. Local modulation of cystic fibrosis conductance regulator: cytoskeleton and compartmentalized cAMP signalling. Br J Pharmacol 169: 1–9, 2013. doi:10.1111/bph.12017. Crossref | PubMed | ISI | Google Scholar21. Barrett KE. Rethinking cholera pathogenesis- no longer all in the same “camp”. Virulence 7: 751–753, 2016. doi:10.1080/21505594.2016.1212156. Crossref | PubMed | ISI | Google Scholar22. Lobo MJ, Amaral MD, Zaccolo M, Farinha CM. EPAC1 activation by cAMP stabilizes CFTR at the membrane by promoting its interaction with NHERF1. J Cell Sci 129: 2599–2612, 2016. doi:10.1242/jcs.185629. Crossref | PubMed | ISI | Google Scholar23. Tao X, Mei F, Agrawal A, Peters CJ, Ksiazek TG, Cheng X, Tseng CT. Blocking of exchange proteins directly activated by cAMP leads to reduced replication of Middle East respiratory syndrome coronavirus. J Virol 88: 3902–3910, 2014. doi:10.1128/JVI.03001-13. Crossref | PubMed | ISI | Google Scholar24. Lazrak A, Iles KE, Liu G, Noah DL, Noah JW, Matalon S. Influenza virus M2 protein inhibits epithelial sodium channels by increasing reactive oxygen species. FASEB J 23: 3829–3842, 2009. doi:10.1096/fj.09-135590. Crossref | PubMed | ISI | Google Scholar25. Kryvenko V, Vadász I. Molecular mechanisms of Na,K-ATPase dysregulation driving alveolar epithelial barrier failure in severe COVID-19. Am J Physiol Lung Cell Mol Physiol. In press. doi:10.1152/ajplung.00056.2021. Link | ISI | Google Scholar26. Dada LA, Chandel NS, Ridge KM, Pedemonte C, Bertorello AM, Sznajder JI. Hypoxia-induced endocytosis of Na,K-ATPase in alveolar epithelial cells is mediated by mitochondrial reactive oxygen species and PKC-zeta. J Clin Invest 111: 1057–1064, 2003. doi:10.1172/JCI16826. Crossref | PubMed | ISI | Google Scholar27. Tokhtaeva E, Sun H, Deiss-Yehiely N, Wen Y, Soni PN, Gabrielli NM, Marcus EA, Ridge KM, Sachs G, Vazquez-Levin M, Sznajder JI, Vagin O, Dada LA. The O-glycosylated ectodomain of FXYD5 impairs adhesion by disrupting cell-cell trans-dimerization of Na,K-ATPase beta1 subunits. J Cell Sci 129: 2394–2406, 2016. doi:10.1242/jcs.186148. Crossref | PubMed | ISI | Google Scholar28. Vagin O, Dada LA, Tokhtaeva E, Sachs G. The Na,K-ATPase alpha1beta1 heterodimer as a cell adhesion molecule in epithelia. Am J Physiol Cell Physiol 302: C1271–C1281, 2012. doi:10.1152/ajpcell.00456.2011. Link | ISI | Google Scholar29. Blanco-Melo D, Nilsson-Payant BE, Liu WC, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, Wang TT, Schwartz RE, Lim JK, Albrecht RA, tenOever BR. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181: 1036–1045.e9, 2020. doi:10.1016/j.cell.2020.04.026. Crossref | PubMed | ISI | Google Scholar30. Bojkova D, Klann K, Koch B, Widera M, Krause D, Ciesek S, Cinatl J, Münch C. Proteomics of SARS-CoV-2-infected host cells reveals therapy targets. Nature 583: 469–472, 2020. doi:10.1038/s41586-020-2332-7. Crossref | PubMed | ISI | Google Scholar31. Chen F, Zhang Y, Sucgang R, Ramani S, Corry D, Kheradmand F, Creighton CJ. Meta-analysis of host transcriptional responses to SARS-CoV-2 infection reveals their manifestation in human tumors. Sci Rep 11: 2459, 2021. doi:10.1038/s41598-021-82221-4. Crossref | PubMed | ISI | Google Scholar32. Jha PK, Vijay A, Halu A, Uchida S, Aikawa M. Gene expression profiling reveals the shared and distinct transcriptional signatures in human lung epithelial cells infected with SARS-CoV-2, MERS-CoV, or SARS-CoV: potential implications in cardiovascular complications of COVID-19. Front Cardiovasc Med 7: 623012, 2021. doi:10.3389/fcvm.2020.623012. Crossref | PubMed | ISI | Google Scholar33. Banerjee AK, Blanco MR, Bruce EA, Honson DD, Chen LM, Chow A, Bhat P, Ollikainen N, Quinodoz SA, Loney C, Thai J, Miller ZD, Lin AE, Schmidt MM, Stewart DG, Goldfarb D, De Lorenzo G, Rihn SJ, Voorhees RM, Botten JW, Majumdar D, Guttman M. SARS-CoV-2 disrupts splicing, translation, and protein trafficking to suppress host defenses. Cell 183: 1325–1339.e21, 2020. doi:10.1016/j.cell.2020.10.004. Crossref | PubMed | ISI | Google Scholar34. Kamura T, Sato S, Iwai K, Czyzyk-Krzeska M, Conaway RC, Conaway JW. Activation of HIF1alpha ubiquitination by a reconstituted von Hippel-Lindau (VHL) tumor suppressor complex. Proc Natl Acad Sci USA 97: 10430–10435, 2000. doi:10.1073/pnas.190332597. Crossref | PubMed | ISI | Google Scholar35. Siu KL, Chan CP, Kok KH, Woo PC, Jin DY. Comparative analysis of the activation of unfolded protein response by spike proteins of severe acute respiratory syndrome coronavirus and human coronavirus HKU1. Cell Biosci 4: 3, 2014. doi:10.1186/2045-3701-4-3. Crossref | PubMed | ISI | Google Scholar36. Watanabe Y, Allen JD, Wrapp D, McLellan JS, Crispin M. Site-specific glycan analysis of the SARS-CoV-2 spike. Science 369: 330–333, 2020. doi:10.1126/science.abb9983. Crossref | PubMed | ISI | Google Scholar37. Olbei M, Hautefort I, Modos D, Treveil A, Poletti M, Gul L, Shannon-Lowe CD, Korcsmaros T. SARS-CoV-2 causes a different cytokine response compared to other cytokine storm-causing respiratory viruses in severely ill patients. Front Immunol 12: 629193, 2021. doi:10.3389/fimmu.2021.629193. Crossref | PubMed | ISI | Google Scholar38. Song P, Li W, Xie J, Hou Y, You C. Cytokine storm induced by SARS-CoV-2. Clin Chim Acta 509: 280–287, 2020. doi:10.1016/j.cca.2020.06.017. Crossref | PubMed | ISI | Google Scholar39. Johnson RM, Vinetz JM. Dexamethasone in the management of COVID-19. BMJ 370: m2648, 2020. doi:10.1136/bmj.m2648. Crossref | PubMed | Google Scholar40. Barquin N, Ciccolella DE, Ridge KM, Sznajder JI. Dexamethasone upregulates the Na-K-ATPase in rat alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 273: L825–L830, 1997. doi:10.1152/ajplung.1997.273.4.L825. Link | ISI | Google Scholar41. Dagenais A, Fréchette R, Clermont ME, Massé C, Privé A, Brochiero E, Berthiaume Y. Dexamethasone inhibits the action of TNF on ENaC expression and activity. Am J Physiol Lung Cell Mol Physiol 291: L1220–L1231, 2006. doi:10.1152/ajplung.00511.2005. Link | ISI | Google Scholar42. Prota LFM, Cebotaru L, Cheng J, Wright J, Vij N, Morales MM, Guggino WB. Dexamethasone regulates CFTR expression in Calu-3 cells with the involvement of chaperones HSP70 and HSP90. PLoS One 7: e47405, 2012. doi:10.1371/journal.pone.0047405. Crossref | PubMed | ISI | Google Scholar43. Short KR, Kroeze EJ, Fouchier RA, Kuiken T. Pathogenesis of influenza-induced acute respiratory distress syndrome. Lancet Infect Dis 14: 57–69, 2014. doi:10.1016/S1473-3099(13)70286-X. Crossref | PubMed | ISI | Google ScholarAUTHOR NOTESCorrespondence: J. I. Sznajder ([email protected]edu). Download PDF Previous Back to Top Next FiguresReferencesRelatedInformation CollectionsAPS Cross-Journal CollectionsCoronavirus-Related Papers Related ArticlesSARS-CoV-2 may hijack GPCR signaling pathways to dysregulate lung ion and fluid transport 02 Mar 2021American Journal of Physiology-Lung Cellular and Molecular PhysiologyMolecular mechanisms of Na,K-ATPase dysregulation driving alveolar epithelial barrier failure in severe COVID-19 22 Jun 2021American Journal of Physiology-Lung Cellular and Molecular PhysiologyThe significance of a lack of rhinorrhea in severe coronavirus 19 lung disease 22 Jun 2021American Journal of Physiology-Lung Cellular and Molecular PhysiologyCited BySerially passaged, conditionally reprogrammed nasal epithelial cells as a model to study epithelial functions and SARS-CoV-2 infectionHanna Schmidt, Lara Guthjahr, Alexander Sauter, Fabian Zech, Rayhane Nchioua, Steffen Stenger, Manfred Frick, Frank Kirchhoff, Paul Dietl, and Oliver H. Wittekindt28 March 2022 | American Journal of Physiology-Cell Physiology, Vol. 322, No. 4Reply to EisenhutReem Abdel Hameid, Estelle Cormet-Boyaka, Wolfgang M. Kuebler, Mohammed Uddin, and Bakhrom K. Berdiev7 July 2021 | American Journal of Physiology-Lung Cellular and Molecular Physiology, Vol. 321, No. 1 Press Release E-cigarette Use during Pregnancy Creates Lung Dysfunction in Babies - December 8, 2022 More from this issue > Volume 320Issue 6June 2021Pages L1183-L1185 Crossmark Copyright & PermissionsPublished by the American Physiological Society.https://doi.org/10.1152/ajplung.00170.2021PubMed33881360History Received 14 April 2021 Accepted 19 April 2021 Published online 22 June 2021 Published in print 1 June 2021 Keywordsalveolar epitheliumCOVID-19ion transportNa,K-ATPaseSARS-CoV-2 Metrics

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call