Open Access

Klebsiella pneumoniae infection inhibits autophagy by alveolar type II epithelial cells

  • Authors:
    • Zhiyun Shi
    • Gang Li
    • Lin Zhang
    • Miao Ma
    • Wei Jia
  • View Affiliations

  • Published online on: August 19, 2020     https://doi.org/10.3892/etm.2020.9123
  • Pages: 3703-3708
  • Copyright: © Shi et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

To investigate the molecular mechanism underlying the interaction between autophagosomes of alveolar type II epithelial (A549) cells and Klebsiella pneumoniae, an in vitro model of K. pneumoniae‑infected A549 cells was established. Western blot analysis and immunofluorescence staining were used to detect the distribution of microtubule‑associated protein 1A/1B‑light chain 3 (LC3) and the expression of the LC3‑phosphatidylethanolamine conjugate (LC3‑II). K. pneumoniae‑infected A549 cells were treated with different concentrations of an autophagy inhibitor or promoter for different time periods to assess the level of autophagy. Western blot analysis and immunofluorescence staining showed that K. pneumoniae could induce autophagy by A549 cells. With an increase in bacterial concentration and time of infection, autophagy gradually increased. The autophagy inhibitor significantly downregulated, while the promoter upregulated, expression of the autophagy‑related protein LC3‑II. Autophagy plays an important role in the resistance of alveolar type II epithelial (A549) cells to K. pneumoniae infection.

Introduction

Klebsiella pneumoniae is a Gram-negative enteric bacillus that is a member of the Enterobacteriaceae family. In immunocompromised patients, K. pneumoniae can infect the lungs, blood, and urinary tract. In recent years, the incidence of nosocomial infections of carbapenem-resistant K. pneumoniae (CRKP) has continued to increase, resulting in exceptionally high mortality rates (1,2). In earlier studies, our group investigated the resistance mechanism and conducted homology analysis and molecular epidemiological studies of 88 clinical isolates of penicillin- and carbapenem-resistant Enterobacteriaceae (25 CRKP) collected in Ningxia, China, from 2011 to 2016. The results of these studies revealed that 100% of carbapenem-resistant K. pneumoniae were also resistant to imipenem. Furthermore, the NDM-1 and KPC-2 genes were found to code for the carbapenemase enzyme. Drug-resistant strains can pass resistant plasmids to sensitive strains of Enterobacteriaceae, although the distribution varied (3-6).

Since there have been relatively few studies of the host innate immune response to K. pneumoniae infection, the virulence and pathogenic mechanism of this species remain unclear. Autophagy is an important component of natural immunity and plays key roles in the host immune response, including inflammation, to infections of pathogenic microbes. Microorganism-mediated autophagy has been shown to clear the host cells of Salmonella sp., Pseudomonas aeruginosa, and group A streptococcus (7-11), and may contribute to the long-term survival and replication in the host cell of Brucella melitensis (12) and Mycobacterium tuberculosis (13). Previous studies have confirmed that autophagy also protects type II alveolar epithelial cells (AECIIs) from M. tuberculosis infection (14) and furthermore conveys important immunomodulatory effects in the lung against microbial infections (15). For example, AECIIs can rapidly phagocytize and kill invading Aspergillus fumigatus (16). The ability of AECIIs to phagocytize M. tuberculosis is 60-70% greater than that of macrophages (17), suggesting that autophagosomes may play a role in the ability of K. pneumoniae to infect AECIIs. In this study, human adenocarcinoma alveolar basal epithelial (A549) cells were used to study the mechanism of autophagy of K. pneumoniae in order to establish a novel approach for effective intervention and treatment of K. pneumoniae infection.

Materials and methods

Bacterial strain and cell type

The reference strain K. pneumoniae K6 (ATCC 700603) was obtained from the American Type Culture Collection and preserved in our laboratory. A549 cells are hypotriploid alveolar basal epithelial cells that are widely used as a model of lung adenocarcinoma, as well as an in vitro model of type II pulmonary epithelial cells, were purchased from The Cell Bank of Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China).

Reagents and instruments

Dulbecco's modified Eagle's medium (DMEM), fetal bovine serum, and trypsin were purchased from HyClone Laboratories, Inc. Cell culture plates were obtained from Axygen Scientific, Inc. Pierce™ Immunoprecipitation Lysis Buffer and the Pierce™ Bicinchoninic Acid (BCA) kit were purchased from Thermo Fisher Scientific, Inc. An enhanced chemiluminescence kit was obtained from Beyotime Institute of Biotechnology. Lysogeny broth (LB) solid medium was obtained from Oxoid Ltd. Rapamycin and 3-methyladenine (3-MA) were purchased from Sigma-Aldrich; Merck KGaA. Antibodies against microtubule-associated protein 1A/1B-light chain 3 (LC3) and β-actin were acquired from Abcam PLC. Alexa Fluor™ 488 goat anti-rabbit secondary antibodies against immunoglobulin G were purchased from Invitrogen; Thermo Fisher Scientific, Inc. An apparatus for sodium dodecyl sulfate-polyacrylamide gel electrophoresis and a wet electroblotting system were acquired from Bio-Rad Laboratories, Inc. A laser confocal fluorescence microscope was purchased from Olympus Corporation.

Bacterial count

Activated single K. pneumoniae colonies were inoculated on conventional LB solid medium and incubated at 37˚C for 24 h. Then a typical single colony was cultivated in LB liquid medium to the logarithmic phase at 37˚C while rotating at 250 rpm. The bacterial culture was diluted in sterile physiological saline to a concentration of 1.2x1011 bacteria/ml.

Cell culture

A549 cells were inoculated into a Petri dish and cultured in a DMEM supplemented with 10% fetal bovine serum, 100 U/ml of penicillin, and 100 µg/ml of streptomycin under an atmosphere of 5% CO2/95% air at 37˚C. At approximately 80% confluence, the medium was discarded and the cells were washed one or two times with phosphate-buffered saline (PBS). After the PBS was removed, the cells were digested for 1-3 min by the addition of 1 ml of trypsin. Then, 3 ml of complete medium was added to terminate the digestion and the cells were transferred to a 15-ml centrifuge tube and centrifuged 300 x g for 5 min at room temperature. After the supernatant was discarded, the cells were resuspended in 3 ml of medium and passaged (1:3) in a petri dish.

Establishment of a cellular infection model

A549 cells were seeded in T25 flasks and grown to a confluence of 80-90%. Next, the cells were digested with trypsin and plated in the wells of 6-well plates at a concentration of 2x106 cells, as determined with a hemocytometer. After culturing overnight, the cells were infected with bacteria at a multiplicity of infection (MOI) of 100:1, 50:1, 10:1, 5:1, or 1:1 and incubated for 3 h. Then, the medium was removed and the cells were washed three times with PBS and then incubated in complete DMEM containing gentamycin for 24 h. The resulting cells were used for immunofluorescence staining and western blot analysis.

Detection of autophagic changes to LC3 by confocal microscopy

A549 cells were cultured in DMEM containing 10% fetal bovine serum. At 80 to 90% confluence, the culture medium was removed and the cells were washed twice with PBS, fixed with 4% paraformaldehyde at room temperature for 10 min, and then washed three times with pre-chilled PBS. Following the addition of PBS containing 0.5% Triton X-100, the cell membranes were lysed on ice for 10 min. The cells were then washed three times with pre-chilled PBS-Triton X-100. PBS containing 3% bovine serum albumin (BSA) was added and blocked at room temperature for 30 min. LC3 antibody was diluted to 1:100 in PBS-1% BSA solution. After removal of the blocking solution, the primary antibody was added and incubated at room temperature for 2 h or at 4˚C overnight. The primary antibody was removed by washing three times with PBS-0.35% Tween-20. Fluorescent secondary antibodies were diluted to 1:400 in PBS-1% BSA solution. After removal of PBS-T, the secondary antibody was added and the samples were incubated for 1 h at room temperature in the dark. For removal of the secondary antibody, the samples were washed three times with PBS-0.35% Tween-20. Following the addition of 100 ng/ml of 4',6-diamidino-2-phenylindole (DAPI) solution, the samples were incubated at room temperature in the dark for 10 min. Then, the DAPI solution was removed and the samples were washed three times with PBS-0.35% Tween-20. Following the addition of anti-fluorescence quenching solution, the samples were placed in the dark at 4˚C or imaged using a laser confocal fluorescence microscope.

Western blot detection of the autophagic protein LC3-II

Triplicate samples of A549 cells were infected with K. pneumoniae at multiple MOIs for various times. Then, the expression levels of LC3-II and LC3-II/LC3-I ratios were detected. The BCA assay was used to detect the concentrations of proteins extracted from K. pneumoniae-infected A549 cells. After separation by vertical electrophoresis on 10% sodium dodecyl sulfate-polyacrylamide gels, the proteins were transferred to polyvinylidene fluoride membranes by semi-dry electroporation. The membranes were sealed and incubated overnight with the primary antibody. The next day, the membranes were washed and then incubated with the secondary antibody. After a final washing, the protein bands on the membranes were visualized using a gel imager. Quantity One 1-D Analysis Software (Bio-Rad Laboratories) was used to quantify the protein bands with β-actin as a reference.

Induction and inhibition of autophagy

Induction and inhibition of autophagy were assessed using four experimental groups: An infection group (Pneu), autophagy inhibition group (Pneu + 3-MA), autophagy induction group (Pneu + rapamycin), and negative control group. At a confluence of 80-90%, the A549 cells were infected at a bacteria:cell ratio of 100:1 and incubated for 3 h. After the medium was removed, the cells were washed three times with PBS and then cultivated in medium containing gentamicin. At the same time, trimethylpurine (3-MA), rapamycin, purine (3-MA), and rapamycin were added to the medium at final concentrations of 5 and 10 µM, respectively. At 24, 48, and 72 h, the cells were stained with immunofluorescent markers and subjected to western blot analysis.

Statistical analysis

Statistical analysis was performed using SPSS software for Windows, version 15.0. (SPSS, Inc.). The data of experiments repeated three times were used for analysis and are presented as the mean ± standard deviation. One-way analysis of variance was used for comparisons between groups. The Tukey test was used to identify differences among three or more groups. At α=0. 05, a probability (p) value of <0.05 was considered statistically significant.

Results

Morphologies of K. pneumoniae-infected cells

The bacteria were added to the cell cultures at ratios of 100:1, 50:1, 10:1, 5:1, and 1:1. As shown in Fig. 1, there were no significant changes to the morphology of A549 cells.

Confocal microscopy of autophagic changes to LC3 in A549 cells

As compared with the control group of cells that were not infected with K. pneumoniae, the autophagosomes of LC3 in the infected group were significantly changed. The immunofluorescence results showed that LC3 content in autophagosomes was lowest at a bacteria:cell ratio of 10:1. As shown in Fig. 2, K. pneumoniae inhibited autophagy and weakened the resistance of A549 cells, resulting in increased necrosis and proliferation of K. pneumoniae.

Western blot detection of the autophagic protein LC3-II

The western blot results showed that LC3-II protein expression was significantly higher in infected A549 cells than in the control uninfected group and the LC3-II/LC3-I ratio was significantly increased (P<0.05), indicating that K. pneumoniae promoted autophagy. As the proportion of bacterial cells (MOI) increased, autophagy also increased. However, as shown in Fig. 3, at a bacteria:cell ratio of 10:1, the level of autophagic cells had relatively decreased as compared with that of the other infection groups.

Immunofluorescence results

The immunofluorescence results revealed significant morphological changes to the LC3 autophagosomes of infected cells as compared with those of the uninfected control cells. With the extension of time and under the action of gentamycin, the invasive abilities of K. pneumoniae were weakened (autophagy increased). As shown in Fig. 4, 3-MA inhibited autophagy, while rapamycin promoted autophagy.

Western blot results

The western blot results (Fig. 5) showed that as compared with the control uninfected group, autophagy was increased in cells infected with K. pneumoniae. Moreover, with time, LC3-II protein expression gradually increased. The addition of 100 nM rapamycin to induce autophagy further upregulated LC3-II protein expression. Moreover, the addition of 10 mM 3-MA to inhibit autophagy significantly downregulated LC3-II protein expression. LC3-II protein expression levels were significantly higher in the infection and autophagy induction groups as compared with the control group (P<0.05). In addition, LC3-II protein expression was significantly lower in the infection group treated with 3-MA (P<0.05).

Discussion

Over the past two decades, K. pneumoniae has surpassed Escherichia coli as the important pathogen isolated from patients with purulent liver abscesses, and has tended to spread globally (18). With the large number and irrational abuse of antibacterial drugs, the drug resistance rate of K. pneumoniae continues to increase (19). The US Centers for Disease Control and Prevention listed Krk-resistant pneumococcus pneumoniae (CRKP) as the highest urgent grade in 2013(20). Moreover, the World Health Organization mentioned CRKP in the global drug resistance surveillance report (21). Therefore, elucidating the pathogenic mechanism underlying K. pneumoniae infection and finding effective control measures is of great value for the treatment and prevention of K. pneumoniae infection.

The role of autophagy in bacteria-infected cells has attracted increasing attention and has been shown to play an important role in host defense, especially in immune cells (22,23). Autophagy can directly affect the immune and inflammatory responses throughout the body. For example, autophagy participates in the clearance of invading bacteria via degradation of autophagic lysosomes and also plays an effective regulatory role in the immune response against pathogen invasion (24). Autophagy and bacterial infection restrict and promote one another. In addition, microbe-mediated autophagy can help the body to clear Salmonella sp., P. aeruginosa, and group A streptococcus (7-11), thereby promoting long-term survival and replication in host cells. For example, M. tuberculosis can survive in macrophages in latent infections (25). Recently, Ato et al found that miR-129-3p can inhibit autophagy through Atg4b, thus contributing to the survival of M. tuberculosis (26). Some Shigella and Listeria species have evolved various mechanisms to disrupt the growth and survival of autophagy systems, and to destroy autophagosomes (27), while others, such as Legionella pneumophila, regulate intracellular transport and inhibit autophagosome formation (28).

LC3 is an autophagosomal membrane protein that is considered to be a specific autophagosome marker. The expression and transformation of LC3-II (LC3-II/LC3-I) are important indicators to evaluate the level of intracellular autophagy. AECIIs serve as the first line of defense against pulmonary exposure to exogenous pathogens and play an extremely important defensive function in lung infections. Hence, in-depth studies of the molecular mechanism underlying the interactions of AECIIs, autophagosomes, and K. pneumoniae are warranted to further elucidate the mechanism underlying the unique immune function of AECIIs for the prevention and treatment of K. pneumoniae infection.

In the present study, human alveolar type II epithelial (A549) cells were cultured in vitro and infected with K. pneumoniae at ratios of 100:1, 50:1, 10:1, 5:1, and 1:1 to establish an infection model. The cells were then collected at 0, 24, 48, and 72 h after infection for detection of relevant indicators. At the same time, A549 cells infected with K. pneumoniae were treated with the autophagy inhibitor 3-MA and the autophagy inducer rapamycin. The immunofluorescence and western blot results showed that in A549 cells, LC3 autophagosome expression, LC3-II protein expression, and the LC3-II/LC3-I ratio were significantly increased relative to the control uninfected cells, indicating that K. pneumoniae promotes autophagy by A549 cells. As the proportion of K. pneumoniae-infected A549 cells had increased, autophagy also increased. However, at a bacteria:cell ratio of 10:1, autophagy relatively decreased. At this ratio, K. pneumoniae can inhibit autophagy and weaken the resistance of A549 cells, resulting in increased necrosis, thereby promoting the proliferation of K. pneumoniae. With prolonged infection, cell autophagy gradually increased. In response to the addition of the autophagy inducer rapamycin, LC3-II protein expression was further upregulated, which promoted the autophagic effect of bacteria in the infected cells. On the contrary, in response to the addition of the autophagy inhibitor 3-MA, LC3-II protein expression was downregulated, which inhibited the autophagic effect of bacteria. These results further confirm that K. pneumoniae can induce autophagy in A549 cells in vitro.

There were some limitations to this study that should be addressed. Due to the COVID-19 pandemic, we were unable to perform proliferation/viability assays to quantitatively assess the responses of A549 cells or to repeat the experiments in vitro and in vivo with other cell lines in order to illustrate the hypothesis of the article.

Autophagy and inflammation are hot topics in the study of infectious diseases. Recent studies have shown that Notch signaling can serve as a substrate and participates in the process of autophagy (29). Autophagy is involved in the regulation of the inflammatory response, since the upregulation and absence of autophagy are closely related to the development of infectious diseases. In future studies, our group plans to use molecular biology techniques, both in vitro and in vivo, to further study autophagy-related targets and related molecular signaling pathways involved with inflammation. The results of the present study provide theoretical and experimental evidence of the pathogenic mechanism for the prevention and treatment of K. pneumoniae infection.

Acknowledgements

Not applicable.

Funding

The study was supported by grants from the First-Class Discipline Construction Founded Project of NingXia Medical University and the School of Clinical Medicine (grant no. NXYLXK2017A05), and the Ningxia Natural Science Foundation Project (grant no. 2019AAC03216).

Availability of data and materials

The datasets used and/or analyzed in the present study are available from the corresponding author on reasonable request.

Authors' contributions

ZS wrote the manuscript and performed western blot analysis. GL and LZ were responsible for the cell culture and transfection. MM and WJ contributed to the analysis of the observation indexes. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Hoxha A, Kärki T, Giambi C, Montano C, Sisto A, Bella A and D'Ancona F: Study Working Group. Attributable mortality of carbapenem-resistant klebsiella pneumoniae infections in a prospective matched cohort study in Italy, 2012-2013. J Hosp Infect. 92:61–66. 2016.PubMed/NCBI View Article : Google Scholar

2 

Zhang X, Gu B, Mei Y, Wen Y and Xia W: Increasing resistance rate to carbapenem among blood culture isolates of Klebsiella pneumoniae, Acinetobacter baumannii and Pseudomonas aeruginosa in a university-affiliated hospital in China, 2004-2011. J Antibiot (Tokyo). 68:115–120. 2015.PubMed/NCBI View Article : Google Scholar

3 

Shi Z, Zhao H, Li G and Jia W: Molecular characteristics of carbapenem resistant enterobacter cloacae in ningxia province, China. Front Microbiol. 8(94)2017.PubMed/NCBI View Article : Google Scholar

4 

Shi ZY, Zhao HJ, Li CY, Li G, Tao J, Li SS, Yao Y and Jia W: Bacterial resistance and genotypic distribution of enterobacteriaceae in burn patients. Chin J Nosocomiol. 26:2660–2663. 2016.

5 

Li G, Zhao HD, Jia W, Zhao M, Zhou XY, Ma H, Wang LL, Li SS, Dong H and Shi ZY: Analysis of clinical distribution and drug resistance of 7 157 strains of Enterobacteriaceae. Int Test Med J. 3:494–497. 2015.

6 

Tängdén T and Giske CG: Global dissemination of extensively drug-resistant carbapenemase-producing enterobacteriaceae: Clinical perspectives on detection, treatment and infection control. J Intern Med. 277:501–512. 2015.PubMed/NCBI View Article : Google Scholar

7 

Noad J, von der Malsburg A, Pathe C, Michel MA, Komander D and Randow F: LUBAC-synthesized linear ubiquitin chains restrict cytosol-invading bacteria by activating autophagy and NF-κB. Nat Microbiol. 2(17063)2017.PubMed/NCBI View Article : Google Scholar

8 

Negroni A, Colantoni E, Vitali R, Palone F, Pierdomenico M, Costanzo M, Cesi V, Cucchiara S and Stronati L: NOD2 induces autophagy to control AIEC bacteria infectiveness in intestinal epithelial cells. Inflamm Res. 65:803–813. 2016.PubMed/NCBI View Article : Google Scholar

9 

Yuan K, Huang C, Fox J, Laturnus D, Carlson E, Zhang B, Yin Q, Gao H and Wu M: Autophagy plays an essential role in the clearance of Pseudomonas aeruginosa by alveolar macrophages. J Cell Sci. 125:507–515. 2012.PubMed/NCBI View Article : Google Scholar

10 

Cemma M, Kim PK and Brumell JH: The ubiquitin-binding adaptor proteins p62/SQSTM1 and NDP52 are recruited independently to bacteria-associated microdomains to target Salmonella to the autophagy pathway. Autophagy. 7:341–345. 2011.PubMed/NCBI View Article : Google Scholar

11 

Nakagawa I, Amano A, Mizushima N, Yamamoto A, Yamaguchi H, Kamimoto T, Nara A, Funao J, Nakata M, Tsuda K, et al: Autophagy defends cells against invading group A streptococcus. Science. 306:1037–1040. 2004.PubMed/NCBI View Article : Google Scholar

12 

Guo F, Zhang H, Chen C, Hu S, Wang Y, Qiao J, Ren Y, Zhang K, Wang Y and Du G: Autophagy favors Brucella melitensis survival in infected macrophages. Cell Mol Biol Lett. 17:249–257. 2012.PubMed/NCBI View Article : Google Scholar

13 

Romagnoli A, Etna MP, Giacomini E, Pardini M, Remoli ME, Corazzari M, Falasca L, Goletti D, Gafa V, Simeone R, et al: ESX-1 dependent impairment of autophagic flux by Mycobacterium tuberculosis in human dendritic cells. Autophagy. 8:1357–1370. 2012.PubMed/NCBI View Article : Google Scholar

14 

Crotzer VL and Blum JS: Autophagy and intracellular surveillance: Modulating MHC class II antigen presentation with stress. Proc Natl Acad Sci USA. 102:7779–7780. 2005.PubMed/NCBI View Article : Google Scholar

15 

Stegemann-Koniszewski S, Jeron A, Gereke M, Geffers R, Kröger A, Gunzer M and Bruder D: Alveolar type II epithelial cells contribute to the anti-influenza A virus response in the lung by integrating pathogen- and microenvironment-derived signals. mBio. 7:e00276–e00216. 2016.PubMed/NCBI View Article : Google Scholar

16 

Zhao YX, Li G and Yu RJ: The effect of retinoic acid on C3 and factor B secretion of human alveolar type II epithelial cells induced with cytokines. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 21:442–444. 2005.PubMed/NCBI(In Chinese).

17 

Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI and Deretic V: Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell. 119:753–766. 2004.PubMed/NCBI View Article : Google Scholar

18 

Liu Y, Wang JY and Jiang W: An increasing prominent disease of klebsiella pneumoniae liver abscess: Etiology, diagnosis, and treatment. Gastroenterol Res Pract. 2013(258514)2013.PubMed/NCBI View Article : Google Scholar

19 

Baraniak A, Izdebski R, Fiett J, Gawryszewska I, Bojarska K, Herda M, Literacka E, Żabicka D, Tomczak H, Pewińska N, et al: NDM-producing enterobacteriaceae in Poland, 2012-14: Inter-regional outbreak of klebsiella pneumoniae ST11 and sporadic cases. J Antimicrob Chemother. 71:85–91. 2016.PubMed/NCBI View Article : Google Scholar

20 

Schuchat A, Tappero J and Blandford J: Global health and the US centers for disease control and prevention. Lancet. 384:98–101. 2014.PubMed/NCBI View Article : Google Scholar

21 

World Health Organization (WHO): Antimicrobial resistance: global report on surveillance. WHO, Geneva, 2014. https://www.who.int/antimicrobial-resistance/publications/surveillancereport/en/.

22 

Deretic V: Autophagy in infection. Curr Opin Cell Biol. 22:252–262. 2010.PubMed/NCBI View Article : Google Scholar

23 

Deretic V: Autophagy in immunity and cell-autonomous defense against intracellular microbes. Immunol Rev. 240:92–104. 2011.PubMed/NCBI View Article : Google Scholar

24 

Deretic V and Levine B: Autophagy, immunity, and microbial adaptations. Cell Host Microbe. 5:527–549. 2009.PubMed/NCBI View Article : Google Scholar

25 

Mizushima N, Yoshimori T and Levine B: Methods in mammalian autophagy. Cell. 2:313–326. 2010.PubMed/NCBI View Article : Google Scholar

26 

Ato K, Akaki T, Shimizu T, Sano C, Ogasawara K and Tomioka H: Invasion and intracellular growth of Mycobacterium tuberculosis and myco-bacterium avium complex adapted to intramacrophagic environment within macrophages and type II alveolar epithelial cells. Kekkaku. 76:53–57. 2001.PubMed/NCBI(In Japanese).

27 

Orsi GB, García-Fernández A, Giordano A, Venditti C, Bencardino A, Gianfreda R, Falcone M, Carattoli A and Venditti M: Risk factors and clinical significance of ertapenem- resistant Klebsiella pneumoniae in hospitalised patients. J Hosp Infect. 78:54–58. 2011.PubMed/NCBI View Article : Google Scholar

28 

Won S, Munoz-Price LS, Lolans K, Hota B, Weinstein RA and Hayden MK: Centers for Disease Control and Prevention Epicenter Program. Emergence and rapid regional spread of Klebsiella pneumoniae carbapenemase-producing enterobacteriaceae. Clin Infect Dis. 53:532–540. 2011.PubMed/NCBI View Article : Google Scholar

29 

Wu X, Fleming A, Ricketts T, Pavel M, Virgin H, Menzies FM and Rubinsztein DC: Autophagy regulates notch degradation and modulates stem cell development and neurogenesis. Nat Commun. 7:10533–10550. 2016.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2020
Volume 20 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shi Z, Li G, Zhang L, Ma M and Jia W: Klebsiella pneumoniae infection inhibits autophagy by alveolar type II epithelial cells. Exp Ther Med 20: 3703-3708, 2020
APA
Shi, Z., Li, G., Zhang, L., Ma, M., & Jia, W. (2020). Klebsiella pneumoniae infection inhibits autophagy by alveolar type II epithelial cells. Experimental and Therapeutic Medicine, 20, 3703-3708. https://doi.org/10.3892/etm.2020.9123
MLA
Shi, Z., Li, G., Zhang, L., Ma, M., Jia, W."Klebsiella pneumoniae infection inhibits autophagy by alveolar type II epithelial cells". Experimental and Therapeutic Medicine 20.4 (2020): 3703-3708.
Chicago
Shi, Z., Li, G., Zhang, L., Ma, M., Jia, W."Klebsiella pneumoniae infection inhibits autophagy by alveolar type II epithelial cells". Experimental and Therapeutic Medicine 20, no. 4 (2020): 3703-3708. https://doi.org/10.3892/etm.2020.9123