Search
- Page Path
-
HOME
> Search
Meta-Analysis
- Exploring COVID-19 Pandemic Disparities with Transcriptomic Meta-analysis from the Perspective of Personalized Medicine.
-
Medi Kori, Ceyda Kasavi, Kazim Yalcin Arga
-
J. Microbiol. 2024;62(9):785-798. Published online July 9, 2024
-
DOI: https://doi.org/10.1007/s12275-024-00154-9
-
-
Abstract
- Infection with SARS-CoV2, which is responsible for COVID-19, can lead to differences in disease development, severity and mortality rates depending on gender, age or the presence of certain diseases. Considering that existing studies ignore these differences, this study aims to uncover potential differences attributable to gender, age and source of sampling as well as viral load using bioinformatics and multi-omics approaches. Differential gene expression analyses were used to analyse the phenotypic differences between SARS-CoV-2 patients and controls at the mRNA level. Pathway enrichment analyses were performed at the gene set level to identify the activated pathways corresponding to the differences in the samples. Drug repurposing analysis was performed at the protein level, focusing on host-mediated drug candidates to uncover potential therapeutic differences. Significant differences (i.e. the number of differentially expressed genes and their characteristics) were observed for COVID-19 at the mRNA level depending on the sample source, gender and age of the samples. The results of the pathway enrichment show that SARS-CoV-2 can be combated more effectively in the respiratory tract than in the blood samples.
Taking into account the different sample sources and their characteristics, different drug candidates were identified. Evaluating disease prediction, prevention and/or treatment strategies from a personalised perspective is crucial. In this study, we not only evaluated the differences in COVID-19 from a personalised perspective, but also provided valuable data for further experimental and clinical efforts. Our findings could shed light on potential pandemics.
Journal Articles
- Enterococcus Phage vB_EfaS_HEf13 as an Anti-Biofilm Agent Against Enterococcus faecalis.
-
Dongwook Lee, Jintaek Im, A Reum Kim, Woohyung Jun, Cheol-Heui Yun, Seung Hyun Han
-
J. Microbiol. 2024;62(8):683-693. Published online June 27, 2024
-
DOI: https://doi.org/10.1007/s12275-024-00150-z
-
-
Abstract
- Enterococcus faecalis is a Gram-positive bacterium that is frequently found in the periapical lesion of patients with apical periodontitis. Its biofilm formation in root canal is closely related to the development of refractory apical periodontitis by providing increased resistance to endodontic treatments.
Phage therapy has recently been considered as an efficient therapeutic strategy in controlling various periodontal pathogens. We previously demonstrated the bactericidal capacities of Enterococcus phage vB_EfaS_HEf13 (phage HEf13) against clinically-isolated E. faecalis strains. Here, we investigated whether phage HEf13 affects biofilm formation and pre-formed biofilm of clinically-isolated E.
faecalis, and its combinatory effect with endodontic treatments, including chlorhexidine (CHX) and penicillin. The phage HEf13 inhibited biofilm formation and disrupted pre-formed biofilms of E. faecalis in a dose- and time-dependent manner. Interestingly, phage HEf13 destroyed E. faecalis biofilm exopolysaccharide (EPS), which is known to be a major component of bacterial biofilm. Furthermore, combined treatment of phage HEf13 with CHX or penicillin more potently inhibited biofilm formation and disrupted pre-formed biofilm than either treatment alone. Confocal laser scanning microscopic examination demonstrated that these additive effects of the combination treatments on disruption of pre-formed biofilm are mediated by relatively enhanced reduction in thickness distribution and biomass of biofilm. Collectively, our results suggest that the effect of phage HEf13 on E. faecalis biofilm is mediated by its EPS-degrading property, and its combination with endodontic treatments more potently suppresses E. faecalis biofilm, implying that phage HEf13 has potential to be used as a combination therapy against E. faecalis infections.
- Quorum Quenching Potential of Reyranella sp. Isolated from Riverside Soil and Description of Reyranella humidisoli sp. nov.
-
Dong Hyeon Lee, Seung Bum Kim
-
J. Microbiol. 2024;62(6):449-461. Published online May 30, 2024
-
DOI: https://doi.org/10.1007/s12275-024-00131-2
-
-
Abstract
- Quorum quenching refers to any mechanism that inhibits quorum sensing processes.
In this study, quorum quenching activity among bacteria inhabiting riverside soil was screened, and a novel Gram-stain-negative, rod shaped bacterial strain designated MMS21-HV4-11(T), which showed the highest level of quorum quenching activity, was isolated and subjected to further analysis. Strain MMS21-HV4-11(T) could be assigned to the genus Reyranella of Alphaproteobacteria based on the 16S rRNA gene sequence, as the strain shared 98.74% sequence similarity with Reyranella aquatilis seoho-37(T), and then 97.87% and 97.80% sequence similarity with Reyranella soli KIS14-15(T) and Reyranella massiliensis 521(T), respectively. The decomposed N-acyl homoserine lactone was restored at high concentrations under acidic conditions, implying that lactonase and other enzyme(s) are responsible for quorum quenching. The genome analysis indicated that strain MMS21-HV4-11(T) had two candidate genes for lactonase and one for acylase, and expected protein structures were confirmed. In the quorum sensing inhibition assay using a plant pathogen Pectobacterium carotovorum KACC 14888, development of soft rot was significantly inhibited by strain MMS21-HV4-11(T).
Besides, the swarming motility by Pseudomonas aeruginosa PA14 was significantly inhibited in the presence of strain MMS21-HV4-11(T). Since the isolate did not display direct antibacterial activity against either of these species, the inhibition was certainly due to quorum quenching activity. In an extended study with the type strains of all known species of Reyranella, all strains were capable of degrading N-acyl homoserine lactones (AHLs), thus showing quorum quenching potential at the genus level. This is the first study on the quorum quenching potential and enzymes responsible in Reyranella. In addition, MMS21-HV4-11(T) could be recognized as a new species through taxonomic characterization, for which the name Reyranella humidisoli sp. nov. is proposed (type strain = MMS21-HV4-11( T) = KCTC 82780( T) = LMG 32365(T)).
- Exploring the Therapeutic Potential of Scorpion‑Derived Css54 Peptide Against Candida albicans
-
Jonggwan Park , Hyeongsun Kim , Da Dam Kang , Yoonkyung Park
-
J. Microbiol. 2024;62(2):101-112. Published online April 8, 2024
-
DOI: https://doi.org/10.1007/s12275-024-00113-4
-
-
21
View
-
0
Download
-
1
Citations
-
Abstract
- Candida albicans (C. albicans) is one of the most common opportunistic fungi worldwide, which is associated with a high
mortality rate. Despite treatment, C. albicans remains the leading cause of life-threatening invasive infections. Consequently,
antimicrobial peptides (AMPs) are potential alternatives as antifungal agents with excellent antifungal activity. We previously
reported that Css54, found in the venom of Centrurodies suffusus suffusus (C. s. suffusus) showed antibacterial activity
against zoonotic bacteria. However, the antifungal activity of Css54 has not yet been elucidated. The obj!ective of this study
was to identify the antifungal activity of Css54 against C. albicans and analyze its mechanism. Css54 showed high antifungal
activity against C. albicans. Css54 also inhibited biofilm formation in fluconazole-resistant fungi. The antifungal mechanism
of action of Css54 was investigated using membrane-related assays, including the membrane depolarization assay and
analysis of the membrane integrity of C. albicans after treatment with Css54. Css54 induced reactive oxygen species (ROS)
production in C. albicans, which affected its antifungal activity. Our results indicate that Css54 causes membrane damage
in C. albicans, highlighting its value as a potential therapeutic agent against C. albicans infection.
- miR-135b Aggravates Fusobacterium nucleatum-Induced Cisplatin Resistance in Colorectal Cancer by Targeting KLF13
-
Wei Zeng , Jia Pan , Guannan Ye
-
J. Microbiol. 2024;62(2):63-73. Published online February 24, 2024
-
DOI: https://doi.org/10.1007/s12275-023-00100-1
-
-
33
View
-
1
Download
-
3
Citations
-
Abstract
- Cisplatin resistance is the main cause of colorectal cancer (CRC) treatment failure, and the cause has been reported to be
related to Fusobacterium nucleatum (Fn) infection. In this study, we explored the role of Fn in regulating cisplatin resistance
of CRC cells and its underlying mechanism involved. The mRNA and protein expressions were examined by qRT-PCR
and western blot. Cell proliferation and cell apoptosis were assessed using CCK8 and flow cytometry assays, respectively.
Dual-luciferase reporter gene assay was adopted to analyze the molecular interactions. Herein, our results revealed that Fn
abundance and miR-135b expression were markedly elevated in CRC tissues, with a favorable association between the two.
Moreover, Fn infection could increase miR-135b expression via a concentration-dependent manner, and it also enhanced
cell proliferation but reduced apoptosis and cisplatin sensitivity by upregulating miR-135b. Moreover, KLF13 was proved
as a downstream target of miR-135b, of which overexpression greatly diminished the promoting effect of miR-135b or
Fn-mediated cisplatin resistance in CRC cells. In addition, it was observed that upstream 2.5 kb fragment of miR-135b
promoter could be interacted by β-catenin/TCF4 complex, which was proved as an effector signaling of Fn. LF3, a blocker
of β-catenin/TCF4 complex, was confirmed to diminish the promoting role of Fn on miR-135b expression. Thus, it could be
concluded that Fn activated miR-135b expression through TCF4/β-catenin complex, thereby inhibiting KLF13 expression
and promoting cisplatin resistance in CRC.
- [Protocol] Use of Cas9 Targeting and Red Recombination for Designer Phage Engineering
-
Shin-Yae Choi , Danitza Xiomara Romero-Calle , Han-Gyu Cho , Hee-Won Bae , You-Hee Cho
-
J. Microbiol. 2024;62(1):1-10. Published online February 1, 2024
-
DOI: https://doi.org/10.1007/s12275-024-00107-2
-
-
Abstract
- Bacteriophages (phages) are natural antibiotics and biological nanoparticles, whose application is significantly boosted by
recent advances of synthetic biology tools. Designer phages are synthetic phages created by genome engineering in a way
to increase the benefits or decrease the drawbacks of natural phages. Here we report the development of a straightforward
genome engineering method to efficiently obtain engineered phages in a model bacterial pathogen, Pseudomonas aeruginosa.
This was achieved by eliminating the wild type phages based on the Streptococcus pyogenes Cas9 (SpCas9) and facilitating
the recombinant generation based on the Red recombination system of the coliphage λ (λRed). The producer (PD) cells of
P. aeruginosa strain PAO1 was created by miniTn7-based chromosomal integration of the genes for SpCas9 and λRed under
an inducible promoter. To validate the efficiency of the recombinant generation, we created the fluorescent phages from a
temperate phage MP29. A plasmid bearing the single guide RNA (sgRNA) gene for selectively targeting the wild type gp35
gene and the editing template for tagging the Gp35 with superfolder green fluorescent protein (sfGFP) was introduced into
the PD cells by electroporation. We found that the targeting efficiency was affected by the position and number of sgRNA.
The fluorescent phage particles were efficiently recovered from the culture of the PD cells expressing dual sgRNA molecules.
This protocol can be used to create designer phages in P. aeruginosa for both application and research purposes.
- Comparative Transcriptomic Analysis of Flagellar‑Associated Genes in Salmonella Typhimurium and Its rnc Mutant
-
Seungmok Han , Ji-Won Byun , Minho Lee
-
J. Microbiol. 2024;62(1):33-48. Published online January 5, 2024
-
DOI: https://doi.org/10.1007/s12275-023-00099-5
-
-
Abstract
- Salmonella enterica serovar Typhimurium (S. Typhimurium) is a globally recognized foodborne pathogen that affects both
animals and humans. Endoribonucleases mediate RNA processing and degradation in the adaptation of bacteria to environmental
changes and have been linked to the pathogenicity of S. Typhimurium. Not much is known about the specific regulatory
mechanisms of these enzymes in S. Typhimurium, particularly in the context of environmental adaptation. Thus, this
study carried out a comparative transcriptomic analysis of wild-type S. Typhimurium SL1344 and its mutant (Δrnc), which
lacks the rnc gene encoding RNase III, thereby elucidating the detailed regulatory characteristics that can be attributed to the
rnc gene. Global gene expression analysis revealed that the Δrnc strain exhibited 410 upregulated and 301 downregulated
genes (fold-change > 1.5 and p < 0.05), as compared to the wild-type strain. Subsequent bioinformatics analysis indicated
that these differentially expressed genes are involved in various physiological functions, in both the wild-type and Δrnc
strains. This study provides evidence for the critical role of RNase III as a general positive regulator of flagellar-associated
genes and its involvement in the pathogenicity of S. Typhimurium.
- Genetic and Functional Characterization of a Salicylate 1‑monooxygenase Located on an Integrative and Conjugative Element (ICE) in Pseudomonas stutzeri AJR13
-
Igor Ivanovski , Gerben J. Zylstra
-
J. Microbiol. 2023;61(12):1025-1032. Published online December 15, 2023
-
DOI: https://doi.org/10.1007/s12275-023-00093-x
-
-
Abstract
- Pseudomonas stutzeri strain AJR13 was isolated for growth on the related compounds biphenyl (BPH) and diphenylmethane
(DPM). The BPH and DPM degradative pathway genes are present on an integrative and conjugative element (ICE) in the
chromosome. Examination of the genome sequence of AJR13 revealed a gene encoding a salicylate 1-monooxygenase (salA)
associated with the ICE even though AJR13 did not grow on salicylate. Transfer of the ICE to the well-studied Pseudomonas
putida KT2440 resulted in a KT2440 strain that could grow on salicylate. Knockout mutagenesis of the salA gene on the
ICE in KT2440 eliminated the ability to grow on salicylate. Complementation of the knockout with the cloned salA gene
restored growth on salicylate. Transfer of the cloned salA gene under control of the lac promoter to KT2440 resulted in a
strain that could grow on salicylate. Heterologous expression of the salA gene in E. coli BL21 DE3 resulted in the production
of catechol from salicylate, confirming that it is indeed a salicylate 1-monooxygenase. Interestingly, transfer of the cloned
salA gene under control of the lac promoter to AJR13 resulted in a strain that could now grow on salicylate, suggesting that
gene expression for the downstream catechol pathway is intact.
- Quantitative Analysis of RNA Polymerase Slippages for Production of P3N‑PIPO Trans‑frame Fusion Proteins in Potyvirids
-
Dongjin Choi , Yoonsoo Hahn
-
J. Microbiol. 2023;61(10):917-927. Published online October 16, 2023
-
DOI: https://doi.org/10.1007/s12275-023-00083-z
-
-
21
View
-
0
Download
-
1
Citations
-
Abstract
- Potyvirids, members of the family Potyviridae, produce the P3N-PIPO protein, which is crucial for the cell-to-cell transport
of viral genomic RNAs. The production of P3N-PIPO requires an adenine (A) insertion caused by RNA polymerase slippage
at a conserved GAA AAA A (
GA6) sequence preceding the PIPO open reading frame. Presently, the slippage rate of
RNA polymerase has been estimated in only a few potyvirids, ranging from 0.8 to 2.1%. In this study, we analyzed publicly
available plant RNA-seq data and identified 19 genome contigs from 13 distinct potyvirids. We further investigated the RNA
polymerase slippage rates at the GA6
motif. Our analysis revealed that the frequency of the A insertion variant ranges from
0.53 to 4.07% in 11 potyviruses (genus Potyvirus). For the two macluraviruses (genus Macluravirus), the frequency of the
A insertion variant was found to be 0.72% and 10.96% respectively. Notably, the estimated RNA polymerase slippage rates
for 12 out of the 13 investigated potyvirids were reported for the first time in this study. Our findings underscore the value of
plant RNA-seq data for quantitative analysis of potyvirid genome variants, specifically at the GA6
slippage site, and contribute
to a more comprehensive understanding of the RNA polymerase slippage phenomenon in potyvirids.
- Genetic Characteristics and Phylogeographic Dynamics of Echovirus
-
Yan Wang , Pir Tariq Shah , Yue Liu , Amina Nawal Bahoussi , Li Xing
-
J. Microbiol. 2023;61(9):865-877. Published online September 15, 2023
-
DOI: https://doi.org/10.1007/s12275-023-00078-w
-
-
Abstract
- Echoviruses belong to the genus Enterovirus in the Picornaviridae family, forming a large group of Enterovirus B (EVB)
within the Enteroviruses. Previously, Echoviruses were classified based on the coding sequence of VP1. In this study,
we performed a reliable phylogenetic classification of 277 sequences isolated from 1992 to 2019 based on the full-length
genomes of Echovirus. In this report, phylogenetic, phylogeographic, recombination, and amino acid variability landscape
analyses were performed to reveal the evolutional characteristics of Echovirus worldwide. Echoviruses were clustered into
nine major clades, e.g., G1–G9. Phylogeographic analysis showed that branches G2–G9 were linked to common strains,
while the branch G1 was only linked to G5. In contrast, strains E12, E14, and E16 clustered separately from their G3 and
G7 clades respectively, and became a separate branch. In addition, we identified a total of 93 recombination events, where
most of the events occurred within the VP1-VP4 coding regions. Analysis of amino acid variation showed high variability in
the a positions of VP2, VP1, and VP3. This study updates the phylogenetic and phylogeographic information of Echovirus
and indicates that extensive recombination and significant amino acid variation in the capsid proteins drove the emergence
of new strains.
- Antiviral Activity Against SARS‑CoV‑2 Variants Using in Silico and in Vitro Approaches
-
Hee-Jung Lee , Hanul Choi , Aleksandra Nowakowska , Lin-Woo Kang , Minjee Kim , Young Bong Kim
-
J. Microbiol. 2023;61(7):703-711. Published online June 26, 2023
-
DOI: https://doi.org/10.1007/s12275-023-00062-4
-
-
21
View
-
0
Download
-
1
Citations
-
Abstract
- Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emergence in 2019 led to global health crises and the persistent
risk of viral mutations. To combat SARS-CoV-2 variants, researchers have explored new approaches to identifying
potential targets for coronaviruses. This study aimed to identify SARS-CoV-2 inhibitors using drug repurposing. In silico
studies and network pharmacology were conducted to validate targets and coronavirus-associated diseases to select potential
candidates, and in vitro assays were performed to evaluate the antiviral effects of the candidate drugs to elucidate the
mechanisms of the viruses at the molecular level and determine the effective antiviral drugs for them. Plaque and cytopathic
effect reduction were evaluated, and real-time quantitative reverse transcription was used to evaluate the antiviral activity
of the candidate drugs against SARS-CoV-2 variants in vitro. Finally, a comparison was made between the molecular docking
binding affinities of fenofibrate and remdesivir (positive control) to conventional and identified targets validated from
protein–protein interaction (PPI). Seven candidate drugs were obtained based on the biological targets of the coronavirus,
and potential targets were identified by constructing complex disease targets and PPI networks. Among the candidates,
fenofibrate exhibited the strongest inhibition effect 1 h after Vero E6 cell infection with SARS-CoV-2 variants. This study
identified potential targets for coronavirus disease (COVID-19) and SARS-CoV-2 and suggested fenofibrate as a potential
therapy for COVID-19.
- Description of Luteibacter aegosomatis sp. nov., Luteibacter aegosomaticola sp. nov., and Luteibacter aegosomatissinici sp. nov. isolated from the Intestines of Aegosoma sinicum Larvae
-
Hae-In Joe , Jee-Won Choi , June-Young Lee , Hojun Sung , Su-Won Jeong , Yun-Seok Jeong , Jae-Yun Lee , Jin-Woo Bae
-
J. Microbiol. 2023;61(6):603-613. Published online May 5, 2023
-
DOI: https://doi.org/10.1007/s12275-023-00051-7
-
-
18
View
-
0
Download
-
2
Citations
-
Abstract
- Three novel bacterial strains, 321T,
335T,
and 353T,
were isolated from the intestines of Aegosoma sinicum larvae collected
from Paju-Si, South Korea. The strains were Gram-negative, obligate aerobe and had rod-shaped cells with a single flagellum.
The three strains belonged to the genus Luteibacter in the family Rhodanobacteraceae and shared < 99.2% similarity
in their 16S rRNA gene sequence and < 83.56% similarity in thier whole genome sequence. Strains 321T,
335T,
and 353T
formed a monophyletic clade with Luteibacter yeojuensis KACC 11405T,
L. anthropi KACC 17855T,
and L. rhizovicinus
KACC 12830T,
with sequence similarities of 98.77–98.91%, 98.44–98.58%, and 97.88–98.02%, respectively. Further
genomic analyses, including the construction of the Up-to-date Bacterial Core Gene (UBCG) tree and assessment of other
genome-related indices, indicated that these strains were novel species belonging to the genus Luteibacter. All three strains
contained ubiquinone Q8 as their major isoprenoid quinone and iso-C15:0 and summed feature 9 (
C16:0 10-methyl and/or
iso-C17:1 ω9c) as their major cellular fatty acids. Phosphatidylethanolamine and diphosphatidylglycerol were the major polar
lipids in all the strains. The genomic DNA G + C contents of strains 321T,
335T,
and 353T
were 66.0, 64.5, and 64.5 mol%,
respectively. Based on multiphasic classification, strains 321T,
335T,
and 353T
were classified into the genus Luteibacter
as the type strains of novel species, for which the names Luteibacter aegosomatis sp. nov., Luteibacter aegosomaticola sp.
nov., and Luteibacter aegosomatissinici sp. nov. are proposed, respectively.
Review
- Influence of Microbiota on Vaccine Effectiveness: “Is the Microbiota the Key to Vaccine‑induced Responses?”
-
So-Hee Hong
-
J. Microbiol. 2023;61(5):483-494. Published online April 13, 2023
-
DOI: https://doi.org/10.1007/s12275-023-00044-6
-
-
15
View
-
0
Download
-
6
Citations
-
Abstract
- Vaccines are one of the most powerful tools for preventing infectious diseases. To effectively fight pathogens, vaccines should
induce potent and long-lasting immune responses that are specific to the pathogens. However, not all vaccines can induce
effective immune responses, and the responses vary greatly among individuals and populations. Although several factors, such
as age, host genetics, nutritional status, and region, affect the effectiveness of vaccines, increasing data have suggested that
the gut microbiota is critically associated with vaccine-induced immune responses. In this review, I discuss how gut microbiota
affects vaccine effectiveness based on the clinical and preclinical data, and summarize possible underlying mechanisms
related to the adjuvant effects of microbiota. A better understanding of the link between vaccine-induced immune responses
and the gut microbiota using high-throughput technology and sophisticated system vaccinology approaches could provide
crucial insights for designing effective personalized preventive and therapeutic vaccination strategies.
Editorial
- Editorial] Bacterial Regulatory Mechanisms for the Control of Cellular Processes: Simple Organisms’ Complex Regulation
-
Jin-Won Lee
-
J. Microbiol. 2023;61(3):273-276. Published online April 3, 2023
-
DOI: https://doi.org/10.1007/s12275-023-00036-6
-
-
20
View
-
0
Download
-
1
Citations
-
Abstract
- Bacteria employ a diverse array of cellular regulatory
mechanisms to successfully adapt and thrive in ever-changing
environments, including but not limited to temperature
changes, fluctuations in nutrient availability, the presence
or absence of electron acceptors such as oxygen, the availability
of metal ions crucial for enzyme activity, and the
existence of antibiotics. Bacteria can virtually modulate
any step of gene expression from transcr!ptional initiation
to posttranslational modification of a protein for the control
of cellular processes. Furthermore, one gene regulator
often controls another in a complex gene regulatory network.
Thus, it is not easy to fully understand the intricacies of
bacterial regulatory mechanisms in various environments. In
this special issue, while acknowledging the challenge of covering
all aspects of bacterial regulatory mechanisms across
diverse environments, seven review articles are included to
provide insight into the recent progress in understanding
such mechanisms from different perspectives: positive regulatory
mechanisms by secondary messenger (cAMP receptor
protein), two-component signal transduction mechanisms
(Rcs and Cpx), diverse regulatory mechanisms by a specific
environmental factor in specific bacteria (oxygen availability
in Mycobacterium and manganese ion availability in Salmonella),
diverse regulatory mechanisms by a specific environmental
factor (temperature and antibiotics), and regulatory
mechanisms by antibiotics in cell wall synthesis.
Bacteria, as ubiquitous organisms that can be found in
almost every environment, carry out complex cellular processes
that allow them to survive and thrive in a variety of
different conditions despite their small size and relative simplicity.
One of the key factors that allows bacteria to carry
out these complex processes is their ability to regulate gene
expression through various mechanisms. Gene expression
is a fundamental biological process by which the genetic
information encoded in a gene is transcribed into an RNA
molecule and subsequently translated into a functional gene
product, often a protein. Furthermore, the activity levels of
proteins may further be altered by posttranslational modification.
Regulation of gene expression refers to the control
of the amount and timing of gene expression, and thus it
can be divided into transcr!ptional, translational, and posttranslational
levels.
Review
- Membrane Proteins as a Regulator for Antibiotic Persistence in Gram‑Negative Bacteria
-
Jia Xin Yee , Juhyun Kim , Jinki Yeom
-
J. Microbiol. 2023;61(3):331-341. Published online February 17, 2023
-
DOI: https://doi.org/10.1007/s12275-023-00024-w
-
-
19
View
-
0
Download
-
1
Citations
-
Abstract
- Antibiotic treatment failure threatens our ability to control bacterial infections that can cause chronic diseases. Persister bacteria
are a subpopulation of physiological variants that becomes highly tolerant to antibiotics. Membrane proteins play crucial
roles in all living organisms to regulate cellular physiology. Although a diverse membrane component involved in persistence
can result in antibiotic treatment failure, the regulations of antibiotic persistence by membrane proteins has not been fully
understood. In this review, we summarize the recent advances in our understanding with regards to membrane proteins in
Gram-negative bacteria as a regulator for antibiotic persistence, highlighting various physiological mechanisms in bacteria.
TOP