1.Immunomodulatory Effect of Epidermal Growth Factor Secreted by Human Umbilical Cord Blood-Derived Mesenchymal Stem Cells on Atopic Dermatitis
Namhee JUNG ; TaeHo KONG ; Yeonsil YU ; Hwanhee PARK ; Eunjoo LEE ; SaeMi YOO ; SongYi BAEK ; Seunghee LEE ; Kyung-Sun KANG
International Journal of Stem Cells 2022;15(3):311-323
Background and Objectives:
Human mesenchymal stem cells (MSCs) are emerging as a treatment for atopic dermatitis (AD), a chronic inflammatory skin disorder that affects a large number of people across the world. Treatment of AD using human umbilical cord blood-derived MSCs (hUCB-MSCs) has recently been studied. However, the mechanism underlying their effect needs to be studied continuously. Thus, the objective of this study was to investigate the immunomodulatory effect of epidermal growth factor (EGF) secreted by hUCB-MSCs on AD.
Methods:
and Results: To explore the mechanism involved in the therapeutic effect of MSCs for AD, a secretome array was performed using culture medium of hUCB-MSCs. Among the list of genes common for epithelium development and skin diseases, we focused on the function of EGF. To elucidate the effect of EGF secreted by hUCB-MSCs, EGF was downregulated in hUCB-MSCs using EGF-targeting small interfering RNA. These cells were then co-cultured with keratinocytes, Th2 cells, and mast cells. Depletion of EGF disrupted immunomodulatory effects of hUCB-MSCs on these AD-related inflammatory cells. In a Dermatophagoides farinae-induced AD mouse model, subcutaneous injection of hUCB-MSCs ameliorated gross scoring, histopathologic damage, and mast cell infiltration. It also significantly reduced levels of inflammatory cytokines including interleukin (IL)-4, tumor necrosis factor (TNF)-α, thymus and activation-regulated chemokine (TARC), and IL-22, as well as IgE levels. These therapeutic effects were significantly attenuated at all evaluation points in mice injected with EGF-depleted hUCB-MSCs.
Conclusions
EGF secreted by hUCB-MSCs can improve AD by regulating inflammatory responses of keratinocytes, Th2 cells, and mast cells.
2.Bioinformatics services for analyzing massive genomic datasets
Gunhwan KO ; Pan-Gyu KIM ; Youngbum CHO ; Seongmun JEONG ; Jae-Yoon KIM ; Kyoung Hyoun KIM ; Ho-Yeon LEE ; Jiyeon HAN ; Namhee YU ; Seokjin HAM ; Insoon JANG ; Byunghee KANG ; Sunguk SHIN ; Lian KIM ; Seung-Won LEE ; Dougu NAM ; Jihyun F. KIM ; Namshin KIM ; Seon-Young KIM ; Sanghyuk LEE ; Tae-Young ROH ; Byungwook LEE
Genomics & Informatics 2020;18(1):e8-
The explosive growth of next-generation sequencing data has resulted in ultra-large-scale datasets and ensuing computational problems. In Korea, the amount of genomic data has been increasing rapidly in the recent years. Leveraging these big data requires researchers to use large-scale computational resources and analysis pipelines. A promising solution for addressing this computational challenge is cloud computing, where CPUs, memory, storage, and programs are accessible in the form of virtual machines. Here, we present a cloud computing-based system, Bio-Express, that provides user-friendly, cost-effective analysis of massive genomic datasets. Bio-Express is loaded with predefined multi-omics data analysis pipelines, which are divided into genome, transcriptome, epigenome, and metagenome pipelines. Users can employ predefined pipelines or create a new pipeline for analyzing their own omics data. We also developed several web-based services for facilitating downstream analysis of genome data. Bio-Express web service is freely available at https://www.bioexpress.re.kr/.
3.Bioinformatics services for analyzing massive genomic datasets
Gunhwan KO ; Pan-Gyu KIM ; Youngbum CHO ; Seongmun JEONG ; Jae-Yoon KIM ; Kyoung Hyoun KIM ; Ho-Yeon LEE ; Jiyeon HAN ; Namhee YU ; Seokjin HAM ; Insoon JANG ; Byunghee KANG ; Sunguk SHIN ; Lian KIM ; Seung-Won LEE ; Dougu NAM ; Jihyun F. KIM ; Namshin KIM ; Seon-Young KIM ; Sanghyuk LEE ; Tae-Young ROH ; Byungwook LEE
Genomics & Informatics 2020;18(1):e8-
The explosive growth of next-generation sequencing data has resulted in ultra-large-scale datasets and ensuing computational problems. In Korea, the amount of genomic data has been increasing rapidly in the recent years. Leveraging these big data requires researchers to use large-scale computational resources and analysis pipelines. A promising solution for addressing this computational challenge is cloud computing, where CPUs, memory, storage, and programs are accessible in the form of virtual machines. Here, we present a cloud computing-based system, Bio-Express, that provides user-friendly, cost-effective analysis of massive genomic datasets. Bio-Express is loaded with predefined multi-omics data analysis pipelines, which are divided into genome, transcriptome, epigenome, and metagenome pipelines. Users can employ predefined pipelines or create a new pipeline for analyzing their own omics data. We also developed several web-based services for facilitating downstream analysis of genome data. Bio-Express web service is freely available at https://www.bioexpress.re.kr/.
4.Tumor Microenvironment Modulation by Neoadjuvant Erlotinib Therapy and Its Clinical Impact on Operable EGFR-Mutant Non–Small Cell Lung Cancer
Beung-Chul AHN ; Charny PARK ; Moon Soo KIM ; Jong Mog LEE ; Jin Ho CHOI ; Hyae Young KIM ; Geon Kook LEE ; Namhee YU ; Youngjoo LEE ; Ji-Youn HAN
Cancer Research and Treatment 2024;56(1):70-80
Purpose:
Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors have greatly improved survival in EGFR-mutant (EGFRm) non–small cell lung cancer (NSCLC); however, their effects on the tumor microenvironment (TME) are unknown. We assessed the changes induced by neoadjuvant erlotinib therapy (NE) in the TME of operable EGFRm NSCLC.
Materials and Methods:
This was a single-arm phase II trial for neoadjuvant/adjuvant erlotinib therapy in patients with stage II/IIIA EGFRm NSCLC (EGFR exon 19 deletion or L858R mutations). Patients received up to 2 cycles of NE (150 mg/day) for 4 weeks, followed by surgery and adjuvant erlotinib or vinorelbine plus cisplatin therapy depending on observed NE response. TME changes were assessed based on gene expression analysis and mutation profiling.
Results:
A total of 26 patients were enrolled; the median age was 61, 69% were female, 88% were stage IIIA, and 62% had L858R mutation. Among 25 patients who received NE, the objective response rate was 72% (95% confidence interval [CI], 52.4 to 85.7). The median disease-free and overall survival (OS) were 17.9 (95% CI, 10.5 to 25.4) and 84.7 months (95% CI, 49.7 to 119.8), respectively. Gene set enrichment analysis in resected tissues revealed upregulation of interleukin, complement, cytokine, transforming growth factor β, and hedgehog pathways. Patients with upregulated pathogen defense, interleukins, and T-cell function pathways at baseline exhibited partial response to NE and longer OS. Patients with upregulated cell cycle pathways at baseline exhibited stable/progressive disease after NE and shorter OS.
Conclusion
NE modulated the TME in EGFRm NSCLC. Upregulation of immune-related pathways was associated with better outcomes.
5.Serotype Distribution and Antimicrobial Resistance of Streptococcus pneumoniae Causing Invasive Pneumococcal Disease in Korea Between 2017 and 2019 After Introduction of the 13-Valent Pneumococcal Conjugate Vaccine
Gyu Ri KIM ; Eun-Young KIM ; Si Hyun KIM ; Hae Kyung LEE ; Jaehyeon LEE ; Jong Hee SHIN ; Young Ree KIM ; Sae Am SONG ; Joseph JEONG ; Young UH ; Yu Kyung KIM ; Dongeun YONG ; Hyun Soo KIM ; Sunjoo KIM ; Young Ah KIM ; Kyeong Seob SHIN ; Seok Hoon JEONG ; Namhee RYOO ; Jeong Hwan SHIN
Annals of Laboratory Medicine 2023;43(1):45-54
Background:
Streptococcus pneumoniae is a serious pathogen causing various infections in humans. We evaluated the serotype distribution and antimicrobial resistance of S. pneumoniae causing invasive pneumococcal disease (IPD) after introduction of pneumococcal conjugate vaccine (PCV)13 in Korea and investigated the epidemiological characteristics of multidrug-resistant (MDR) isolates.
Methods:
S. pneumoniae isolates causing IPD were collected from 16 hospitals in Korea between 2017 and 2019. Serotyping was performed using modified sequential multiplex PCR and the Quellung reaction. Antimicrobial susceptibility tests were performed using the broth microdilution method. Multilocus sequence typing was performed on MDR isolates for epidemiological investigations.
Results:
Among the 411 S. pneumoniae isolates analyzed, the most prevalent serotype was 3 (12.2%), followed by 10A (9.5%), 34 (7.3%), 19A (6.8%), 23A (6.3%), 22F (6.1%), 35B (5.8%), 11A (5.1%), and others (40.9%). The coverage rates of PCV7, PCV10, PCV13, and pneumococcal polysaccharide vaccine (PPSV)23 were 7.8%, 7.8%, 28.7%, and 59.4%, respectively. Resistance rates to penicillin, ceftriaxone, erythromycin, and levofloxacin were 13.1%, 9.2%, 80.3%, and 4.1%, respectively. MDR isolates accounted for 23.4% of all isolates. Serotypes 23A, 11A, 19A, and 15B accounted for the highest proportions of total isolates at 18.8%, 16.7%, 14.6%, and 8.3%, respectively. Sequence type (ST)166 (43.8%) and ST320 (12.5%) were common among MDR isolates.
Conclusions
Non-PCV13 serotypes are increasing among invasive S. pneumoniae strains causing IPD. Differences in antimicrobial resistance were found according to the specific serotype. Continuous monitoring of serotypes and antimicrobial resistance is necessary for the appropriate management of S. pneumoniae infections.
6.Serotype Distribution and Antimicrobial Resistance of Invasive and Noninvasive Streptococcus pneumoniae Isolates in Korea between 2014 and 2016
Dong Chul PARK ; Si Hyun KIM ; Dongeun YONG ; In Bum SUH ; Young Ree KIM ; Jongyoun YI ; Wonkeun SONG ; Sae Am SONG ; Hee Won MOON ; Hae Kyung LEE ; Kyoung Un PARK ; Sunjoo KIM ; Seok Hoon JEONG ; Jaehyeon LEE ; Joseph JEONG ; Yu Kyung KIM ; Miae LEE ; Jihyun CHO ; Jong Wan KIM ; Kyeong Seob SHIN ; Sang Hyun HWANG ; Jae Woo CHUNG ; Hye In WOO ; Chae Hoon LEE ; Namhee RYOO ; Chulhun L CHANG ; Hyun Soo KIM ; Jayoung KIM ; Jong Hee SHIN ; Soo Hyun KIM ; Mi Kyung LEE ; Seong Gyu LEE ; Sook Jin JANG ; Kyutaeg LEE ; HunSuk SUH ; Yong Hak SOHN ; Min Jung KWON ; Hee Joo LEE ; Ki Ho HONG ; Kwang Sook WOO ; Chul Min PARK ; Jeong Hwan SHIN
Annals of Laboratory Medicine 2019;39(6):537-544
BACKGROUND: Several factors contribute to differences in Streptococcus pneumoniae serotype distribution. We investigated the serotype distribution and antimicrobial resistance of S. pneumoniae isolated between 2014 and 2016 in Korea. METHODS: We collected a total of 1,855 S. pneumoniae isolates from 44 hospitals between May 2014 and May 2016, and analyzed the serotypes by sequential multiplex PCR. We investigated the distribution of each serotype by patient age, source of the clinical specimen, and antimicrobial resistance pattern. RESULTS: The most common serotypes were 11A (10.1%), followed by 19A (8.8%), 3 (8.5%), 34 (8.1%), 23A (7.3%), and 35B (6.2%). The major invasive serotypes were 3 (12.6%), 19A (7.8%), 34 (7.8%), 10A (6.8%), and 11A (6.8%). Serotypes 10A, 15B, 19A, and 12F were more common in patients ≤5 years old, while serotype 3 was more common in patients ≥65 years old compared with the other age groups. The coverage rates of pneumococcal conjugate vaccine (PCV)7, PCV10, PCV13, and pneumococcal polysaccharide vaccine 23 were 11.8%, 12.12%, 33.3%, and 53.6%, respectively. Of the 1,855 isolates, 857 (46.2%) were multi-drug resistant (MDR), with serotypes 11A and 19A predominant among the MDR strains. The resistance rates against penicillin, cefotaxime, and levofloxacin were 22.8%, 12.5%, and 9.4%, respectively. CONCLUSIONS: There were significant changes in the major S. pneumoniae serotypes in the community. Non-PCV13 serotypes increased in patients ≤5 years old following the introduction of national immunization programs with the 10- and 13-polyvalent vaccines.
Cefotaxime
;
Humans
;
Immunization Programs
;
Korea
;
Levofloxacin
;
Multiplex Polymerase Chain Reaction
;
Penicillins
;
Pneumococcal Vaccines
;
Pneumonia
;
Serogroup
;
Streptococcus pneumoniae
;
Streptococcus
;
Vaccines